We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Disrupted intricacy of histone H3K4 methylation in neurodevelopmental disorders

    Christina N Vallianatos

    Department of Human Genetics, University of Michigan, 5815 Medical Science II, Ann Arbor, MI 48109, USA

    Predoctoral Training Program in Genetics, University of Michigan, 5815 Medical Science II, Ann Arbor, MI 48109, USA

    &
    Shigeki Iwase

    *Author for correspondence:

    E-mail Address: siwase@umich.edu

    Department of Human Genetics, University of Michigan, 5815 Medical Science II, Ann Arbor, MI 48109, USA

    Published Online:https://doi.org/10.2217/epi.15.1

    Methylation of histone H3 lysine 4 (H3K4me) is an intricately regulated posttranslational modification, which is broadly associated with enhancers and promoters of actively transcribed genomic loci. Recent advances in next-generation sequencing have identified a number of H3K4me regulators mutated in neurodevelopmental disorders including intellectual disabilities, autism spectrum disorders, and schizophrenia. Here, we aim to summarize the molecular function of H3K4me-regulating enzymes in brain development and function. We describe four H3K4me methyltransferases (KMT2A, KMT2C, KMT2D, KMT2F), four demethylases (KDM1A, KDM5A, KDM5B, KDM5C), and two reader proteins (PHF21A, PHF8) mutated in neurodevelopmental disorders. Understanding the role of these chromatin regulators in the development and maintenance of neural connections will advance therapeutic opportunities for prevention and treatment of these lifelong neurodevelopmental disorders.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1 Najmabadi H, Hu H, Garshasbi M et al. Deep sequencing reveals 50 novel genes for recessive cognitive disorders. Nature 478(7367), 57–63 (2011).• First large-scale exome sequencing study identifying numerous chromatin gene mutations in recessive intellectual disabilities.
    • 2 Takata A, Xu B, Ionita-Laza I, Roos JL, Gogos JA, Karayiorgou M. Loss-of-function variants in schizophrenia risk and SETD1A as a candidate susceptibility gene. Neuron 82(4), 773–780 (2014).
    • 3 Iossifov I, O'roak BJ, Sanders SJ et al. The contribution of de novo coding mutations to autism spectrum disorder. Nature 515(7526), 216–221 (2014).
    • 4 De Rubeis S, He X, Goldberg AP et al. Synaptic, transcriptional and chromatin genes disrupted in autism. Nature 515(7526), 209–215 (2014).• Recent large-scale exome sequencing study highlighting involvement of histone methylation in pathogenesis of ASD.
    • 5 Heintzman ND, Hon GC, Hawkins RD et al. Histone modifications at human enhancers reflect global cell-type-specific gene expression. Nature 459(7243), 108–112 (2009).
    • 6 Heintzman ND, Stuart RK, Hon G et al. Distinct and predictive chromatin signatures of transcriptional promoters and enhancers in the human genome. Nat. Genet. 39(3), 311–318 (2007).
    • 7 Barski A, Cuddapah S, Cui K et al. High-resolution profiling of histone methylations in the human genome. Cell 129(4), 823–837 (2007).
    • 8 Zhu J, Adli M, Zou JY et al. Genome-wide chromatin state transitions associated with developmental and environmental cues. Cell 152(3), 642–654 (2013).
    • 9 Cheung I, Shulha HP, Jiang Y et al. Developmental regulation and individual differences of neuronal H3K4me3 epigenomes in the prefrontal cortex. Proc. Natl Acad. Sci. USA 107(19), 8824–8829 (2010).
    • 10 Lan F, Collins RE et al. Recognition of unmethylated histone H3 lysine 4 links BHC80 to LSD1-mediated gene repression. Nature 448(7154), 718–722 (2007).• First characterization of a reader protein recognizing unmethylated H3K4.
    • 11 Ooi SK, Qiu C, Bernstein E et al. DNMT3L connects unmethylated lysine 4 of histone H3 to de novo methylation of DNA. Nature 448(7154), 714–717 (2007).
    • 12 Ruthenburg AJ, Allis CD, Wysocka J. Methylation of lysine 4 on histone H3: intricacy of writing and reading a single epigenetic mark. Mol. Cell. 25(1), 15–30 (2007).
