Journal of Inflammation (Aug 2020)

Anatabine ameliorates intestinal inflammation and reduces the production of pro-inflammatory factors in a dextran sulfate sodium mouse model of colitis

  • Pedro A. Ruiz Castro,
  • Ulrike Kogel,
  • Giuseppe Lo Sasso,
  • Blaine W. Phillips,
  • Alain Sewer,
  • Bjorn Titz,
  • Llenalia Garcia,
  • Athanasios Kondylis,
  • Emmanuel Guedj,
  • Dariusz Peric,
  • David Bornand,
  • Remi Dulize,
  • Celine Merg,
  • Maica Corciulo,
  • Nikolai V. Ivanov,
  • Manuel C. Peitsch,
  • Julia Hoeng

DOI
https://doi.org/10.1186/s12950-020-00260-6
Journal volume & issue
Vol. 17, no. 1
pp. 1 – 17

Abstract

Read online

Abstract Background Inflammatory bowel disease (IBD) is the collective term for chronic immune-mediated diseases of unknown, multifactorial etiology, arising from the interplay between genetic and environmental factors and including two main disease manifestations: ulcerative colitis (UC) and Crohn’s disease. In the last few decades, naturally occurring alkaloids have gained interest because of their substantial anti-inflammatory effects in several animal models of disease. Studies on mouse models of IBD have demonstrated the anti-inflammatory action of the main tobacco alkaloid, nicotine. In addition, anatabine, a minor tobacco alkaloid also present in peppers, tomato, and eggplant presents anti-inflammatory properties in vivo and in vitro. In this study, we aimed to evaluate the anti-inflammatory properties of nicotine and anatabine in a dextran sulfate sodium (DSS) mouse model of UC. Results Oral administration of anatabine, but not nicotine, reduced the clinical symptoms of DSS-induced colitis. The result of gene expression analysis suggested that anatabine had a restorative effect on global DSS-induced gene expression profiles, while nicotine only had limited effects. Accordingly, MAP findings revealed that anatabine reduced the colonic abundance of DSS-associated cytokines and increased IL-10 abundance. Conclusions Our results support the amelioration of inflammatory effects by anatabine in the DSS mouse model of UC, and suggest that anatabine constitutes a promising therapeutic agent for IBD treatment.

Keywords