Trends in Cell Biology
Volume 20, Issue 11, November 2010, Pages 662-671
Journal home page for Trends in Cell Biology

Review
Histone demethylases in development and disease

https://doi.org/10.1016/j.tcb.2010.08.011Get rights and content

Histone modifications serve as regulatory marks that are instrumental for the control of transcription and chromatin architecture. Strict regulation of gene expression patterns is crucial during development and differentiation, where diverse cell types evolve from common predecessors. Since the first histone lysine demethylase was discovered in 2004, a number of demethylases have been identified and implicated in the control of gene expression programmes and cell fate decisions. Histone demethylases are now emerging as important players in developmental processes and have been linked to human diseases such as neurological disorders and cancer.

Introduction

Eukaryotic DNA is packed into chromatin whose basic repeating unit is the nucleosome. The nucleosome contains 145-147 base pairs of DNA wrapped nearly twice around an octamer of the core histones, H2A, H2B, H3 and H4. In addition, the linker histone, H1, helps further compact the nucleosomal DNA into higher-order structures. Histones, especially their flexible tails extending from the nucleosomal core, are subjected to numerous post-translational modifications, which include methylation, acetylation, phosphorylation, ubiquitylation and SUMOylation. Histone methylation occurs on lysines and arginines. Adding complexity, residues can exist in different methylated forms with lysines (K), being either mono- (me1), di- (me2) or tri-methylated (me3), and arginines (R) being monomethylated or symmetrically or asymmetrically dimethylated. In general, trimethylation of H3K4, H3K36 and H3K79 are found in euchromatic regions with transcriptional activity, whereas H3K9me3/me2, H4K20me3 and H3K27me3 are associated with transcriptionally silenced chromatin. Histone modifications are believed to be important for coordinating both transient changes in gene transcription, as well as for maintaining differential patterns of gene expression during organismal development. Chromatin architecture also impinges on genomic stability, being linked to processes such as DNA repair and chromosome segregation.

Histone modifications have been suggested to be part of a ‘code’ that is read by proteins via specific binding domains, and in this way translated into a functional signal [1]. Thus histone modifications can influence chromatin condensation, and poise genes for either transcriptional activation or repression, depending on how the modification is read and translated in a specific context. While individual histone marks have been correlated with either active or silenced transcriptional states, many modifications have several, seemingly opposing roles, and the combination of marks, as well as their genomic context, appear to be essential for the biological output (reviewed in 2, 3, 4).

Our current knowledge regarding histone methylation stems in large part from the study of histone methyltransferases. Several of these enzymes are essential for development, and deregulated expression has been linked to human disorders such as cancer (reviewed in [5]). The more recent discovery of histone demethylases (Box 1) made a significant impact on the perception of histone methylation as permanent, inheritable marks supporting more dynamic roles in gene regulation.

Two classes of histone demethylases have thus far been identified. The proteins of the KDM1 (Lysine (K) Demethylase 1) family are FAD-dependent amine oxidases, which can act only on mono- and dimethylated lysines (Figure 1, Figure 2, Table 1). The Jumonji C (JmjC) domain is a signature motif for the other class of demethylases, which are Fe(II) and 2-oxoglutarate-dependent enzymes. Based on sequence homology in the JmjC domain and the overall architecture of associated motifs, JmjC domain-containing proteins have been classified into different groups, several of which have been found to possess histone demethylase activity (Figure 1, Figure 2, Table 1).

Here we review the current knowledge regarding histone demethylases with a particular focus on their potential roles in development and disease.

Section snippets

The KDM1 family

The first protein demonstrated to possess histone demethylase activity was mammalian KDM1A/AOF2 (amine oxidase (flavin containing) domain 2)/LSD1 (lysine-specific demethylase 1). In an elegant study, Shi and co-workers found that KDM1A could demethylate H3K4me2/me1 [7]. Subsequent studies have shown histone demethylase activity for the closely related KDM1B/AOF1/LSD2, as well as for orthologues in other species 8, 9, 10, 11.