    • 13 Greer EL, Shi Y. Histone methylation: a dynamic mark in health, disease and inheritance. Nat. Rev. Genet. 13(5), 343–357 (2012).•• An overview of histone methylation regulation and consequences on health and disease.
    • 14 Shulha HP, Cheung I, Guo Y, Akbarian S, Weng Z. Coordinated cell type-specific epigenetic remodeling in prefrontal cortex begins before birth and continues into early adulthood. PLoS Genet. 9(4), e1003433 (2013).
    • 15 Soshnikova N, Duboule D. Epigenetic regulation of Hox gene activation: the waltz of methyls. Bioessays 30(3), 199–202 (2008).
    • 16 Wynder C, Stalker L, Doughty ML. Role of H3K4 demethylases in complex neurodevelopmental diseases. Epigenomics 2(3), 407–418 (2010).•• Excellent review of H3K4 demethylases in neurodevelopmental disorders.
    • 17 Shen E, Shulha H, Weng Z, Akbarian S. Regulation of histone H3K4 methylation in brain development and disease. Philos. Trans. R Soc. Lond. B Biol. Sci. 369(1652), (2014).•• Excellent recent review of H3K4 writer and eraser enzymes in neurodevelopmental disorders, with an emphasis on human studies and animal models.
    • 18 Tunovic S, Barkovich J, Sherr EH, Slavotinek AM. De novo ANKRD11 and KDM1A gene mutations in a male with features of KBG syndrome and Kabuki syndrome. Am. J. Med. Genet. A 164(7), 1744–1749 (2014).
    • 19 Athanasakis E, Licastro D, Faletra F et al. Next generation sequencing in nonsyndromic intellectual disability: from a negative molecular karyotype to a possible causative mutation detection. Am. J. Med. Genet. A 164(1), 170–176 (2014).
    • 20 Kim HG, Kim HT, Leach NT et al. Translocations disrupting PHF21A in the Potocki-Shaffer syndrome region are associated with intellectual disability and craniofacial anomalies. Am. J. Hum. Genet. 91(1), 56–72 (2012).
    • 21 Laumonnier F, Holbert S, Ronce N et al. Mutations in PHF8 are associated with X linked mental retardation and cleft lip/cleft palate. J. Med. Genet. 42(10), 780–786 (2005).
    • 22 Allis CD, Berger SL, Cote J et al. New nomenclature for chromatin-modifying enzymes. Cell 131(4), 633–636 (2007).
    • 23 Shilatifard A. The COMPASS family of histone H3K4 methylases: mechanisms of regulation in development and disease pathogenesis. Annu. Rev. Biochem. 81, 65–95 (2012).
    • 24 Dou YL, Milne TA, Ruthenburg AJ et al. Regulation of MLL1 H3K4 methyltransferase activity by its core components. Nat. Struct. Mol. Biol. 13(8), 713–719 (2006).
    • 25 Zieminvanderpoel S, Mccabe NR, Gill HJ et al. Identification of a gene, Mll, that spans the breakpoint in 11q23 translocations associated with human leukemias. Proc. Natl Acad. Sci. USA 88(23), 10735–10739 (1991).
    • 26 Cimino G, Moir DT, Canaani O et al. Cloning of All-1, the locus involved in leukemias with the T(4–11)(Q21-Q23), T(9–11)(P22-Q23), and T(11–19)(Q23-P13) chromosome translocations. Cancer Res. 51(24), 6712–6714 (1991).
    • 27 Strom SP, Lozano R, Lee H et al. De novo variants in the KMT2A (MLL) gene causing atypical Wiedemann-Steiner syndrome in two unrelated individuals identified by clinical exome sequencing. BMC Med. Genet. 15, 49 (2014).
    • 28 Jones WD, Dafou D, Mcentagart M et al. De novo mutations in MLL cause Wiedemann-Steiner syndrome. Am. J. Hum. Genet. 91(2), 358–364 (2012).
    • 29 Miyake N, Koshimizu E, Okamoto N et al. MLL2 and KDM6A mutations in patients with Kabuki syndrome. Am. J. Med. Genet. A 161(9), 2234–2243 (2013).
    • 30 Zeleznik-Le NJ, Harden AM, Rowley JD. 11q23 translocations split the “AT-hook” cruciform DNA-binding region and the transcriptional repression domain from the activation domain of the mixed-lineage leukemia (MLL) gene. Proc. Natl Acad. Sci. USA 91(22), 10610–10614 (1994).
