952
Views
8
CrossRef citations to date
0
Altmetric
Research Paper

A histone H3.3K36M mutation in mice causes an imbalance of histone modifications and defects in chondrocyte differentiation

, , & ORCID Icon
Pages 1123-1134 | Received 17 Jul 2020, Accepted 07 Oct 2020, Published online: 16 Nov 2020

References

  • Ono N, Ono W, Nagasawa T, et al. A subset of chondrogenic cells provides early mesenchymal progenitors in growing bones. Nat Cell Biol. 2014;16(12):1157–1167.
  • Yang L, Tsang KY, Tang HC, et al. Hypertrophic chondrocytes can become osteoblasts and osteocytes in endochondral bone formation. Proc Natl Acad Sci U S A. 2014;111:12097–12102.
  • Zhou X, von der Mark K, Henry S, et al. Chondrocytes transdifferentiate into osteoblasts in endochondral bone during development, postnatal growth and fracture healing in mice. PLoS Genet. 2014;10:e1004820.
  • Kozhemyakina E, Lassar AB, Zelzer E. A pathway to bone: signaling molecules and transcription factors involved in chondrocyte development and maturation. Development. 2015;142:817–831.
  • Schwartzentruber J, Korshunov A, Liu XY, et al. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature. 2012;482:226–231.
  • Maze I, Noh KM, Soshnev AA, et al. Every amino acid matters: essential contributions of histone variants to mammalian development and disease. Nat Rev Genet. 2014;15:259–271.
  • Nacev BA, Feng L, Bagert JD, et al. The expanding landscape of ‘oncohistone’ mutations in human cancers. Nature. 2019;567:473–478.
  • Behjati S, Tarpey PS, Presneau N, et al. Distinct H3F3A and H3F3B driver mutations define chondroblastoma and giant cell tumor of bone. Nat Genet. 2013;45:1479–1482.
  • Fang D, Gan H, Lee JH, et al. The histone H3.3K36M mutation reprograms the epigenome of chondroblastomas. Science. 2016;352:1344–1348.
  • Lu C, Jain SU, Hoelper D, et al. Histone H3K36 mutations promote sarcomagenesis through altered histone methylation landscape. Science. 2016;352:844–849.
  • Yang S, Zheng X, Lu C, et al. Molecular basis for oncohistone H3 recognition by SETD2 methyltransferase. Genes Dev. 2016;30:1611–1616.
  • Schmitges FW, Prusty AB, Faty M, et al. Histone methylation by PRC2 is inhibited by active chromatin marks. Mol Cell. 2011;42:330–341.
  • Streubel G, Watson A, Jammula SG, et al. The H3K36me2 methyltransferase Nsd1 demarcates PRC2-mediated H3K27me2 and H3K27me3 domains in embryonic stem cells. Mol Cell. 2018;70:371–379 e375.
  • Sakai K, Miyazaki J. A transgenic mouse line that retains Cre recombinase activity in mature oocytes irrespective of the cre transgene transmission. Biochem Biophys Res Commun. 1997;237:318–324.
  • Chen W, DiFrancesco LM. Chondroblastoma: an update. Arch Pathol Lab Med. 2017;141:867–871.
  • Logan M, Martin JF, Nagy A, et al. Expression of cre recombinase in the developing mouse limb bud driven by a Prxl enhancer. Genesis. 2002;33:77–80.
  • Jin H, Kasper LH, Larson JD, et al. ChIPseqSpikeInFree: a ChIP-seq normalization approach to reveal global changes in histone modifications without spike-in. Bioinformatics. 2020;36:1270–1272.
  • Bracken AP, Dietrich N, Pasini D, et al. Genome-wide mapping of Polycomb target genes unravels their roles in cell fate transitions. Genes Dev. 2006;20:1123–1136.
  • Zenk F, Loeser E, Schiavo R, et al. Germ line-inherited H3K27me3 restricts enhancer function during maternal-to-zygotic transition. Science. 2017;357:212–216.
  • Cruz-Molina S, Respuela P, Tebartz C, et al. PRC2 facilitates the regulatory topology required for poised enhancer function during pluripotent stem cell differentiation. Cell Stem Cell. 2017;20:689–705 e689.
  • Shen Y, Yue F, McCleary DF, et al. A map of the cis-regulatory sequences in the mouse genome. Nature. 2012;488:116–120.
  • Suomi S, Taipaleenmaki H, Seppanen A, et al. MicroRNAs regulate osteogenesis and chondrogenesis of mouse bone marrow stromal cells. Gene Regul Syst Bio. 2008;2:177–191.
  • Watanabe T, Sato T, Amano T, et al. Dnm3os, a non-coding RNA, is required for normal growth and skeletal development in mice. Dev Dyn. 2008;237:3738–3748.
  • Mirzamohammadi F, Papaioannou G, Kobayashi T. MicroRNAs in cartilage development, homeostasis, and disease. Curr Osteoporos Rep. 2014;12:410–419.
  • Roberto VP, Gavaia P, Nunes MJ, et al. Evidences for a new role of miR-214 in chondrogenesis. Sci Rep. 2018;8:3704.
  • Qin L, Beier F. EGFR signaling: friend or foe for cartilage? JBMR Plus. 2019;3:e10177.
  • Yamamoto S, Uchida Y, Ohtani T, et al. Hoxa13 regulates expression of common Hox target genes involved in cartilage development to coordinate the expansion of the autopodal anlage. Dev Growth Differ. 2019;61:228–251.
  • Wellik DM, Capecchi MR. Hox10 and Hox11 genes are required to globally pattern the mammalian skeleton. Science. 2003;301:363–367.
  • Mirzamohammadi F, Papaioannou G, Inloes JB, et al. Polycomb repressive complex 2 regulates skeletal growth by suppressing Wnt and TGF-beta signalling. Nat Commun. 2016;7:12047.
  • Massip L, Ectors F, Deprez P, et al. Expression of Hoxa2 in cells entering chondrogenesis impairs overall cartilage development. Differentiation. 2007;75:256–267.
  • Li Y, Trojer P, Xu C-F, et al. The target of the NSD family of histone lysine methyltransferases depends on the nature of the substrate. J Biol Chem. 2009;49:34283–34295.
  • Zhuang L, Jang Y, Park YK, et al. Depletion of Nsd2-mediated histone H3K36 methylation impairs adipose tissue development and function. Nat Commun. 2018;9:1796.
  • Cong L, Ran FA, Cox D, et al. Multiplex genome engineering using CRISPR/Cas systems. Science. 2013;339:819–823.
  • Brind’Amour J, Liu S, Hudson M, et al. An ultra-low-input native ChIP-seq protocol for genome-wide profiling of rare cell populations. Nat Commun. 2015;6:6033.
  • Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods. 2012;9:357–359.
  • Wang L, Wang S, Li W. RSeQC: quality control of RNA-seq experiments. Bioinformatics. 2012;28:2184–2185.
  • Li H, Handsaker B, Wysoker A, et al., Genome Project Data Processing, S. The sequence alignment/map format and SAMtools. Bioinformatic. 2009;25:2078–2079.
  • Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics. 2010;26:841–842.
  • Ramirez F, Ryan DP, Gruning B, et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 2016;44:W160–165.
  • Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low memory requirements. Nat Methods. 2015;12:357–360.
  • Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15:550.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.