Highlighting the importance of KDM1A for normal development, targeted

The KDM2 cluster

The first JmjC domain-containing protein shown to be a histone demethylase was KDM2A/JHDM1A (JmjC domain-containing histone demethylation protein 1A)/FBXL11 (F-box and leucine-rich repeat protein 11) [29]. Mammalian KDM2A and KDM2B/JHDM1B/FBXL10, as well as the homologues in Drosophila and S. cerevisiae, have been demonstrated to catalyze H3K36me2/me1 demethylation 29, 30, 31. In addition, mammalian KDM2B has been suggested to act as an H3K4me3 demethylase 32, 33, but discrepancy exists about

The KDM3 cluster

KDM3A/JHDM2A/JMJD1A (jumonji domain-containing 1a)/TSGA (testis-specific gene A) is a histone demethylase specific for H3K9me2/me1 [43]. Two KDM3A homologues exist in mammalian cells - KDM3B/JHDM2B/JMJD1B and JMJD1C/JHDM2C/TRIP8 (thyroid receptor interacting protein 8) - but demethylase activities of these proteins have not been reported so far.

Studies of genetrap and knockout mice have demonstrated important roles for KDM3A in germ cell development and metabolism 44, 45, 46, 47. KDM3A is

The KDM4 cluster

The KDM4 proteins were the first published demethylases that showed activity towards trimethylated lysines. There are four members of the KDM4 cluster in mammalian cells - KDM4A/JHDM3A/JMJD2A, KDM4B/JHDM3B/JMJD2B, KDM4C/JHDM3C/JMJD2C/GASC1 (gene amplified in squamous cell carcinoma 1) and KDM4D/JHDM3D/JMJD2D. The KDM4 proteins catalyze the demethylation of H3K9me3/me2 and/or H3K36me3/me2, with the substrate specificity varying between family members 50, 51, 52, 53, 54. Moreover, the KDM4

The KDM5 cluster

The KDM5 proteins catalyze the demethylation of H3K4me3/me2 62, 63, 64, 65, 66, 67. In mammalian cells, the KDM5 family is constituted by KDM5A/JARID1A (Jumonji, AT-rich interactive domain 1A)/RBP2 (retinoblastoma-binding protein 2), KDM5B/JARID1B/PLU-1, KDM5C/JARID1C/SMCX (selected mouse cDNA on the X) and KDM5D/JARID1D/SMCY.

Important developmental functions have been demonstrated for the KDM5 proteins in C. elegans and Drosophila. In C. elegans, deletion of the JmjC domain of RBR-2

The KDM6 cluster

The KDM6 cluster in mammalian cells consists of KDM6A/UTX (ubiquitously transcribed X chromosome tetraticopeptide repeat protein), UTY and KDM6B/JMJD3. Whereas KDM6A and KDM6B are histone demethylases specific for H3K27me3/me2, no activity has so far been reported for UTY 85, 86, 87, 88.

The importance of H3K27 demethylases in normal development is underscored by studies in C. elegans and D. rerio. In C. elegans, mutation of one of the three KDM6B homologues disrupts gonadal development [85],

The PHF cluster

Recently, histone demethylase activity has been reported for the PHF (plant homeodomain finger protein) cluster of the JmjC family of proteins. Mammalian PHF8 possesses H3K9me2/me1 activity 99, 100, 101, 102, whereas JHDM1D/KIAA1718 targets both H3K9me2/me1 and H3K27me2/me1 99, 103, 104. PHF2 has been suggested to demethylate H3K9me1, but this has yet to be confirmed by in vitro studies [105].