    • 31 Wysocka J, Swigut T, Milne TA et al. WDR5 associates with histone H3 methylated at K4 and is essential for H3K4 methylation and vertebrate development. Cell 121(6), 859–872 (2005).
    • 32 Musselman CA, Kutateladze TG. Handpicking epigenetic marks with PHD fingers. Nucleic Acids Res. 39(21), 9061–9071 (2011).
    • 33 Wang Z, Song J, Milne TA et al. Pro isomerization in MLL1 PHD3-bromo cassette connects H3K4me readout to CyP33 and HDAC-mediated repression. Cell 141(7), 1183–1194 (2010).
    • 34 Wang J, Muntean AG, Wu L, Hess JL. A subset of mixed lineage leukemia proteins has plant homeodomain (PHD)-mediated E3 ligase activity. J. Biol. Chem. 287(52), 43410–43416 (2012).
    • 35 Rossler T, Marschalek R. An alternative splice process renders the MLL protein either into a transcriptional activator or repressor. Pharmazie 68(7), 601–607 (2013).
    • 36 Johnson MB, Kawasawa YI, Mason CE et al. Functional and evolutionary insights into human brain development through global transcriptome analysis. Neuron 62(4), 494–509 (2009).
    • 37 Lim DA, Huang YC, Swigut T et al. Chromatin remodelling factor Mll1 is essential for neurogenesis from postnatal neural stem cells. Nature 458(7237), 529–533 (2009).
    • 38 Wang P, Lin C, Smith ER et al. Global analysis of H3K4 methylation defines MLL family member targets and points to a role for MLL1-mediated H3K4 methylation in the regulation of transcriptional initiation by RNA polymerase II. Mol. Cell. Biol. 29(22), 6074–6085 (2009).
    • 39 Denissov S, Hofemeister H, Marks H et al. Mll2 is required for H3K4 trimethylation on bivalent promoters in embryonic stem cells, whereas Mll1 is redundant. Development 141(3), 526–537 (2014).
    • 40 Lee JE, Wang CC, Xu SLY et al. H3K4 mono- and di-methyltransferase MLL4 is required for enhancer activation during cell differentiation. Elife 2, e01503 (2013).
    • 41 Hu DQ, Gao X, Morgan MA, Herz HM, Smith ER, Shilatifard A. The MLL3/MLL4 branches of the COMPASS family function as major histone H3K4 monomethylases at enhancers. Mol. Cell. Biol. 33(23), 4745–4754 (2013).
    • 42 Tan YC, Chow VT. Novel human HALR (MLL3) gene encodes a protein homologous to ALR and to ALL-1 involved in leukemia, and maps to chromosome 7q36 associated with leukemia and developmental defects. Cancer Detect. Prev. 25(5), 454–469 (2001).
    • 43 Ruault M, Brun ME, Ventura M, Roizes G, De Sario A. MLL3, a new human member of the TRX/MLL gene family, maps to 7q36, a chromosome region frequently deleted in myeloid leukaemia. Gene 284(1–2), 73–81 (2002).
    • 44 Kleefstra T, Kramer JM, Neveling K et al. Disruption of an EHMT1-associated chromatin-modification module causes intellectual disability. Am. J. Hum. Genet. 91(1), 73–82 (2012).
    • 45 Nagase T, Kikuno R, Ishikawa K, Hirosawa M, Ohara O. Prediction of the coding sequences of unidentified human genes. XVII. The complete sequences of 100 new cDNA clones from brain which code for large proteins in vitro. DNA Res. 7(2), 143–150 (2000).
    • 46 Cheng J, Blum R, Bowman C et al. A role for H3K4 monomethylation in gene repression and partitioning of chromatin readers. Mol. Cell. 53(6), 979–992 (2014).
    • 47 Ng SB, Bigham AW, Buckingham KJ et al. Exome sequencing identifies MLL2 mutations as a cause of Kabuki syndrome. Nat. Genet. 42(9), 790–793 (2010).
    • 48 Paulussen AD, Stegmann AP, Blok MJ et al. MLL2 mutation spectrum in 45 patients with Kabuki syndrome. Hum. Mutat. 32(2), E2018–E2025 (2011).
