Your browser doesn't support javascript.
loading
Interleukin-33 Promotes REG3γ Expression in Intestinal Epithelial Cells and Regulates Gut Microbiota.
Xiao, Yi; Huang, Xiangsheng; Zhao, Ye; Chen, Feidi; Sun, Mingming; Yang, Wenjing; Chen, Liang; Yao, Suxia; Peniche, Alex; Dann, Sara M; Sun, Jiaren; Golovko, George; Fofanov, Yuriy; Miao, Yinglei; Liu, Zhanju; Chen, Daiwen; Cong, Yingzi.
Afiliação
  • Xiao Y; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas; Key Laboratory of Animal Disease-Resistance Nutrition, Animal Nutrition Institute, Sichuan Agricultural University, Yaan, Sichuan, China.
  • Huang X; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas.
  • Zhao Y; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas.
  • Chen F; Department of Pathology, University of Texas Medical Branch, Galveston, Texas.
  • Sun M; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas; Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.
  • Yang W; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas; Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.
  • Chen L; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas; Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.
  • Yao S; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas.
  • Peniche A; Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas.
  • Dann SM; Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas.
  • Sun J; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas.
  • Golovko G; Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas.
  • Fofanov Y; Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas.
  • Miao Y; Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
  • Liu Z; Department of Gastroenterology, The Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.
  • Chen D; Key Laboratory of Animal Disease-Resistance Nutrition, Animal Nutrition Institute, Sichuan Agricultural University, Yaan, Sichuan, China. Electronic address: dwchen@sicau.edu.cn.
  • Cong Y; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas; Department of Pathology, University of Texas Medical Branch, Galveston, Texas. Electronic address: yicong@utmb.edu.
Article em En | MEDLINE | ID: mdl-30831322
ABSTRACT
BACKGROUND &

AIMS:

Regenerating islet-derived protein (REG3γ), an antimicrobial peptide, typically expressed by intestinal epithelial cells (IEC), plays crucial roles in intestinal homeostasis and controlling gut microbiota. However, the mechanisms that regulate IEC expression of REG3γ are still largely unclear. In this study, we investigated whether and how interleukin (IL) 33, an alarmin produced by IEC in response to injury, regulates REG3γ expression in IEC, thus contributing to intestinal homeostasis.

METHODS:

IEC were isolated from wild-type and IL33-/- mice to determine expression of REG3γ and other antimicrobial peptides by quantitative real-time polymerase chain reaction and Western blot. IEC cell lines were used for mechanistic studies. 16S rRNA pyrosequencing analysis was used for measuring gut microbiota. Citrobacter rodentium was used for enteric infections.

RESULTS:

The expression of REG3γ, but not ß-defensins, in IECs of IL33-/- mice was significantly lower than wild-type mice. IL33 treatment induced IEC expression of REG3γ in both mice and human cell lines. Mechanistically, IL33 activated STAT3, mTOR, and ERK1/2 in IEC. Inhibition of these pathways abrogated IL33-induction of REG3γ. IL33-/- mice demonstrated higher bacteria loads and altered microbiota composition. IL33 did not directly inhibit bacterial growth, but promoted wild-type, not REG3γKO, IECs to kill bacteria in vitro. Consistently, C rodentium infection induced IEC IL33 expression, and IL33-/- mice demonstrated an impaired bacterial clearance with C rodentium infection.

CONCLUSIONS:

Our study demonstrated that IL33, which is produced by IEC in response to injury and inflammatory stimulation, in turn promotes IEC expression of REG3γ, and controls the gut microbiota of the host.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Bactérias / RNA Ribossômico 16S / Interleucina-33 / Proteínas Associadas a Pancreatite / Mucosa Intestinal Limite: Animals / Humans Idioma: En Revista: Cell Mol Gastroenterol Hepatol Ano de publicação: 2019 Tipo de documento: Article País de afiliação: China

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Bactérias / RNA Ribossômico 16S / Interleucina-33 / Proteínas Associadas a Pancreatite / Mucosa Intestinal Limite: Animals / Humans Idioma: En Revista: Cell Mol Gastroenterol Hepatol Ano de publicação: 2019 Tipo de documento: Article País de afiliação: China