Intriguingly, mutations in the human PHF8 gene might, like KDM5C mutations, be relevant for the

JMJD6

JMJD6/PSR/PTDSR (phosphatidylserine receptor) is the first demethylase described to target arginine residues and was reported to demethylate H3R2me2 and H4R3me2 [109]. However this activity has been questioned recently, and it has been demonstrated that JMJD6 has strong lysyl hydroxylation activity towards the splicing factor U2AF2 (U2 small nuclear ribonucleoprotein auxiliary factor) [110]. Originally, JMJD6 was identified as the phosphatidylserine receptor responsible for recognizing

Perspectives and concluding remarks

Since the discovery of the first bona fide histone demethylase, several proteins have been demonstrated to possess demethylase activity, and more demethylases will most likely be identified in the coming years. With the identification of histone demethylases, it became clear that histone methylation patterns are the result of equilibria between opposing activities. Tremendous progress has been made towards unravelling the cellular functions of histone demethylases, but many important questions

Acknowledgements

We thank Jesper Christensen and Paul Cloos for help with illustrations and members of the Helin lab for critical comments on the manuscript. MTP was supported by fellowships from P. Carl Petersen's Foundation and the Danish Cancer Society. The work in the Helin lab is supported by grants from the Danish National Research Foundation, the Danish Cancer Society, The Lundbeck Foundation, the Novo Nordisk Foundation, the Danish Medical Research Council, and the Excellence Program of the University

References (119)

  • F. Forneris

    LSD1: oxidative chemistry for multifaceted functions in chromatin regulation

    Trends Biochem. Sci.

    (2008)
  • L. Di Stefano et al.

    Mutation of Drosophila Lsd1 disrupts H3-K4 methylation, resulting in tissue-specific defects during development

    Curr. Biol.

    (2007)
  • D.J. Katz

    A C. elegans LSD1 demethylase contributes to germline immortality by reprogramming epigenetic memory

    Cell

    (2009)
  • N.P. Blackledge

    CpG islands recruit a histone H3 lysine 36 demethylase

    Mol. Cell

    (2010)
  • K. Yamane

    JHDM2A, a JmjC-containing H3K9 demethylase, facilitates transcription activation by androgen receptor

    Cell

    (2006)
  • Z. Liu

    Jmjd1a demethylase-regulated histone modification is essential for cAMP-response element modulator-regulated gene expression and spermatogenesis

    J. Biol. Chem.

    (2010)
  • C.H. Lin

    Heterochromatin protein 1a stimulates histone H3 lysine 36 demethylation by the Drosophila KDM4A demethylase

    Mol. Cell

    (2008)
  • J.R. Whetstine

    Reversal of histone lysine trimethylation by the JMJD2 family of histone demethylases

    Cell

    (2006)
  • P. Trojer

    Dynamic histone H1 isotype 4 methylation and demethylation by histone lysine methyltransferase G9a/KMT1C and the jumonji domain-containing JMJD2/KDM4 proteins

    J. Biol. Chem.

    (2009)
  • M.T. Lorbeck

    The histone demethylase Dmel\Kdm4A controls genes required for life span and male-specific sex determination in Drosophila

    Gene

    (2010)
  • J. Christensen

    RBP2 belongs to a family of demethylases, specific for tri-and dimethylated lysine 4 on histone 3

    Cell

    (2007)
  • S. Iwase

    The X-linked mental retardation gene SMCX/JARID1C defines a family of histone H3 lysine 4 demethylases

    Cell

    (2007)
  • R.J. Klose

    The retinoblastoma binding protein RBP2 is an H3K4 demethylase

    Cell

    (2007)
  • M.G. Lee

    Physical and functional association of a trimethyl H3K4 demethylase and Ring6a/MBLR, a polycomb-like protein

    Cell

    (2007)
  • K. Yamane

    PLU-1 is an H3K4 demethylase involved in transcriptional repression and breast cancer cell proliferation

    Mol. Cell

    (2007)
  • Y.M. Moshkin

    Histone chaperones ASF1 and NAP1 differentially modulate removal of active histone marks by LID-RPD3 complexes during NOTCH silencing

