全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Epigenetics in Friedreich's Ataxia: Challenges and Opportunities for Therapy

DOI: 10.1155/2013/852080

Full-Text   Cite this paper   Add to My Lib

Abstract:

Friedreich's ataxia (FRDA) is an autosomal recessive neurodegenerative disorder caused by homozygous expansion of a GAA·TTC trinucleotide repeat within the first intron of the FXN gene, leading to reduced FXN transcription and decreased levels of frataxin protein. Recent advances in FRDA research have revealed the presence of several epigenetic modifications that are either directly or indirectly involved in this FXN gene silencing. Although epigenetic marks may be inherited from one generation to the next, modifications of DNA and histones can be reversed, indicating that they are suitable targets for epigenetic-based therapy. Unlike other trinucleotide repeat disorders, such as Huntington disease, the large expansions of GAA·TTC repeats in FRDA do not produce a change in the frataxin amino acid sequence, but they produce reduced levels of normal frataxin. Therefore, transcriptional reactivation of the FXN gene provides a good therapeutic option. The present paper will initially focus on the epigenetic changes seen in FRDA patients and their role in the silencing of FXN gene and will be concluded by considering the potential epigenetic therapies. 1. Introduction FRDA is a rare autosomal recessive neurodegenerative disorder that affects approximately 1-2 in 50,000 Caucasians [1]. In 96% of FRDA patients, the disease is caused by homozygous expansion of GAA·TTC repeats in intron 1 of the FXN gene [2]. Unaffected individuals have up to 40 GAA·TTC repeats, with a premutation range from 41 to 65 GAA repeats. The affected individuals contain 66 to 1700 GAA·TTC repeats [3], most commonly between 600 and 900 GAA·TTC repeats. In most cases, the GAA·TTC repeat number of the smaller allele is directly related to the age of onset and the severity of the disease. However, a small proportion of patients (approximately 4%) are compound heterozygous, having one allele with a GAA·TTC repeat expansion and the other allele with an inactivating (or loss-of-function) intragenic mutation, such as a point mutation [4, 5] or a deletion/duplication [6–9]. To date, no confirmed FRDA patients have been identified without at least one GAA·TTC repeat expansion. The exact mechanism underlying the GAA·TTC repeat expansion in FRDA is not fully understood, but evidence has been put forward for the involvement of abnormal DNA replication, transcription, or repair [10–12]. In FRDA patients, the expanded GAA·TTC repeats produce a marked reduction in the mitochondrial protein frataxin, ranging from 4% to 29% of normal levels [13]. However, asymptomatic carriers produce about 50% of

References

[1]  M. Cossée, M. Schmitt, V. Campuzano et al., “Evolution of the Friedreich's ataxia trinucleotide repeat expansion: founder effect and premutations,” Proceedings of the National Academy of Sciences of the United States of America, vol. 94, no. 14, pp. 7452–7457, 1997.
[2]  V. Campuzano, L. Montermini, M. D. Moltò et al., “Friedreich's ataxia: autosomal recessive disease caused by an intronic GAA triplet repeat expansion,” Science, vol. 271, no. 5254, pp. 1423–1427, 1996.
[3]  M. Pandolfo, “The molecular basis of Friedreich ataxia,” Advances in Experimental Medicine and Biology, vol. 516, pp. 99–118, 2002.
[4]  M. Cossee, A. Durr, M. Schmitt, et al., “Friedreich's ataxia: point mutations and clinical presentation of compound heterozygotes,” Annals of Neurology, vol. 45, no. 2, pp. 200–206, 1999.
[5]  C. Gellera, B. Castellotti, C. Mariotti et al., “Frataxin gene point mutations in Italian Friedreich ataxia patients,” Neurogenetics, vol. 8, no. 4, pp. 289–299, 2007.
[6]  E. C. Deutsch, A. B. Santani, S. L. Perlman et al., “A rapid, noninvasive immunoassay for frataxin: utility in assessment of Friedreich ataxia,” Molecular Genetics and Metabolism, vol. 101, no. 2-3, pp. 238–245, 2010.
[7]  M. V. Evans-Galea, L. A. Corben, J. Hasell, et al., “A novel deletion-insertion mutation identified in exon 3 of FXN in two siblings with a severe Friedreich ataxia phenotype,” Neurogenetics, vol. 12, no. 4, pp. 307–313, 2011.
[8]  C. H. Zühlke, A. Dalski, M. Habeck et al., “Extension of the mutation spectrum in Friedreich's ataxia: detection of an exon deletion and novel missense mutations,” European Journal of Human Genetics, vol. 12, no. 11, pp. 979–982, 2004.
[9]  M. Anheim, L. L. Mariani, P. Calvas, et al., “Exonic deletions of FXN and early-onset Friedreich ataxia,” Archives of Neurology, vol. 69, no. 7, pp. 912–916, 2012.