    • 49 Micale L, Augello B, Fusco C et al. Mutation spectrum of MLL2 in a cohort of Kabuki syndrome patients. Orphanet J. Rare Dis. 6, 38 (2011).
    • 50 Li Y, Bogershausen N, Alanay Y et al. A mutation screen in patients with Kabuki syndrome. Hum. Genet. 130(6), 715–724 (2011).
    • 51 Hannibal MC, Buckingham KJ, Ng SB et al. Spectrum of MLL2 (ALR) mutations in 110 cases of Kabuki syndrome. Am. J. Med. Genet. A 155A(7), 1511–1516 (2011).
    • 52 Banka S, Veeramachaneni R, Reardon W et al. How genetically heterogeneous is Kabuki syndrome?: MLL2 testing in 116 patients, review and analyses of mutation and phenotypic spectrum. Eur. J. Hum. Genet. 20(4), 381–388 (2012).
    • 53 Bogershausen N, Wollnik B. Unmasking Kabuki syndrome. Clin. Genet. 83(3), 201–211 (2013).
    • 54 Banka S, Howard E, Bunstone S et al. MLL2 mosaic mutations and intragenic deletion-duplications in patients with Kabuki syndrome. Clin. Genet. 83(5), 467–471 (2013).
    • 55 Dhar SS, Lee SH, Kan PY et al. Trans-tail regulation of MLL4-catalyzed H3K4 methylation by H4R3 symmetric dimethylation is mediated by a tandem PHD of MLL4. Genes Dev. 26(24), 2749–2762 (2012).
    • 56 Cho YW, Hong T, Hong S et al. PTIP associates with MLL3- and MLL4-containing histone H3 lysine 4 methyltransferase complex. J. Biol. Chem. 282(28), 20395–20406 (2007).
    • 57 Lee JH, Skalnik DG. CpG-binding protein (CXXC finger protein 1) is a component of the mammalian set1 histone H3-Lys(4) methyltransferase complex, the analogue of the yeast Set1/COMPASS complex. J. Biol. Chem. 280(50), 41725–41731 (2005).
    • 58 Wu M, Wang PF, Lee JS et al. Molecular regulation of H3K4 trimethylation by Wdr82, a component of human Set1/COMPASS. Mol. Cell. Biol. 28(24), 7337–7344 (2008).
    • 59 Ardehali MB, Mei A, Zobeck KL, Caron M, Lis JT, Kusch T. Drosophila Set1 is the major histone H3 lysine 4 trimethyltransferase with role in transcription. EMBO J. 30(14), 2817–2828 (2011).
    • 60 Hallson G, Hollebakken RE, Li T et al. dSet1 is the main H3K4 di- and tri-methyltransferase throughout Drosophila development. Genetics 190(1), 91–100 (2012).
    • 61 Mohan M, Herz HM, Smith ER et al. The COMPASS family of H3K4 methylases in Drosophila. Mol. Cell. Biol. 31(21), 4310–4318 (2011).
    • 62 Mersman DP, Du HN, Fingerman IM, South PF, Briggs SD. Charge-based interaction conserved within histone H3 lysine 4 (H3K4) methyltransferase complexes is needed for protein stability, histone methylation, and gene expression. J. Biol. Chem. 287(4), 2652–2665 (2012).
    • 63 Clouaire T, Webb S, Skene P et al. Cfp1 integrates both CpG content and gene activity for accurate H3K4me3 deposition in embryonic stem cells. Genes Dev. 26(15), 1714–1728 (2012).
    • 64 Clouaire T, Webb S, Bird A. Cfp1 is required for gene expression dependent H3K4me3 and H3K9 acetylation in embryonic stem cells. Genome Biol. 15(9), 451 (2014).
    • 65 Lauberth SM, Nakayama T, Wu X et al. H3K4me3 interactions with TAF3 regulate preinitiation complex assembly and selective gene activation. Cell 152(5), 1021–1036 (2013).
    • 66 Vermeulen M, Mulder KW, Denissov S et al. Selective anchoring of TFIID to nucleosomes by trimethylation of histone H3 lysine 4. Cell 131(1), 58–69 (2007).
    • 67 Shi Y, Matson C et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 119(7), 941–953 (2004).• First characterization of a histone demethylase.
