Specific Aim 1: examine the change of histone modifications on PPARgamma promoter in the early phase of adipogenesis. By ChIP assays, we have observed robust change of multiple histone acetylation and methylation on PPARgamma promoter in the early phase of adipogenesis.
Specific Aim 2 : investigate the functional significance of the histone modification change on PPARgamma promoter by depleting the responsible enzymes.We show recently that histone H3K27 methyltransferase Ezh2 represses Wnt genes to facilitate adipogenesis. Specifically, we show Ezh2 and its H3K27 methyltransferase activity are required for adipogenesis. Ezh2 directly represses Wnt1, -6, -10a, and -10b genes in preadipocytes and during adipogenesis. Deletion of Ezh2 eliminates H3K27me3 on Wnt promoters and derepresses Wnt expression, which leads to activation of Wnt/beta-catenin signaling and inhibition of adipogenesis. Ectopic expression of the wild-type (WT) Ezh2, but not the enzymatically inactive F667I mutant, prevents the loss of H3K27me3 and the defects in adipogenesis in Ezh2(-/-) preadipocytes. The adipogenesis defects in Ezh2(-/-) cells can be rescued by expression of adipogenic transcription factors PPARgamma, C/EBPalpha, or inhibitors of Wnt/beta-catenin signaling. Interestingly, Ezh2(-/-) cells show marked increase of H3K27 acetylation globally as well as on Wnt promoters. These results indicate that H3K27 methyltransferase Ezh2 directly represses Wnt genes to facilitate adipogenesis and suggest that acetylation and trimethylation on H3K27 play opposing roles in regulating Wnt expression (Wang L. et al., Proc Natl Acad Sci U S A (107): 7317-22, 2010).
Specific Aim 3 : investigate the roles of other epigenetic mechanisms, in particular chromatin remodeling and non-coding RNAs, in regulation of PPARgamma expression and adipogenesis.

Project Start
Project End
Budget Start
Budget End
Support Year
3
Fiscal Year
2011
Total Cost
$587,230
Indirect Cost
City
State
Country
Zip Code
Zhuang, Lenan; Jang, Younghoon; Park, Young-Kwon et al. (2018) Depletion of Nsd2-mediated histone H3K36 methylation impairs adipose tissue development and function. Nat Commun 9:1796
Katz, Liora S; Xu, Shiliyang; Ge, Kai et al. (2018) T3 and Glucose Coordinately Stimulate ChREBP-Mediated Ucp1 Expression in Brown Adipocytes From Male Mice. Endocrinology 159:557-569
Park, Young-Kwon; Wang, Limin; Giampietro, Anne et al. (2017) Distinct Roles of Transcription Factors KLF4, Krox20, and Peroxisome Proliferator-Activated Receptor ? in Adipogenesis. Mol Cell Biol 37:
Park, Young-Kwon; Ge, Kai (2017) Glucocorticoid Receptor Accelerates, but Is Dispensable for, Adipogenesis. Mol Cell Biol 37:
Lai, Binbin; Lee, Ji-Eun; Jang, Younghoon et al. (2017) MLL3/MLL4 are required for CBP/p300 binding on enhancers and super-enhancer formation in brown adipogenesis. Nucleic Acids Res 45:6388-6403
Froimchuk, Eugene; Jang, Younghoon; Ge, Kai (2017) Histone H3 lysine 4 methyltransferase KMT2D. Gene 627:337-342
Lee, Ji-Eun; Park, Young-Kwon; Park, Sarah et al. (2017) Brd4 binds to active enhancers to control cell identity gene induction in adipogenesis and myogenesis. Nat Commun 8:2217
Ang, Siang-Yun; Uebersohn, Alec; Spencer, C Ian et al. (2016) KMT2D regulates specific programs in heart development via histone H3 lysine 4 di-methylation. Development 143:810-21
Ray Chaudhuri, Arnab; Callen, Elsa; Ding, Xia et al. (2016) Replication fork stability confers chemoresistance in BRCA-deficient cells. Nature 535:382-7
Zhang, Jiyuan; Dominguez-Sola, David; Hussein, Shafinaz et al. (2015) Disruption of KMT2D perturbs germinal center B cell development and promotes lymphomagenesis. Nat Med 21:1190-8

Showing the most recent 10 out of 26 publications