1,950
Views
42
CrossRef citations to date
0
Altmetric
Review

Emerging role of SETDB1 as a therapeutic target

, , , , , , & show all
Pages 319-331 | Received 14 Jul 2016, Accepted 21 Dec 2016, Published online: 20 Jan 2017

References

  • Goldberg AD, Allis CD, Bernstein E. Epigenetics: a landscape takes shape . Cell. 2007;128:635–638.
  • Bernstein BE, Meissner A, Lander ES. The mammalian epigenome. Cell. 2007;128:669–681.
  • Bird A. Perceptions of epigenetics. Nature. 2007;447:396–398.
  • Saito Y, Hibino S, Saito H. Alterations of epigenetics and microRNA in hepatocellular carcinoma. Hepatol Res. 2014;44:31–42.
  • Sharma S, Kelly TK, Jones PA. Epigenetics in cancer. Carcinogenesis. 2010;31:27–36.
  • Vendetti FP, Rudin CM. Epigenetic therapy in non-small-cell lung cancer: targeting DNA methyltransferases and histone deacetylases. Expert Opin Biol Ther. 2013;13:1273–1285.
  • Stylianou E. Epigenetics: the fine-tuner in inflammatory bowel disease? Curr Opin Gastroenterol. 2013;29:370–377.
  • Kwon MJ, Shin YK. Epigenetic regulation of cancer-associated genes in ovarian cancer. Int J Mol Sci. 2011;12:983–1008.
  • Baylin SB, Ohm JE. Epigenetic gene silencing in cancer - a mechanism for early oncogenic pathway addiction? Nat Rev Cancer. 2006;6:107–116.
  • Yoo CB, Jones PA. Epigenetic therapy of cancer: past, present and future. Nat Rev Drug Discov. 2006;5:37–50.
  • Herz HM, Garruss A, Shilatifard A. SET for life: biochemical activities and biological functions of SET domain-containing proteins. Trends Biochem Sci. 2013;38:621–639.
  • Harte PJ, Wu W, Carrasquillo MM, et al. Assignment of a novel bifurcated SET domain gene, SETDB1, to human chromosome band 1q21 by in situ hybridization and radiation hybrids. Cytogenet Cell Genet. 1999;84:83–86.
  • Blackburn ML, Chansky HA, Zielinska-Kwiatkowska A, et al. Genomic structure and expression of the mouse ESET gene encoding an ERG-associated histone methyltransferase with a SET domain. Biochim Biophys Acta. 2003;1629:8–14.
  • Rea S, Eisenhaber F, O’Carroll D, et al. Regulation of chromatin structure by site-specific histone H3 methyltransferases. Nature. 2000;406:593–599.
  • Schultz DC, Ayyanathan K, Negorev D, et al. SETDB1: a novel KAP-1-associated histone H3, lysine 9-specific methyltransferase that contributes to HP1-mediated silencing of euchromatic genes by KRAB zinc-finger proteins. Genes Dev. 2002;16:919–932.
  • Kang YK. SETDB1 in early embryos and embryonic stem cells. Curr Issues Mol Biol. 2015;17:1–10.
  • Bird A. Molecular biology. Methylation talk between histones and DNA. Science. 2001;294:2113–2115.
  • Ponting CP. Tudor domains in proteins that interact with RNA. Trends Biochem Sci. 1997;22:51–52.
  • Bird AP, Wolffe AP. Methylation-induced repression–belts, braces, and chromatin. Cell. 1999;99:451–454.
  • Lawson KA, Teteak CJ, Gao J, et al. ESET histone methyltransferase regulates osteoblastic differentiation of mesenchymal stem cells during postnatal bone development. FEBS Lett. 2013;587:3961–3967.
  • Terns MP, Terns RM. Macromolecular complexes: SMN–the master assembler. Curr Biol. 2001;11:R862–4.
  • Morris GE. The Cajal body. Biochim Biophys Acta. 2008;1783:2108–2115.
  • Yang L, Mei Q, Zielinska-Kwiatkowska A, et al. An ERG (ets-related gene)-associated histone methyltransferase interacts with histone deacetylases 1/2 and transcription co-repressors mSin3A/B. Biochem J. 2003;369:651–657.
  • Bucher P. Weight matrix descriptions of four eukaryotic RNA polymerase II promoter elements derived from 502 unrelated promoter sequences. J Mol Biol. 1990;212:563–578.