[10]  G. S. Chandok, M. P. Patel, S. M. Mirkin, and M. M. Krasilnikova, “Effects of Friedreich's ataxia GAA repeats on DNA replication in mammalian cells,” Nucleic Acids Research, vol. 40, no. 9, pp. 3964–3974, 2012.
[11]  N. Sakamoto, K. Ohshima, L. Montermini, M. Pandolfo, and R. D. Wells, “Sticky DNA, a self-associated complex formed at long GAA·TTC repeats in intron 1 of the frataxin gene, inhibits transcription,” Journal of Biological Chemistry, vol. 276, no. 29, pp. 27171–27177, 2001.
[12]  V. Ezzatizadeh, R. M. Pinto, C. Sandi, et al., “The mismatch repair system protects against intergenerational GAA repeat instability in a Friedreich ataxia mouse model,” Neurobiology of Disease, vol. 46, no. 1, pp. 165–171, 2012.
[13]  V. Campuzano, L. Montermini, Y. Lutz et al., “Frataxin is reduced in Friedreich ataxia patients and is associated with mitochondrial membranes,” Human Molecular Genetics, vol. 6, no. 11, pp. 1771–1780, 1997.
[14]  L. Pianese, M. Turano, M. S. Lo Casale et al., “Real time PCR quantification of frataxin mRNA in the peripheral blood leucocytes of Friedreich ataxia patients and carriers,” Journal of Neurology, Neurosurgery and Psychiatry, vol. 75, no. 7, pp. 1061–1063, 2004.
[15]  J. L. Bradley, J. C. Blake, S. Chamberlain, P. K. Thomas, J. M. Cooper, and A. H. V. Schapira, “Clinical, biochemical and molecular genetic correlations in Friedreich's ataxia,” Human Molecular Genetics, vol. 9, no. 2, pp. 275–282, 2000.
[16]  D. Waldvogel, P. van Gelderen, and M. Hallett, “Increased iron in the dentate nucleus of patients with Friedrich's ataxia,” Annals of Neurology, vol. 46, no. 1, pp. 123–125, 1999.
[17]  A. H. Koeppen, S. C. Michael, M. D. Knutson et al., “The dentate nucleus in Friedreich's ataxia: the role of iron-responsive proteins,” Acta Neuropathologica, vol. 114, no. 2, pp. 163–173, 2007.
[18]  F. Foury and O. Cazzalini, “Deletion of the yeast homologue of the human gene associated with Friedreich's ataxia elicits iron accumulation in mitochondria,” FEBS Letters, vol. 411, no. 2-3, pp. 373–377, 1997.
[19]  A. Wong, J. Yang, P. Cavadini et al., “The Friedreich's ataxia mutation confers cellular sensitivity to oxidant stress which is rescued by chelators of iron and calcium and inhibitors of apoptosis,” Human Molecular Genetics, vol. 8, no. 3, pp. 425–430, 1999.
[20]  J. B. Schulz, S. Boesch, K. Bürk et al., “Diagnosis and treatment of Friedreich ataxia: a European perspective,” Nature Reviews Neurology, vol. 5, no. 4, pp. 222–234, 2009.
[21]  E. Grabczyk, M. Mancuso, and M. C. Sammarco, “A persistent RNA·DNA hybrid formed by transcription of the Friedreich ataxia triplet repeat in live bacteria, and by T7 RNAP in vitro,” Nucleic Acids Research, vol. 35, no. 16, pp. 5351–5359, 2007.
[22]  R. D. Wells, “DNA triplexes and Friedreich ataxia,” FASEB Journal, vol. 22, no. 6, pp. 1625–1634, 2008.
[23]  A. Savellev, C. Everett, T. Sharpe, Z. Webster, and R. Festenstein, “DNA triplet repeats mediate heterochromatin-protein-1-sensitive variegated gene silencing,” Nature, vol. 422, no. 6934, pp. 909–913, 2003.
[24]  C. Beisel and R. Paro, “Silencing chromatin: comparing modes and mechanisms,” Nature Reviews Genetics, vol. 12, no. 2, pp. 123–135, 2011.
[25]  K. D. Robertson, “DNA methylation, methyltransferases, and cancer,” Oncogene, vol. 20, no. 24, pp. 3139–3155, 2001.
[26]  J. Goffin and E. Eisenhauer, “DNA methyltransferase inhibitors—state of the art,” Annals of Oncology, vol. 13, no. 11, pp. 1699–1716, 2002.
[27]  T. H. Bestor, “The DNA methyltransferases of mammals,” Human Molecular Genetics, vol. 9, no. 16, pp. 2395–2402, 2000.
