Precision Mapping of Coexisting Modifications in Histone H3 Tails from Embryonic Stem Cells by ETD-MS/MS

2013 ◽  
Vol 85 (17) ◽  
pp. 8232-8239 ◽  
Author(s):  
Hye Ryung Jung ◽  
Simone Sidoli ◽  
Simon Haldbo ◽  
Richard R. Sprenger ◽  
Veit Schwämmle ◽  
...  
2015 ◽  
Vol 35 (22) ◽  
pp. 3909-3920 ◽  
Author(s):  
Tatsuya Ohhata ◽  
Mika Matsumoto ◽  
Martin Leeb ◽  
Shinwa Shibata ◽  
Satoshi Sakai ◽  
...  

One of the two X chromosomes in female mammals is inactivated by the noncodingXistRNA. In mice, X chromosome inactivation (XCI) is regulated by the antisense RNATsix, which repressesXiston the active X chromosome. In the absence ofTsix, PRC2-mediated histone H3 lysine 27 trimethylation (H3K27me3) is established over theXistpromoter. Simultaneous disruption ofTsixand PRC2 leads to derepression ofXistand in turn silencing of the single X chromosome in male embryonic stem cells. Here, we identified histone H3 lysine 36 trimethylation (H3K36me3) as a modification that is recruited byTsixcotranscriptionally and extends over theXistpromoter. Reduction of H3K36me3 by expression of a mutated histone H3.3 with a substitution of methionine for lysine at position 36 causes a significant derepression ofXist. Moreover, depletion of the H3K36 methylaseSetd2leads to upregulation ofXist, suggesting H3K36me3 as a modification that contributes to the mechanism ofTsixfunction in regulating XCI. Furthermore, we found that reduction of H3K36me3 does not facilitate an increase in H3K27me3 over theXistpromoter, indicating that additional mechanisms exist by whichTsixblocks PRC2 recruitment to theXistpromoter.


2021 ◽  
Author(s):  
Elana Bryan ◽  
Marie Warburton ◽  
Kimberly M. Webb ◽  
Katy A. McLaughlin ◽  
Christos Spanos ◽  
...  

SummaryPromoters of developmental genes in embryonic stem cells (ESCs) are marked by histone H3 lysine 4 trimethylation (H3K4me3) and H3K27me3 in an asymmetric nucleosomal conformation, with each sister histone H3 carrying only one mark. These bivalent domains are thought to poise genes for timely activation upon differentiation. Here we show that asymmetric bivalent nucleosomes recruit repressive H3K27me3 binders but fail to enrich activating H3K4me3 binders, despite presence of H3K4me3, thereby promoting a poised state. Strikingly, the bivalent mark combination further attracts chromatin proteins that are not recruited by each mark individually, including the histone acetyltransferase complex KAT6B (MORF). Knockout of KAT6B blocks neuronal differentiation, demonstrating that bivalency-specific readers are critical for proper ESC differentiation. These findings reveal how histone mark bivalency directly promotes establishment of a poised state at developmental genes, while highlighting how nucleosomal asymmetry is critical for histone mark readout and function.


2020 ◽  
Vol 3 (1) ◽  
Author(s):  
Azra Fatima ◽  
Dilber Irmak ◽  
Alireza Noormohammadi ◽  
Markus M. Rinschen ◽  
Aniruddha Das ◽  
...  

2009 ◽  
Vol 41 (2) ◽  
pp. 246-250 ◽  
Author(s):  
Bo Wen ◽  
Hao Wu ◽  
Yoichi Shinkai ◽  
Rafael A Irizarry ◽  
Andrew P Feinberg

2007 ◽  
Vol 21 (20) ◽  
pp. 2545-2557 ◽  
Author(s):  
Y.-H. Loh ◽  
W. Zhang ◽  
X. Chen ◽  
J. George ◽  
H.-H. Ng

2015 ◽  
Vol 35 (24) ◽  
pp. 4158-4169 ◽  
Author(s):  
Timothy S. Carey ◽  
Zubing Cao ◽  
Inchul Choi ◽  
Avishek Ganguly ◽  
Catherine A. Wilson ◽  
...  

During mouse preimplantation development, the generation of the inner cell mass (ICM) and trophoblast lineages comprises upregulation ofNanogexpression in the ICM and its silencing in the trophoblast. However, the underlying epigenetic mechanisms that differentially regulateNanogin the first cell lineages are poorly understood. Here, we report that BRG1 (Brahma-related gene 1) cooperates with histone deacetylase 1 (HDAC1) to regulateNanogexpression. BRG1 depletion in preimplantation embryos andCdx2-inducible embryonic stem cells (ESCs) revealed that BRG1 is necessary forNanogsilencing in the trophoblast lineage. Conversely, in undifferentiated ESCs, loss of BRG1 augmentedNanogexpression. Analysis of histone H3 within theNanogproximal enhancer revealed that H3 lysine 9/14 (H3K9/14) acetylation increased in BRG1-depleted embryos and ESCs. Biochemical studies demonstrated that HDAC1 was present in BRG1-BAF155 complexes and BRG1-HDAC1 interactions were enriched in the trophoblast lineage. HDAC1 inhibition triggered an increase in H3K9/14 acetylation and a corresponding rise inNanogmRNA and protein, phenocopying BRG1 knockdown embryos and ESCs. Lastly, nucleosome-mapping experiments revealed that BRG1 is indispensable for nucleosome remodeling at theNanogenhancer during trophoblast development. In summary, our data suggest that BRG1 governsNanogexpression via a dual mechanism involving histone deacetylation and nucleosome remodeling.


Sign in / Sign up

Export Citation Format

Share Document