  • Huang S. Histone methyltransferases, diet nutrients and tumour suppressors. Nat Rev Cancer. 2002;2:469–476.
  • Tachibana K, Gotoh E, Kawamata N, et al. Analysis of the subcellular localization of the human histone methyltransferase SETDB1. Biochem Biophys Res Commun. 2015;465:725–731.
  • Cho S, Park JS, Kang YK. Regulated nuclear entry of over-expressed SETDB1. Genes Cells. 2013;18:694–703.
  • Loyola A, Tagami H, Bonaldi T, et al. The HP1alpha-CAF1-SETDB1-containing complex provides H3K9me1 for Suv39-mediated K9me3 in pericentric heterochromatin. EMBO Rep. 2009;10:769–775.
  • Matsui T, Leung D, Miyashita H, et al. Proviral silencing in embryonic stem cells requires the histone methyltransferase ESET. Nature. 2010;464:927–931.
  • Minkovsky A, Sahakyan A, Rankin-Gee E, et al. The MBD1-ATF7IP-SETDB1 pathway contributes to the maintenance of X chromosome inactivation. Epigenetics Chromatin. 2014;7:12.
  • Song YJ, Choi JH, Lee H. Setdb1 is required for myogenic differentiation of C2C12 myoblast cells via maintenance of Myod expression. Mol Cells. 2015;38:362–372.
  • Yang L, Lawson KA, Teteak CJ, et al. ESET histone methyltransferase is essential to hypertrophic differentiation of growth plate chondrocytes and formation of epiphyseal plates. Dev Biol. 2013;380(1):99–110.
  • Wong CM, Wei L, Law CT, et al. Up-regulation of histone methyltransferase SETDB1 by multiple mechanisms in hepatocellular carcinoma promotes cancer metastasis. Hepatology. 2016;63:474–487.
  • Fischle W, Wang Y, Allis CD. Histone and chromatin cross-talk. Curr Opin Cell Biol. 2003;15:172–183.
  • Li F, Ye B, Hong L, et al. Epigenetic modifications of histone h4 in lung neuroendocrine tumors. Appl Immunohistochem Mol Morphol. 2011;19:389–394.
  • Aagaard L, Laible G, Selenko P, et al. Functional mammalian homologues of the Drosophila PEV-modifier Su(var)3-9 encode centromere-associated proteins which complex with the heterochromatin component M31. Embo J. 1999;18:1923–1938.
  • Tachibana M, Sugimoto K, Nozaki M, et al. G9a histone methyltransferase plays a dominant role in euchromatic histone H3 lysine 9 methylation and is essential for early embryogenesis. Genes Dev. 2002;16:1779–1791.
  • Ogawa H, Ishiguro K, Gaubatz S, et al. A complex with chromatin modifiers that occupies E2F- and Myc-responsive genes in G0 cells. Science. 2002;296:1132–1136.
  • Yang L, Xia L, Wu DY, et al. Molecular cloning of ESET, a novel histone H3-specific methyltransferase that interacts with ERG transcription factor. Oncogene. 2002;21:148–152.
  • Barski A, Cuddapah S, Cui K, et al. High-resolution profiling of histone methylations in the human genome. Cell. 2007;129:823–837.
  • Wang H, An W, Cao R, et al. mAM facilitates conversion by ESET of dimethyl to trimethyl lysine 9 of histone H3 to cause transcriptional repression. Mol Cell. 2003;12:475–487.
  • Binda O, LeRoy G, Bua DJ, et al. Trimethylation of histone H3 lysine 4 impairs methylation of histone H3 lysine 9: regulation of lysine methyltransferases by physical interaction with their substrates. Epigenetics. 2010;5:767–775.
  • Keniry A, Gearing LJ, Jansz N, et al. Setdb1-mediated H3K9 methylation is enriched on the inactive X and plays a role in its epigenetic silencing. Epigenetics Chromatin. 2016;9:16.
  • Sripathy SP, Stevens J, Schultz DC. The KAP1 corepressor functions to coordinate the assembly of de novo HP1-demarcated microenvironments of heterochromatin required for KRAB zinc finger protein-mediated transcriptional repression. Mol Cell Biol. 2006;26:8623–8638.