[28]  S. Pradhan, A. Bacolla, R. D. Wells, and R. J. Roberts, “Recombinant human DNA (cytosine-5) methyltransferase. I. Expression, purification, and comparison of novo and maintenance methylation,” Journal of Biological Chemistry, vol. 274, no. 46, pp. 33002–33010, 1999.
[29]  C. Beard, E. Li, and R. Jaenisch, “Loss of methylation activates Xist in somatic but not in embryonic cells,” Genes and Development, vol. 9, no. 19, pp. 2325–2334, 1995.
[30]  M. Okano, D. W. Bell, D. A. Haber, and E. Li, “DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development,” Cell, vol. 99, no. 3, pp. 247–257, 1999.
[31]  D. Watanabe, I. Suetake, T. Tada, and S. Tajima, “Stage- and cell-specific expression of Dnmt3a and Dnmt3b during embryogenesis,” Mechanisms of Development, vol. 118, no. 1-2, pp. 187–190, 2002.
[32]  M. C. Lorincz, D. R. Dickerson, M. Schmitt, and M. Groudine, “Intragenic DNA methylation alters chromatin structure and elongation efficiency in mammalian cells,” Nature Structural and Molecular Biology, vol. 11, no. 11, pp. 1068–1075, 2004.
[33]  E. N. Gal-Yam, G. Egger, L. Iniguez et al., “Frequent switching of polycomb repressive marks and DNA hypermethylation in the PC3 prostate cancer cell line,” Proceedings of the National Academy of Sciences of the United States of America, vol. 105, no. 35, pp. 12979–12984, 2008.
[34]  Y. Schlesinger, R. Straussman, I. Keshet et al., “Polycomb-mediated methylation on Lys27 of histone H3 pre-marks genes for de novo methylation in cancer,” Nature Genetics, vol. 39, no. 2, pp. 232–236, 2007.
[35]  P. A. Jones, “Functions of DNA methylation: islands, start sites, gene bodies and beyond,” Nature Reviews Genetics, vol. 13, no. 7, pp. 484–492, 2012.
[36]  E. Greene, L. Mahishi, A. Entezam, D. Kumari, and K. Usdin, “Repeat-induced epigenetic changes in intron 1 of the frataxin gene and its consequences in Friedreich ataxia,” Nucleic Acids Research, vol. 35, no. 10, pp. 3383–3390, 2007.
[37]  H. Sugawara, K. Iwamoto, M. Bundo, et al., “Comprehensive DNA methylation analysis of human peripheral blood leukocytes and lymphoblastoid cell lines,” Epigenetics, vol. 6, no. 4, pp. 508–515, 2011.
[38]  S. Al-Mahdawi, R. M. Pinto, O. Ismail et al., “The Friedreich ataxia GAA repeat expansion mutation induces comparable epigenetic changes in human and transgenic mouse brain and heart tissues,” Human Molecular Genetics, vol. 17, no. 5, pp. 735–746, 2008.
[39]  M. V. Evans-Galea, N. Carrodus, S. M. Rowley, et al., “FXN methylation predicts expression and clinical outcome in Friedreich ataxia,” Annals of Neurology, vol. 71, no. 4, pp. 487–497, 2012.
[40]  I. Castaldo, M. Pinelli, A. Monticelli et al., “DNA methylation in intron 1 of the frataxin gene is related to GAA repeat length and age of onset in Friedreich ataxia patients,” Journal of Medical Genetics, vol. 45, no. 12, pp. 808–812, 2008.
[41]  A. Naumann, N. Hochstein, S. Weber, E. Fanning, and W. Doerfler, “A distinct DNA-methylation boundary in the 5′- upstream sequence of the FMR1 promoter binds nuclear proteins and is lost in fragile X syndrome,” American Journal of Human Genetics, vol. 85, no. 5, pp. 606–616, 2009.
[42]  A. López Castel, M. Nakamori, S. Tomé et al., “Expanded CTG repeat demarcates a boundary for abnormal CpG methylation in myotonic dystrophy patient tissues,” Human Molecular Genetics, vol. 20, no. 1, pp. 1–15, 2011.
[43]  V. Dion, Y. Lin, L. Hubert Jr., R. A. Waterland, and J. H. Wilson, “Dnmt1 deficiency promotes CAG repeat expansion in the mouse germline,” Human Molecular Genetics, vol. 17, no. 9, pp. 1306–1317, 2008.
[44]  R. T. Libby, K. A. Hagerman, V. V. Pineda et al., “CTCF cis-regulates trinucleotide repeat instability in an epigenetic manner: a novel basis for mutational hot spot determination,” PLoS Genetics, vol. 4, no. 11, Article ID e1000257, 2008.
[45]  T. Kouzarides, “Chromatin modifications and their function,” Cell, vol. 128, no. 4, pp. 693–705, 2007.
[46]  V. G. Allfrey, R. Faulkner, and A. E. Mirsky, “Acetylation and methylation of histones and their possible role in the regulation of RNA synthesis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 51, pp. 786–794, 1964.
