Tpl2 ablation promotes intestinal inflammation and tumorigenesis in Apcmin mice by inhibiting IL-10 secretion and regulatory T-cell generation

OB Serebrennikova, C Tsatsanis… - Proceedings of the …, 2012 - National Acad Sciences
OB Serebrennikova, C Tsatsanis, C Mao, E Gounaris, W Ren, LD Siracusa, AG Eliopoulos
Proceedings of the National Academy of Sciences, 2012National Acad Sciences
To address the role of Tpl2, a MAP3K8 that regulates innate/adaptive immunity and
inflammation, in intestinal tumorigenesis, we crossed a Tpl2 KO allele into the Apcmin/+
genetic background. Here, we show that Apcmin/+/Tpl2−/− mice exhibit a fivefold increase in
the number of intestinal adenomas. Bone marrow transplantation experiments revealed that
the enhancement of polyposis was partially hematopoietic cell-driven. Consistent with this
observation, Tpl2 ablation promoted intestinal inflammation. IL-10 levels and regulatory T …
To address the role of Tpl2, a MAP3K8 that regulates innate/adaptive immunity and inflammation, in intestinal tumorigenesis, we crossed a Tpl2 KO allele into the Apcmin/+ genetic background. Here, we show that Apcmin/+/Tpl2−/− mice exhibit a fivefold increase in the number of intestinal adenomas. Bone marrow transplantation experiments revealed that the enhancement of polyposis was partially hematopoietic cell-driven. Consistent with this observation, Tpl2 ablation promoted intestinal inflammation. IL-10 levels and regulatory T-cell numbers were lower in the intestines of Tpl2−/− mice, independent of Apc and polyp status, suggesting that they were responsible for the initiation of the enhancement of tumorigenesis caused by the ablation of Tpl2. The low IL-10 levels correlated with defects in mTOR activation and Stat3 phosphorylation in Toll-like receptor-stimulated macrophages and with a defect in inducible regulatory T-cell generation and function. Both polyp numbers and inflammation increased progressively with time. The rate of increase of both, however, was more rapid in Apcmin/+/Tpl2−/− mice, suggesting that the positive feedback initiated by inflammatory signals originating in developing polyps is more robust in these mice. This may be because these mice have a higher intestinal polyp burden as a result of the enhancement of tumor initiation.
National Acad Sciences