  • Ryan RF, Schultz DC, Ayyanathan K, et al. KAP-1 corepressor protein interacts and colocalizes with heterochromatic and euchromatic HP1 proteins: a potential role for Krüppel-associated box-zinc finger proteins in heterochromatin-mediated gene silencing. Mol Cell Biol. 1999;19:4366–4378.
  • Lechner MS, Begg GE, Speicher DW, et al. Molecular determinants for targeting heterochromatin protein 1-mediated gene silencing: direct chromoshadow domain-KAP-1 corepressor interaction is essential. Mol Cell Biol. 2000;20:6449–6465.
  • Yang L, Mei Q, Zielinska-Kwiatkowska A, et al. An ERG (ets-related gene)-associated histone methyltransferase interacts with histone deacetylases 1/2 and transcription co-repressors mSin3A/B. Biochem J. 2003;369:651–657.
  • Hammond EM, Mandell DJ, Salim A, et al. Genome-wide analysis of p53 under hypoxic conditions. Mol Cell Biol. 2006;26:3492–3504.
  • Cho S, Park JS, Kang YK. AGO2 and SETDB1 cooperate in promoter-targeted transcriptional silencing of the androgen receptor gene. Nucleic Acids Res. 2014;42:13545–13556.
  • Li H, Rauch T, Chen ZX, et al. The histone methyltransferase SETDB1 and the DNA methyltransferase DNMT3A interact directly and localize to promoters silenced in cancer cells. J Biol Chem. 2006;281:19489–19500.
  • Thompson PJ, Dulberg V, Moon KM, et al. hnRNP K coordinates transcriptional silencing by SETDB1 in embryonic stem cells. PLoS Genet. 2015;11:e1004933.
  • Bernardi R, Pandolfi PP. Structure, dynamics and functions of promyelocytic leukaemia nuclear bodies. Nat Rev Mol Cell Biol. 2007;8:1006–1016.
  • De Thé H, Le Bras M, Lallemand-Breitenbach V. The cell biology of disease: acute promyelocytic leukemia, arsenic, and PML bodies. J Cell Biol. 2012;198:11–21.
  • Cho S, Park JS, Kang YK. Dual functions of histone-lysine N-methyltransferase SETDB1 protein at promyelocytic leukemia-nuclear body (PML-NB): maintaining PML-NB structure and regulating the expression of its associated genes. J Biol Chem. 2011;286:41115–41124.
  • Cairo S, De Falco F, Pizzo M, et al. PML interacts with Myc, and Myc target gene expression is altered in PML-null fibroblasts. Oncogene. 2005;24:2195–2203.
  • Cho S, Park JS, Kwon S, et al. Dynamics of SETDB1 expression in early mouse development. Gene Expr Patterns. 2012;12:213–218.
  • Dodge JE, Kang YK, Beppu H, et al. Histone H3-K9 methyltransferase ESET is essential for early development. Mol Cell Biol. 2004;24:2478–2486.
  • Tachibana M, Ueda J, Fukuda M, et al. Histone methyltransferases G9a and GLP form heteromeric complexes and are both crucial for methylation of euchromatin at H3-K9. Genes Dev. 2005;19:815–826.
  • Murry CE, Keller G. Differentiation of embryonic stem cells to clinically relevant populations: lessons from embryonic development. Cell. 2008;132:661–680.
  • Yuan P, Han J, Guo G, et al. Eset partners with Oct4 to restrict extraembryonic trophoblast lineage potential in embryonic stem cells. Genes Dev. 2009;23:2507–2520.
  • Chen J, Liu H, Liu J, et al. H3K9 methylation is a barrier during somatic cell reprogramming into iPSCs. Nat Genet. 2013;45:34–42.
  • Ceol CJ, Houvras Y, Jane-Valbuena J, et al. The histone methyltransferase SETDB1 is recurrently amplified in melanoma and accelerates its onset. Nature. 2011;471:513–517.
  • Sepsa A, Levidou G, Gargalionis A, et al. Emerging role of linker histone variant H1x as a biomarker with prognostic value in astrocytic gliomas.A multivariate analysis including trimethylation of H3K9 and H4K20. PLoS One. 2015;10:e0115101.
  • Sun Y, Wei M, Ren SC, et al. Histone methyltransferase SETDB1 is required for prostate cancer cell proliferation, migration and invasion. Asian J Androl. 2014;16:319–324.
