User: Guest  Login
Document type:
Journal Article
Author(s):
Münch, Natasha Stephens; Fang, Hsin-Yu; Ingermann, Jonas; Maurer, H Carlo; Anand, Akanksha; Kellner, Victoria; Sahm, Vincenz; Wiethaler, Maria; Baumeister, Theresa; Wein, Frederik; Einwächter, Henrik; Bolze, Florian; Klingenspor, Martin; Haller, Dirk; Kavanagh, Maria; Lysaght, Joanne; Friedman, Richard; Dannenberg, Andrew J; Pollak, Michael; Holt, Peter R; Muthupalani, Sureshkumar; Fox, James G; Whary, Mark T; Lee, Yoomi; Ren, Tony Y; Elliot, Rachael; Fitzgerald, Rebecca; Steiger, Katja; Schmid,...     »
Title:
High-Fat Diet Accelerates Carcinogenesis in a Mouse Model of Barrett's Esophagus via Interleukin 8 and Alterations to the Gut Microbiome.
Abstract:
BACKGROUND & AIMS: Barrett's esophagus (BE) is a precursor to esophageal adenocarcinoma (EAC). Progression from BE to cancer is associated with obesity, possibly due to increased abdominal pressure and gastroesophageal reflux disease, although this pathogenic mechanism has not been proven. We investigated whether environmental or dietary factors associated with obesity contribute to the progression of BE to EAC in mice. METHODS: Tg(ED-L2-IL1RN/IL1B)#Tcw mice (a model of BE, called L2-IL1B mice) were fed a chow (control) or high-fat diet (HFD) or were crossbred with mice that express human interleukin (IL) 8 (L2-IL1B/IL8 mice). Esophageal tissues were collected and analyzed for gene expression profiles and by quantitative polymerase chain reaction, immunohistochemistry, and flow cytometry. Organoids were established from BE tissue of mice and cultured with serum from lean or obese individuals or with neutrophils from L2-IL1B mice. Feces from mice were analyzed by 16s ribosomal RNA sequencing and compared to 16s sequencing data from patients with dysplasia or BE. L2-IL1B were mice raised in germ-free conditions. RESULTS: L2-IL1B mice fed an HFD developed esophageal dysplasia and tumors more rapidly than mice fed the control diet; the speed of tumor development was independent of body weight. The acceleration of dysplasia by the HFD in the L2-IL1B mice was associated with a shift in the gut microbiota and an increased ratio of neutrophils to natural killer cells in esophageal tissues compared with mice fed a control diet. We observed similar differences in the microbiomes from patients with BE that progressed to EAC vs patients with BE that did not develop into cancer. Tissues from dysplasias of L2-IL1B mice fed the HFD contained increased levels of cytokines that are produced in response to CXCL1 (the functional mouse homolog of IL8, also called KC). Serum from obese patients caused organoids from L2-IL1B/IL8 mice to produce IL8. BE tissues from L2-IL1B mice fed the HFD and from L2-IL1B/IL8 mice contained increased numbers of myeloid cells and cells expressing Cxcr2 and Lgr5 messenger RNAs (epithelial progenitors) compared with mice fed control diets. BE tissues from L2-IL1B mice raised in germ-free housing had fewer progenitor cells and developed less dysplasia than in L2-IL1 mice raised under standard conditions; exposure of fecal microbiota from L2-IL1B mice fed the HFD to L2-IL1B mice fed the control diet accelerated tumor development. CONCLUSIONS: In a mouse model of BE, we found that an HFD promoted dysplasia by altering the esophageal microenvironment and gut microbiome, thereby inducing inflammation and stem cell expansion, independent of obesity.
Journal title abbreviation:
Gastroenterology
Year:
2019
Journal volume:
157
Journal issue:
2
Pages contribution:
492-506.e2
Fulltext / DOI:
doi:10.1053/j.gastro.2019.04.013
Pubmed ID:
http://view.ncbi.nlm.nih.gov/pubmed/30998992
Print-ISSN:
0016-5085
TUM Institution:
Frauenklinik und Poliklinik; II. Medizinische Klinik und Poliklinik (Gastroenterologie); Institut für Allgemeine Pathologie und Pathologische Anatomie
 BibTeX