    • 68 Sirmaci A, Spiliopoulos M, Brancati F et al. Mutations in ANKRD11 cause KBG syndrome, characterized by intellectual disability, skeletal malformations, and macrodontia. Am. J. Hum. Genet. 89(2), 289–294 (2011).
    • 69 Wang JX, Scully K, Zhu X et al. Opposing LSD1 complexes function in developmental gene activation and repression programmes. Nature 446(7138), 882–887 (2007).
    • 70 Wang J, Hevi S, Kurash JK et al. The lysine demethylase LSD1 (KDM1) is required for maintenance of global DNA methylation. Nat. Genet. 41(1), 125–129 (2009).
    • 71 Whyte WA, Bilodeau S, Orlando DA et al. Enhancer decommissioning by LSD1 during embryonic stem cell differentiation. Nature 482(7384), 221–225 (2012).
    • 72 Ballas N, Battaglioli E, Atouf F et al. Regulation of neuronal traits by a novel transcriptional complex. Neuron 31(3), 353–365 (2001).
    • 73 Andres ME, Burger C, Peral-Rubio MJ et al. CoREST: a functional corepressor required for regulation of neural-specific gene expression. Proc. Natl Acad. Sci. USA 96(17), 9873–9878 (1999).
    • 74 Hakimi MA, Dong Y, Lane WS, Speicher DW, Shiekhattar R. A candidate X-linked mental retardation gene is a component of a new family of histone deacetylase-containing complexes. J. Biol. Chem. 278(9), 7234–7239 (2003).
    • 75 Zibetti C, Adamo A, Binda C et al. Alternative splicing of the histone demethylase LSD1/KDM1 contributes to the modulation of neurite morphogenesis in the mammalian nervous system. J. Neurosci. 30(7), 2521–2532 (2010).
    • 76 Garcia-Bassets I, Kwon YS, Telese F et al. Histone methylation-dependent mechanisms impose ligand dependency for gene activation by nuclear receptors. Cell 128(3), 505–518 (2007).
    • 77 Iwase S, Lan F et al. The X-linked mental retardation gene SMCX/JARID1C defines a family of histone H3 lysine 4 demethylases. Cell 128(6), 1077–1088 (2007).• Discovery of the first H3K4me3 demethylase and first link between the dynamic nature of histone methylation and human cognitive development.
    • 78 Yamane K, Tateishi K, Klose RJ et al. PLU-1 is an H3K4 demethylase involved in transcriptional repression and breast cancer cell proliferation. Mol. Cell. 25(6), 801–812 (2007).
    • 79 Xiang Y, Zhu Z, Han G et al. JARID1B is a histone H3 lysine 4 demethylase up-regulated in prostate cancer. Proc. Natl Acad. Sci. USA 104(49), 19226–19231 (2007).
    • 80 Hayakawa T, Ohtani Y, Hayakawa N et al. RBP2 is an MRG15 complex component and down-regulates intragenic histone H3 lysine 4 methylation. Genes to Cells 12(6), 811–826 (2007).
    • 81 Klose RJ, Yan Q, Tothova Z et al. The retinoblastoma binding protein RBP2 is an H3K4 demethylase. Cell 128(5), 889–900 (2007).
    • 82 Christensen J, Agger K, Cloos PA et al. RBP2 belongs to a family of demethylases, specific for tri-and dimethylated lysine 4 on histone 3. Cell 128(6), 1063–1076 (2007).
    • 83 Tsukada Y, Fang J, Erdjument-Bromage H et al. Histone demethylation by a family of JmjC domain-containing proteins. Nature 439(7078), 811–816 (2006).
    • 84 Defeo-Jones D, Huang PS, Jones RE et al. Cloning of cDNAs for cellular proteins that bind to the retinoblastoma gene-product. Nature 352(6332), 251–254 (1991).
    • 85 Chen Z, Zang J, Whetstine J et al. Structural insights into histone demethylation by JMJD2 family members. Cell 125(4), 691–702 (2006).
    • 86 Lopez-Bigas N, Kisiel TA, Dewaal DC et al. Genome-wide analysis of the H3K4 histone demethylase RBP2 reveals a transcriptional program controlling differentiation. Mol. Cell. 31(4), 520–530 (2008).