    Mol. Cell

    (2009)
  • E.V. Benevolenskaya

    Binding of pRB to the PHD protein RBP2 promotes cellular differentiation

    Mol. Cell

    (2005)
  • N. Lopez-Bigas

    Genome-wide analysis of the H3K4 histone demethylase RBP2 reveals a transcriptional program controlling differentiation

    Mol. Cell

    (2008)
  • C. van Oevelen

    A role for mammalian Sin3 in permanent gene silencing

    Mol. Cell

    (2008)
  • J. Zeng

    The histone demethylase RBP2 Is overexpressed in gastric cancer and its inhibition triggers senescence of cancer cells

    Gastroenterology

    (2010)
  • P.J. Lu

    A novel gene (PLU-1) containing highly conserved putative DNA/chromatin binding motifs is specifically up-regulated in breast cancer

    J. Biol. Chem.

    (1999)
  • L.R. Jensen

    Mutations in the JARID1C gene, which is involved in transcriptional regulation and chromatin remodeling, cause X-linked mental retardation

    Am. J. Hum. Genet.

    (2005)
  • F. De Santa

    The histone H3 lysine-27 demethylase Jmjd3 links inflammation to inhibition of polycomb-mediated gene silencing

    Cell

    (2007)
  • Y.W. Cho

    PTIP associates with MLL3- and MLL4-containing histone H3 lysine 4 methyltransferase complex

    J. Biol. Chem.

    (2007)
  • D. Kleine-Kohlbrecher

    A functional link between the histone demethylase PHF8 and the transcription factor ZNF711 in X-linked mental retardation

    Mol. Cell

    (2010)
  • T. Jenuwein et al.

    Translating the histone code

    Science

    (2001)
  • E.I. Campos et al.

    Histones: annotating chromatin

    Annu. Rev. Genet.

    (2009)
  • S.D. Taverna

    How chromatin-binding modules interpret histone modifications: lessons from professional pocket pickers

    Nat. Struct. Mol. Biol.

    (2007)
  • M. Opel

    Genome-wide studies of histone demethylation catalysed by the fission yeast homologues of mammalian LSD1

    PLoS One

    (2007)
  • J. Wang

    The lysine demethylase LSD1 (KDM1) is required for maintenance of global DNA methylation

    Nat. Genet.

    (2009)
  • J. Wang

    Opposing LSD1 complexes function in developmental gene activation and repression programmes

    Nature

    (2007)
  • G. Sun

    The histone demethylase LSD1 regulates neural stem cell proliferation

    Mol. Cell Biol.

    (2010)
  • X. Hu

    LSD1-mediated epigenetic modification is required for TAL1 function and hematopoiesis

    Proc. Natl. Acad. Sci. U. S. A.

    (2009)
  • S.T. Su

    Involvement of histone demethylase LSD1 in Blimp-1-mediated gene repression during plasma cell differentiation

    Mol. Cell Biol.

    (2009)
  • P. Kahl

    Androgen receptor coactivators lysine-specific histone demethylase 1 and four and a half LIM domain protein 2 predict risk of prostate cancer recurrence

    Cancer Res.

    (2006)
  • S. Lim

    Lysine-specific demethylase 1 (LSD1) is highly expressed in ER-negative breast cancers and a biomarker predicting aggressive biology

    Carcinogenesis

    (2010)
  • E. Metzger

    LSD1 demethylates repressive histone marks to promote androgen-receptor-dependent transcription

    Nature

    (2005)
  • J.H. Schulte

    Lysine-specific demethylase 1 is strongly expressed in poorly differentiated neuroblastoma: implications for therapy

    Cancer Res.

    (2009)
  • J. Huang

    p53 is regulated by the lysine demethylase LSD1

    Nature

    (2007)
  • D.N. Ciccone

    KDM1B is a histone H3K4 demethylase required to establish maternal genomic imprints

    Nature

    (2009)
  • Cited by (317)

    View all citing articles on Scopus
    View full text