  • Lindgren D, Sjödahl G, Lauss M, et al. Integrated genomic and gene expression profiling identifies two major genomic circuits in urothelial carcinoma. PLoS One. 2012;7:e38863.
  • Sun QY, Ding LW, Xiao JF, et al. SETDB1 accelerates tumourigenesis by regulating the WNT signalling pathway. J Pathol. 2015;235:559–570.
  • Zhang H, Cai K, Wang J, et al. MiR-7, inhibited indirectly by lincRNA HOTAIR, directly inhibits SETDB1 and reverses the EMT of breast cancer stem cells by downregulating the STAT3 pathway. Stem Cells. 2014;32:2858–2868.
  • Regina C, Compagnone M, Peschiaroli A, et al. Setdb1, a novel interactor of ΔNp63, is involved in breast tumorigenesis. Oncotarget. 2016;7:28836-28848.
  • Fei Q, Shang K, Zhang J, et al. Histone methyltransferase SETDB1 regulates liver cancer cell growth through methylation of p53. Nat Commun. 2015;6:8651.
  • Liu L, Kimball S, Liu H, et al. Genetic alterations of histone lysine methyltransferases and their significance in breast cancer. Oncotarget. 2015;6:2466–2482.
  • Crum CP, McKeon FD. p63 in epithelial survival, germ cell surveillance, and neoplasia. Annu Rev Pathol. 2010;5:349–371.
  • Fang Y(1), Xue JL, Shen Q, et al. MicroRNA-7 inhibits tumor growth and metastasis by targeting the phosphoinositide 3-kinase/Akt pathway in hepatocellular carcinoma. Hepatology. 2012;55:1852–1862.
  • Ma L, Young J, Prabhala H, et al. miR-9, a MYC/MYCN-activated microRNA, regulates E-cadherin and cancer metastasis. Nat Cell Biol. 2010;12:247–256.
  • Bartkova J, Horejsí Z, Koed K, et al. DNA damage response as a candidate anti-cancer barrier in early human tumorigenesis. Nature. 2005;434:864–870.
  • Gorgoulis VG, Vassiliou LV, Karakaidos P, et al. Activation of the DNA damage checkpoint and genomic instability in human precancerous lesions. Nature. 2005;434:907–913.
  • Wang S, Tian C, Xing G, et al. ARF-dependent regulation of ATM and p53 associated KZNF (Apak) protein activity in response to oncogenic stress. FEBS Lett. 2010;584:3909–3915.
  • Olcina MM, Leszczynska KB, Senra JM, et al. H3K9me3 facilitates hypoxia-induced p53-dependent apoptosis through repression of Apak. Oncogene. 2016;35:793–799.
  • Spyropoulou A, Gargalionis A, Dalagiorgou G, et al. Role of histone lysine methyltransferases SUV39H1 and SETDB1 in gliomagenesis: modulation of cell proliferation, migration, and colony formation. Neuromolecular Med. 2014;16:70–82.
  • Kriegel AJ, Liu Y, Fang Y, et al. The miR-29 family: genomics, cell biology, and relevance to renal and cardiovascular injury. Physiol Genomics. 2012;44:237–244.
  • Rodriguez-Paredes M, De Paz AM, Simó-Riudalbas L, et al. Gene amplification of the histone methyltransferase SETDB1 contributes to human lung tumorigenesis. Oncogene. 2014;33:2807–2813.
  • Lafuente-Sanchis A, Zúñiga Á, Galbis JM, et al. Prognostic value of ERCC1, RRM1, BRCA1 and SETDB1 in early stage of non-small cell lung cancer. Clin Transl Oncol. 2015;18:798-804.
  • Inoue Y, Matsuura S, Kurabe N, et al. Clinicopathological and survival analysis of Japanese patients with resected non-small-cell lung cancer harboring NKX2-1, SETDB1, MET, HER2, SOX2, FGFR1, or PIK3CA gene amplification. J Thorac Oncol. 2015;10:1590–1600.
  • Wu PC, Lu JW, Yang JY, et al. H3K9 histone methyltransferase, KMT1E/SETDB1, cooperates with the SMAD2/3 pathway to suppress lung cancer metastasis. Cancer Res. 2014;74:7333–7343.
  • Na HH, Noh HJ, Cheong HM, et al. SETDB1 mediated FosB expression increases the cell proliferation rate during anticancer drug therapy. BMB Rep. 2016;49:238–243.