    • 87 Shao GB, Chen JC, Zhang LP et al. Dynamic patterns of histone H3 lysine 4 methyltransferases and demethylases during mouse preimplantation development. In vitro Cell. Dev. Biol. Anim. 50(7), 603–613 (2014).
    • 88 Chicas A, Kapoor A, Wang X et al. H3K4 demethylation by Jarid1a and Jarid1b contributes to retinoblastoma-mediated gene silencing during cellular senescence. Proc. Natl Acad. Sci. USA 109(23), 8971–8976 (2012).
    • 89 Ditacchio L, Le HD, Vollmers C et al. Histone lysine demethylase JARID1a activates CLOCK-BMAL1 and influences the circadian clock. Science 333(6051), 1881–1885 (2011).
    • 90 Dey BK, Stalker L, Schnerch A, Bhatia M, Taylor-Papidimitriou J, Wynder C. The histone demethylase KDM5b/JARID1b plays a role in cell fate decisions by blocking terminal differentiation. Mol. Cell. Biol. 28(17), 5312–5327 (2008).
    • 91 Schmitz SU, Albert M, Malatesta M et al. Jarid1b targets genes regulating development and is involved in neural differentiation. EMBO J. 30(22), 4586–4600 (2011).
    • 92 Kidder BL, Hu G, Zhao K. KDM5B focuses H3K4 methylation near promoters and enhancers during embryonic stem cell self-renewal and differentiation. Genome Biol. 15(2), R32 (2014).
    • 93 Catchpole S, Spencer-Dene B, Hall D et al. PLU-1/JARID1B/KDM5B is required for embryonic survival and contributes to cell proliferation in the mammary gland and in ER+ breast cancer cells. Int. J. Oncol. 38(5), 1267–1277 (2011).
    • 94 Scibetta AG, Santangelo S, Coleman J et al. Functional analysis of the transcription repressor PLU-1/JARID1B. Mol. Cell. Biol. 27(20), 7220–7235 (2007).
    • 95 Xie L, Pelz C, Wang W et al. KDM5B regulates embryonic stem cell self-renewal and represses cryptic intragenic transcription. EMBO J. 30(8), 1473–1484 (2011).
    • 96 Li X, Liu L, Yang S et al. Histone demethylase KDM5B is a key regulator of genome stability. Proc. Natl Acad. Sci. USA 111(19), 7096–7101 (2014).
    • 97 Jensen LR, Amende M, Gurok U et al. Mutations in the JARID1C gene, which is involved in transcriptional regulation and chromatin remodeling, cause X-linked mental retardation. Am. J. Hum. Genet. 76(2), 227–236 (2005).
    • 98 Goncalves TF, Goncalves AP, Fintelman Rodrigues N, Dos Santos JM, Pimentel MM, Santos-Reboucas CB. KDM5C mutational screening among males with intellectual disability suggestive of X-linked inheritance and review of the literature. Eur. J. Med. Genet. 57(4), 138–144 (2014).
    • 99 Ropers HH, Hamel BC. X-linked mental retardation. Nat. Rev. Genet. 6(1), 46–57 (2005).
    • 100 Tahiliani M, Mei PC, Fang R et al. The histone H3K4 demethylase SMCX links REST target genes to X-linked mental retardation. Nature 447(7144), 601–605 (2007).
    • 101 Tzschach A, Lenzner S, Moser B et al. Novel JARID1C/SMCX mutations in patients with X-linked mental retardation. Hum. Mutat. 27(4), 389 (2006).
    • 102 Adegbola A, Gao H, Sommer S, Browning M. A novel mutation in JARID1C/SMCX in a patient with autism spectrum disorder (ASD). Am. J. Med. Genet. A 146A(4), 505–511 (2008).
    • 103 Simensen RJ, Rogers RC, Collins JS, Abidi F, Schwartz CE, Stevenson RE. Short-term memory deficits in carrier females with KDM5C mutations. Genet. Couns. 23(1), 31–40 (2012).
    • 104 Agulnik AI, Mitchell MJ, Lerner JL, Woods DR, Bishop CE. A mouse Y-chromosome gene encoded by a region essential for spermatogenesis and expression of male-specific minor histocompatibility antigens. Hum. Molec. Genet. 3(6), 873–878 (1994).
