Gastrointestinal tract dysbiosis enhances distal tumor progression through suppression of leukocyte trafficking

SV Jenkins, MS Robeson, RJ Griffin, CM Quick… - Cancer research, 2019 - AACR
SV Jenkins, MS Robeson, RJ Griffin, CM Quick, ER Siegel, MJ Cannon, KB Vang
Cancer research, 2019AACR
The overall use of antibiotics has increased significantly in recent years. Besides fighting
infections, antibiotics also alter the gut microbiota. Commensal bacteria in the
gastrointestinal tract are crucial to maintain immune homeostasis, and microbial imbalance
or dysbiosis affects disease susceptibility and progression. We hypothesized that antibiotic-
induced dysbiosis of the gut microbiota would suppress cytokine profiles in the host, thereby
leading to changes in the tumor microenvironment. The induced dysbiosis was …
Abstract
The overall use of antibiotics has increased significantly in recent years. Besides fighting infections, antibiotics also alter the gut microbiota. Commensal bacteria in the gastrointestinal tract are crucial to maintain immune homeostasis, and microbial imbalance or dysbiosis affects disease susceptibility and progression. We hypothesized that antibiotic-induced dysbiosis of the gut microbiota would suppress cytokine profiles in the host, thereby leading to changes in the tumor microenvironment. The induced dysbiosis was characterized by alterations in bacterial abundance, composition, and diversity in our animal models. On the host side, antibiotic-induced dysbiosis caused elongated small intestines and ceca, and B16-F10 melanoma and Lewis lung carcinoma progressed more quickly than in control mice. Mechanistic studies revealed that this progression was mediated by suppressed TNFα levels, both locally and systemically, resulting in reduced expression of tumor endothelial adhesion molecules, particularly intercellular adhesion molecule-1 (ICAM-1) and a subsequent decrease in the number of activated and effector CD8+ T cells in the tumor. However, suppression of ICAM-1 or its binding site, the alpha subunit of lymphocyte function-associated antigen-1, was not seen in the spleen or thymus during dysbiosis. TNFα supplementation in dysbiotic mice was able to increase ICAM-1 expression and leukocyte trafficking into the tumor. Overall, these results demonstrate the importance of commensal bacteria in supporting anticancer immune surveillance, define an important role of tumor endothelial cells within this process, and suggest adverse consequences of antibiotics on cancer control.
Significance
Antibiotic-induced dysbiosis enhances distal tumor progression by altering host cytokine levels, resulting in suppression of tumor endothelial adhesion molecules and activated and effector CD8+ T cells in the tumor.
AACR