  • Siderovski DP, Blum S, Forsdyke RE, et al. A set of human putative lymphocyte G0/G1 switch genes includes genes homologous to rodent cytokine and zinc finger protein-encoding genes. DNA Cell Biol. 1990;9:579–587.
  • Lewis EB. The bithorax complex: the first fifty years. Int J Dev Biol. 1998;42:403–415.
  • Miura S, Maesawa C, Shibazaki M, et al. Immunohistochemistry for histone h3 lysine 9 methyltransferase and demethylase proteins in human melanomas. Am J Dermatopathol. 2014;36:211–216.
  • Kostaki M, Manona AD, Stavraka I, et al. High-frequency p16(INK) (4A) promoter methylation is associated with histone methyltransferase SETDB1 expression in sporadic cutaneous melanoma. Exp Dermatol. 2014;23:332–338.
  • Li Q, Li Y, Wang Y, et al. Quantitative proteomic study of human prostate cancer cells with different metastatic potentials. Int J Oncol. 2016;48:1437–1446.
  • Yoshikawa Y, Sato A, Tsujimura T, et al. Biallelic germline and somatic mutations in malignant mesothelioma: multiple mutations in transcription regulators including mSWI/SNF genes. Int J Cancer. 2015;136:560–571.
  • Kang HC, Kim HK, Lee S, et al. Whole exome and targeted deep sequencing identify genome-wide allelic loss and frequent SETDB1 mutations in malignant pleural mesotheliomas. Oncotarget. 2016;7:8321–8331.
  • Guo G, Chmielecki J, Goparaju C, et al. Whole-exome sequencing reveals frequent genetic alterations in BAP1, NF2, CDKN2A, and CUL1 in malignant pleural mesothelioma. Cancer Res. 2015;75:264–269.
  • Macgregor S, Montgomery GW, Liu JZ, et al. Genome-wide association study identifies a new melanoma susceptibility locus at 1q21.3. Nat Genet. 2011;43:1114–1118.
  • Ryu H, Lee J, Hagerty SW, et al. ESET/SETDB1 gene expression and histone H3 (K9) trimethylation in Huntington’s disease. Proc Natl Acad Sci U S A. 2006;103:19176–19181.
  • Kennedy BJ, Torkelson JL. Long-term follow-up of stage III testicular carcinoma treated with mithramycin (plicamycin). Med Pediatr Oncol. 1995;24:327–328.
  • Dutcher JP, Coletti D, Paietta E, et al. A pilot study of alpha-interferon and plicamycin for accelerated phase of chronic myeloid leukemia. Leuk Res. 1997;21:375–380.
  • Albertini V, Jain A, Vignati S, et al. Novel GC-rich DNA-binding compound produced by a genetically engineered mutant of the mithramycin producer Streptomyces argillaceus exhibits improved transcriptional repressor activity: implications for cancer therapy. Nucleic Acids Res. 2006;34:1721–1734.
  • Plicamycin. NLM USA. Clinical and research information on drug-induced liver injury. Available from:http://livertox.nih.gov/Plicamycin.htm
  • Monto RW, Talley RW, Caldwell MJ, et al. Observations on the mechanism of hemorrhagic toxicity in mithramycin (NSC 24559) therapy. Cancer Res. 1969;29:697–704.
  • ClinicalTrials.gov. Mithramycin for lung, esophagus, and other chest cancers 2012. Available from:https://clinicaltrials.gov/ct2/show/NCT01624090
  • Vizcaíno C, Mansilla S, Núñez LE, et al. Novel mithramycins abrogate the involvement of protein factors in the transcription of cell cycle control genes. Biochem Pharmacol. 2012;84:1133–1142.
  • Vizcaíno C, Mansilla S, Portugal J. Sp1 transcription factor: a long-standing target in cancer chemotherapy. Pharmacol Ther. 2015;152:111–124.
  • Spengler ML, Brattain MG. Sumoylation inhibits cleavage of Sp1 N-terminal negative regulatory domain and inhibits Sp1-dependent transcription. J Biol Chem. 2006;281:5567–5574.
  • Remsing LL, González AM, Nur-e-Alam M, et al. Mithramycin SK, a novel antitumor drug with improved therapeutic index, mithramycin SA, and demycarosyl-mithramycin SK: three new products generated in the mithramycin producer Streptomyces argillaceus through combinatorial biosynthesis. J Am Chem Soc. 2003;125:5745–5753.