    • 105 Kent-First MG, Maffitt M, Muallem A et al. Gene sequence and evolutionary conservation of human SMCY. Nat. Genet. 14(2), 128–129 (1996).
    • 106 Xu J, Burgoyne PS, Arnold AP. Sex differences in sex chromosome gene expression in mouse brain. Hum. Mol. Genet. 11(12), 1409–1419 (2002).
    • 107 Aguilar-Valles A, Vaissiere T, Griggs EM et al. Methamphetamine-associated memory is regulated by a writer and an eraser of permissive histone methylation. Biol. Psychiatry 76(1), 57–65 (2014).
    • 108 Xu J, Deng X, Disteche CM. Sex-specific expression of the X-linked histone demethylase gene Jarid1c in brain. PLoS ONE 3(7), e2553 (2008).
    • 109 Poeta L, Fusco F, Drongitis D et al. A regulatory path associated with X-linked intellectual disability and epilepsy links KDM5C to the polyalanine expansions in ARX. Am. J. Hum. Genet. 92(1), 114–125 (2013).
    • 110 Outchkourov NS, Muino JM, Kaufmann K et al. Balancing of histone H3K4 methylation states by the Kdm5c/SMCX histone demethylase modulates promoter and enhancer function. Cell. Rep. 3(4), 1071–1079 (2013).
    • 111 Jensen LR, Bartenschlager H, Rujirabanjerd S et al. A distinctive gene expression fingerprint in mentally retarded male patients reflects disease-causing defects in the histone demethylase KDM5C. Pathogenetics 3(1), 2 (2010).
    • 112 Shaffer LG, Hecht JT, Ledbetter DH, Greenberg F. Familial interstitial deletion 11(p11.12p12) associated with parietal foramina, brachymicrocephaly, and mental retardation. Am. J. Med. Genet. 45(5), 581–583 (1993).
    • 113 Potocki L, Shaffer LG. Interstitial deletion of 11(p11.2p12): a newly described contiguous gene deletion syndrome involving the gene for hereditary multiple exostoses (EXT2). Am. J. Med. Genet. 62(3), 319–325 (1996).
    • 114 Iwase S, Januma A, Miyamoto K et al. Characterization of BHC80 in BRAF-HDAC complex, involved in neuron-specific gene repression. Biochem. Biophys. Res. Commun. 322(2), 601–608 (2004).
    • 115 Iwase S, Shono N, Honda A et al. A component of BRAF-HDAC complex, BHC80, is required for neonatal survival in mice. FEBS Lett. 580(13), 3129–3135 (2006).
    • 116 Klajn A, Ferrai C, Stucchi L et al. The rest repression of the neurosecretory phenotype is negatively modulated by BHC80, a protein of the BRAF/HDAC complex. J. Neurosci. 29(19), 6296–6307 (2009).
    • 117 Hakimi MA, Bochar DA, Chenoweth J, Lane WS, Mandel G, Shiekhattar R. A core-BRAF35 complex containing histone deacetylase mediates repression of neuronal-specific genes. Proc. Natl Acad. Sci. USA 99(11), 7420–7425 (2002).
    • 118 Shi YJ, Matson C, Iwase S, Baba T, Shi Y. Regulation of LSD1 histone demethylase activity by its associated factors. Mol. Cell. 19(6), 857–864 (2005).
    • 119 Siderius LE, Hamel BCJ, Van Bokhoven H et al. X-linked mental retardation associated with cleft lip palate maps to Xp11.3-q21.3. Am. J. Med. Genet. 85(3), 216–220 (1999).
    • 120 Koivisto AM, Ala-Mello S, Lemmela S, Komu HA, Rautio J, Jarvela I. Screening of mutations in the PHF8 gene and identification of a novel mutation in a Finnish family with XLMR and cleft lip/cleft palate. Clin. Genet. 72(2), 145–149 (2007).
    • 121 Abidi FE, Miano MG, Murray JC, Schwartz CE. A novel mutation in the PHF8 gene is associated with X-linked mental retardation with cleft lip/cleft palate. Clin. Genet. 72(1), 19–22 (2007).
    • 122 Qiao Y, Liu X, Harvard C et al. Autism-associated familial microdeletion of Xp11.22. Clin. Genet. 74(2), 134–144 (2008).
