TY - JOUR T1 - MTG16 (CBFA2T3) represses E protein-dependent transcription to regulate colonic secretory cell differentiation, epithelial regeneration, and tumorigenesis JF - bioRxiv DO - 10.1101/2021.11.03.467178 SP - 2021.11.03.467178 AU - Rachel E. Brown AU - Justin Jacobse AU - Shruti A. Anant AU - Koral M. Blunt AU - Bob Chen AU - Paige N. Vega AU - Chase T. Jones AU - Jennifer M. Pilat AU - Frank Revetta AU - Aidan H. Gorby AU - Kristy R. Stengel AU - Yash A. Choksi AU - Kimmo Palin AU - M. Blanca Piazuelo AU - M. Kay Washington AU - Ken S. Lau AU - Jeremy A. Goettel AU - Scott W. Hiebert AU - Sarah P. Short AU - Christopher S. Williams Y1 - 2021/01/01 UR - http://biorxiv.org/content/early/2021/11/04/2021.11.03.467178.abstract N2 - Aberrant epithelial differentiation and regeneration pathways contribute to colon pathologies including inflammatory bowel disease (IBD) and colitis-associated cancer (CAC). MTG16 (also known as CBFA2T3) is a transcriptional corepressor expressed in the colonic epithelium. MTG16 interaction partners include E box-binding basic helix-loop-helix transcription factors (E proteins). MTG16-deficient mice exhibit worse colitis and increased tumor burden in inflammatory carcinogenesis. In this study, we sought to understand the role of MTG16 colonic epithelial homeostasis and the mechanisms by which MTG16 protects the epithelium in colitis and CAC. We demonstrated that MTG16 deficiency enabled enteroendocrine cell differentiation from secretory precursor cells at the expense of goblet cells. Transcriptomic analysis implicated dysregulated E protein function in MTG16-deficient colon crypts. Using a novel mouse model with a point mutation that disrupts MTG16:E protein complex formation (Mtg16P209T), we established that enteroendocrine:goblet cell balance was dependent on MTG16:E protein interactions and that the shift in lineage allocation was associated with enhanced expression of Neurog3, the central driver of enteroendocrine lineage specification. Furthermore, Mtg16 was upregulated in the previously described Ascl2+, de-differentiating cells that replenish the stem cell compartment in response to colon injury. Mtg16 expression was also increased in dextran sulfate sodium (DSS)-treated mouse colon crypts and in IBD patients compared to unaffected controls. We determined that the effects of MTG16 in regeneration are also dependent on its repression of E proteins, as the colonic epithelium failed to regenerate following DSS-induced injury in our novel mutant mouse model. Finally, we revealed that uncoupling MTG16:E protein interactions contributes to the enhanced tumorigenicity in Mtg16-/- colon in the azoxymethane(AOM)/DSS-induced model of CAC. Collectively, our results demonstrate that MTG16, via its repression of E protein targets, is a key regulator of cell fate decisions during colonic differentiation and regeneration.Competing Interest StatementThe authors have declared no competing interest. ER -