RT Journal Article SR Electronic T1 Drosophila Kruppel homolog 1 represses lipolysis through interaction with dFOXO JF bioRxiv FD Cold Spring Harbor Laboratory SP 165456 DO 10.1101/165456 A1 Ping Kang A1 Kai Chang A1 Ying Liu A1 Mark Bouska A1 Galina Karashchuk A1 Rachel Thakore A1 Wenjing Zheng A1 Stephanie Post A1 Colin S. Brent A1 Sheng Li A1 Marc Tatar A1 Hua Bai YR 2017 UL http://biorxiv.org/content/early/2017/07/18/165456.abstract AB Transcriptional coordination is a vital process contributing to metabolic homeostasis. As one of the key nodes in the metabolic network, the forkhead transcription factor FOXO has been shown to interact with diverse transcription co-factors and integrate signals from multiple pathways to control metabolism, oxidative stress response, and cell cycle. Recently, insulin/FOXO signaling has been implicated in the regulation of insect development via the interaction with insect hormones, such as ecdysone and juvenile hormone. In this study, we identified an interaction between dFOXO and the zinc finger transcription factor Kruppel homolog 1 (Kr-h1), one of the key players in juvenile hormone signaling in Drosophila. We found that Kr-h1 mutants have reduced triglyceride storage, decreased insulin signaling and delayed larval development. Notably, Kr-h1 physically and genetically interacts with dFOXO in vitro and in vivo to regulate the transcriptional activation of adipose lipase brummer (bmm). The transcriptional co-regulation by Kr-h1 and dFOXO may represent a broad mechanism by which Kruppel-like factors integrate with insulin signaling to maintain metabolic homeostasis and coordinate organism growth.