    • 123 Qi HH, Sarkissian M, Hu GQ et al. Histone H4K20/H3K9 demethylase PHF8 regulates zebrafish brain and craniofacial development. Nature 466(7305), 503–507 (2010).
    • 124 Loenarz C, Ge W, Coleman ML et al. PHF8, a gene associated with cleft lip/palate and mental retardation, encodes for an Nepsilon-dimethyl lysine demethylase. Hum. Mol. Genet. 19(2), 217–222 (2010).
    • 125 Fortschegger K, De Graaf P, Outchkourov NS, Van Schaik FM, Timmers HT, Shiekhattar R. PHF8 targets histone methylation and RNA polymerase II to activate transcription. Mol. Cell. Biol. 30(13), 3286–3298 (2010).
    • 126 Feng WJ, Yonezawa M, Ye J, Jenuwein T, Grummt I. PHF8 activates transcription of rRNA genes through H3K4me3 binding and H3K9me1/2 demethylation. Nat. Struct. Mol. Biol. 17(4), 445–U483 (2010).
    • 127 Kleine-Kohlbrecher D, Christensen J, Vandamme J et al. A functional link between the histone demethylase PHF8 and the transcription factor ZNF711 in X-linked mental retardation. Mol. Cell. 38(2), 165–178 (2010).
    • 128 Qiu JH, Shi GA, Jia YH et al. The X-linked mental retardation gene PHF8 is a histone demethylase involved in neuronal differentiation. Cell Res. 20(8), 908–918 (2010).
    • 129 Horton JR, Upadhyay AK, Qi HH, Zhang X, Shi Y, Cheng XD. Enzymatic and structural insights for substrate specificity of a family of jumonji histone lysine demethylases. Nat. Struct. Mol. Biol. 17(1), U38–U52 (2010).
    • 130 Asensio-Juan E, Gallego C, Martinez-Balbas MA. The histone demethylase PHF8 is essential for cytoskeleton dynamics. Nucleic Acids Res. 40(19), 9429–9440 (2012).
    • 131 Lupo G, Harris WA, Lewis KE. Mechanisms of ventral patterning in the vertebrate nervous system. Nat. Rev. Neurosci. 7(2), 103–114 (2006).
    • 132 Cau E, Blader P. Notch activity in the nervous system: to switch or not switch? Neural Dev. 4, 36 (2009).
    • 133 Tsai MC, Manor O, Wan Y et al. Long noncoding RNA as modular scaffold of histone modification complexes. Science 329(5992), 689–693 (2010).
    • 134 Wang KC, Yang YW, Liu B et al. A long noncoding RNA maintains active chromatin to coordinate homeotic gene expression. Nature 472(7341), 120–124 (2011).
    • 135 Yang YW, Flynn RA, Chen Y et al. Essential role of lncRNA binding for WDR5 maintenance of active chromatin and embryonic stem cell pluripotency. Elife 3, e02046 (2014).
    • 136 Shulha HP, Crisci JL, Reshetov D et al. Human-specific histone methylation signatures at transcription start sites in prefrontal neurons. PLoS Biol. 10(11), e1001427 (2012).
    • 137 Bell CG, Wilson GA, Beck S. Human-specific CpG ‘beacons’ identify human-specific prefrontal cortex H3K4me3 chromatin peaks. Epigenomics 6(1), 21–31 (2014).
    • 138 Gupta S, Kim SY, Artis S et al. Histone methylation regulates memory formation. J. Neurosci. 30(10), 3589–3599 (2010).
    • 139 Kerimoglu C, Agis-Balboa RC, Kranz A et al. Histone-methyltransferase MLL2 (KMT2B) is required for memory formation in mice. J. Neurosci. 33(8), 3452–3464 (2013).
    • 140 Cao F, Townsend EC, Karatas H et al. Targeting MLL1 H3K4 methyltransferase activity in mixed-lineage leukemia. Mol. Cell. 53(2), 247–261 (2014).
    • 141 Grembecka J, He S, Shi A et al. Menin-MLL inhibitors reverse oncogenic activity of MLL fusion proteins in leukemia. Nat. Chem. Biol. 8(3), 277–284 (2012).
    • 142 Helin K, Dhanak D. Chromatin proteins and modifications as drug targets. Nature 502(7472), 480–488 (2013).