TY - JOUR T1 - DOT1L primarily acts as a transcriptional repressor in hematopoietic progenitor cells JF - bioRxiv DO - 10.1101/2020.10.15.341255 SP - 2020.10.15.341255 AU - Shaon Borosha AU - Anamika Ratri AU - Sami M. Housami AU - Shubham Rai AU - Subhra Ghosh AU - Carrie A. Malcom AU - V. Praveen Chakravarthi AU - Jay L. Vivian AU - Timothy A. Fields AU - M.A. Karim Rumi AU - Patrick E. Fields Y1 - 2020/01/01 UR - http://biorxiv.org/content/early/2020/10/16/2020.10.15.341255.abstract N2 - DOT1L is essential for early hematopoiesis but the precise mechanisms remain largely unclear. The only known function of DOT1L is histone H3 lysine 79 (H3K79) methylation. We generated two mouse models; a Dot1L-knockout (Dot1L-KO), and another possessing a point mutation in its methyltransferase domain (Dot1L-MM) to determine the role of its catalytic activity during early hematopoiesis. We observed that Dot1L-KO embryos suffered from severe anemia, while Dot1L-MM embryos showed minimal to no anemia. However, ex vivo culture of Dot1L-MM hematopoietic progenitors (HPCs) exhibited defective development of myeloid and mixed progenitors. DOT1L is a well-recognized, cell-type specific epigenetic regulator of gene expression. To elucidate the mechanisms underlying such diverse hematopoietic properties of Dot1L-KO and Dot1L-MM HPCs, we examined their whole transcriptomes. Extensively self-renewing erythroblast (ESRE) cultures were established using yolk sac (YS) cells collected on embryonic day 10.5 (E10.5). Dot1l-KO and Dot1l-MM cells expanded significantly less than the wildtype cells and showed slower progression through the cell cycle. Total RNA extracted from the wildtype and Dot1l-mutant ESRE cells were subjected to RNA-seq analyses. We observed that the majority (~82%) of the differentially expressed genes (DEGs) were upregulated in both of the Dot1L-mutants, which suggests that DOT1L predominantly acts as a transcriptional repressor in HPCs. We also observed that about ~40% of the DEGs were unique to either of the mutant group, suggesting that DOT1L possesses both methyltransferase domain-dependent and -independent functions. We further analyzed Gene Ontology and signaling pathways relevant to the DEGs common to both mutant groups and those that were unique to either group. Among the common DEGs, we observed upregulation of CDK inhibitors, which explains the cell cycle arrest in both of the Dot1L-mutant progenitors.Competing Interest StatementThe authors have declared no competing interest. ER -