  • Malek A, Núñez LE, Magistri M, et al. Modulation of the activity of Sp transcription factors by mithramycin analogues as a new strategy for treatment of metastatic prostate cancer. PLoS One. 2012;7:e35130.
  • Scott D, Chen JM, Bae Y, et al. Semi-synthetic mithramycin SA derivatives with improved anticancer activity. Chem Biol Drug Des. 2013;81:615–624.
  • Exploring novel opportunities for aureolic acids as anticancer drugs. Biochemistry and pharmacology: openAcess. Available from:http://www.omicsgroup.org/journals/exploring-novel-opportunities-for-aureolic-acids-as-anticancer-drugs-2167-0501.1000e140.php?aid=11095
  • Lee JK, Kim KC. DZNep, inhibitor of S-adenosylhomocysteine hydrolase,down-regulates expression of SETDB1 H3K9me3 HMTase in human lung cancer cells. Biochem Biophys Res Commun. 2013;438:647–652.
  • Bray M, Driscoll J, Huggins JW. Treatment of lethal Ebola virus infection in mice with a single dose of an S-adenosyl-L-homocysteine hydrolase inhibitor. Antiviral Res. 2000;45:135–147.
  • Mayers DL, Mikovits JA, Joshi B, et al. Anti-human immunodeficiency virus 1 (HIV-1) activities of 3-deazaadenosine analogs: increased potency against 3ʹ-azido-3ʹ-deoxythymidine-resistant HIV-1 strains. Proc Natl Acad Sci U S A. 1995;92:215–219.
  • De Clercq E, Cools M, Balzarini J, et al. Broad-spectrum antiviral activities of neplanocin A, 3-deazaneplanocin A, and their 5ʹ-nor derivatives. Antimicrob Agents Chemother. 1989;33:1291–1297.
  • Noh HJ, Kim KA, Kim KC. p53 down-regulates SETDB1 gene expression during paclitaxel induced-cell death. Biochem Biophys Res Commun. 2014;446:43–48.
  • Gambari R, Brognara E, Spandidos DA, et al. Targeting oncomiRNAs and mimicking tumor suppressor miRNAs: new trends in the development of miRNA therapeutic strategies in oncology (review). Int J Oncol. 2016;49:5–32.
  • Sun L, Fang J. E3-independent constitutive monoubiquitination complements histone methyltransferase activity of SETDB1. Mol Cell. 2016;62:958–966.
  • Researchers find new way to control genes often involved in cancer growth. Discovery of controlling SETDB1 through monoubiquitination could lead to new target therapies. Sci Daily. 2016. Available from: https://www.sciencedaily.com/releases/2016/07/160708105517.htm
  • Okamura M, Inagaki T, Tanaka T, et al. Role of histone methylation and demethylation in adipogenesis and obesity. Organogenesis. 2010;6:24–32.
  • Wakabayashi K, Okamura M, Tsutsumi S, et al. The peroxisome proliferator-activated receptor gamma/retinoid X receptor alpha heterodimer targets the histone modification enzyme PR-SET7/SETD8 gene and regulates adipogenesis through a positive feedback loop. Mol Cell Biol. 2009;29:3544–3555.
  • Jindal RD, Pillai AK, Mahadik SP, et al. Decreased BDNF in patients with antipsychotic naïve first episode schizophrenia. Schizophr Res. 2010;119:47–51.
  • Guidotti A, Auta J, Davis JM, et al. Decrease in reelin and glutamic acid decarboxylase67 (GAD67) expression in schizophrenia and bipolar disorder: a postmortem brain study. Arch Gen Psychiatry. 2000;57:1061–1069.
  • Chase KA, Gavin DP, Guidotti A, et al. Histone methylation at H3K9: evidence for a restrictive epigenome in schizophrenia. Schizophr Res. 2013;149:15–20.
  • Chase KA, Rosen C, Rubin LH, et al. Evidence of a sex-dependent restrictive epigenome in schizophrenia. J Psychiatr Res. 2015;65:87–94.
  • Cukier HN, Lee JM, Ma D, et al. The expanding role of MBD genes in autism: identification of a MECP2 duplication and novel alterations in MBD5, MBD6, and SETDB1. Autism Res. 2012;5:385–397.
  • Karberg S. Switching on epigenetic therapy. Cell. 2009;139:1029–1031.

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.