[47]  M. Tan, H. Luo, S. Lee, et al., “Identification of 67 histone marks and histone lysine crotonylation as a new type of histone modification,” Cell, vol. 146, no. 6, pp. 1016–1028, 2011.
[48]  A. Razin, “CpG methylation, chromatin structure and gene silencing—a three-way connection,” EMBO Journal, vol. 17, no. 17, pp. 4905–4908, 1998.
[49]  G. Blander and L. Guarente, “The Sir2 family of protein deacetylases,” Annual Review of Biochemistry, vol. 73, pp. 417–435, 2004.
[50]  K. N. Bhalla, “Epigenetic and chromatin modifiers as targeted therapy of hematologic malignancies,” Journal of Clinical Oncology, vol. 23, no. 17, pp. 3971–3993, 2005.
[51]  P. A. Marks and M. Dokmanovic, “Histone deacetylase inhibitors: discovery and development as anticancer agents,” Expert Opinion on Investigational Drugs, vol. 14, no. 12, pp. 1497–1511, 2005.
[52]  K. B. Glaser, “HDAC inhibitors: clinical update and mechanism-based potential,” Biochemical Pharmacology, vol. 74, no. 5, pp. 659–671, 2007.
[53]  L. N. Pan, J. Lu, and B. Huang, “HDAC inhibitors: a potential new category of anti-tumor agents,” Cellular & Molecular Immunology, vol. 4, no. 5, pp. 337–343, 2007.
[54]  D. H. Cho, C. P. Thienes, S. E. Mahoney, E. Analau, G. N. Filippova, and S. J. Tapscott, “Antisense transcription and heterochromatin at the DM1 CTG repeats are constrained by CTCF,” Molecular Cell, vol. 20, no. 3, pp. 483–489, 2005.
[55]  B. Coffee, F. Zhang, S. Ceman, S. T. Warren, and D. Reines, “Histone modifications depict an aberrantly heterochromatinized FMR1 gene in fragile X syndrome,” American Journal of Human Genetics, vol. 71, no. 4, pp. 923–932, 2002.
[56]  D. Kumari and K. Usdin, “The distribution of repressive histone modifications on silenced FMR1 alleles provides clues to the mechanism of gene silencing in fragile X syndrome,” Human Molecular Genetics, vol. 19, no. 23, pp. 4634–4642, 2010.
[57]  L. E. Kernochan, M. L. Russo, N. S. Woodling et al., “The role of histone acetylation in SMN gene expression,” Human Molecular Genetics, vol. 14, no. 9, pp. 1171–1182, 2005.
[58]  N. Sakamoto, P. D. Chastain, P. Parniewski et al., “Sticky DNA: self-association properties of long GAA·TTC repeats in R·R·Y triplex structures from Friedreich's ataxia,” Molecular Cell, vol. 3, no. 4, pp. 465–475, 1999.
[59]  N. Sakamoto, J. E. Larson, R. R. Iyer, L. Montermini, M. Pandolfo, and R. D. Wells, “GGA·TCC-interrupted triplets in long GAA·TTC repeats inhibit the formation of triplex and sticky DNA structures, alleviate transcription inhibition, and reduce genetic instabilities,” Journal of Biological Chemistry, vol. 276, no. 29, pp. 27178–27187, 2001.
[60]  D. Herman, K. Jenssen, R. Burnett, E. Soragni, S. L. Perlman, and J. M. Gottesfeld, “Histone deacetylase inhibitors reverse gene silencing in Friedreich's ataxia,” Nature Chemical Biology, vol. 2, no. 10, pp. 551–558, 2006.
[61]  T. Punga and M. Bühler, “Long intronic GAA repeats causing Friedreich ataxia impede transcription elongation,” EMBO Molecular Medicine, vol. 2, no. 4, pp. 120–129, 2010.
[62]  M. Rai, E. Soragni, K. Jenssen et al., “HDAC inihibitors correct frataxin deficiency in a Friedreich ataxia mouse model,” PLoS One, vol. 3, no. 4, Article ID e1958, 2008.
[63]  I. De Biase, Y. K. Chutake, P. M. Rindler, and S. I. Bidichandani, “Epigenetic silencing in Friedreich ataxia is associated with depletion of CTCF (CCCTC-binding factor) and antisense transcription,” PLoS One, vol. 4, no. 11, Article ID e7914, 2009.
[64]  C. Martin and Y. Zhang, “The diverse functions of histone lysine methylation,” Nature Reviews Molecular Cell Biology, vol. 6, no. 11, pp. 838–849, 2005.
[65]  N. Kourmouli, P. Jeppesen, S. Mahadevhaiah et al., “Heterochromatin and tri-methylated lysine 20 of histone H4 in animals,” Journal of Cell Science, vol. 117, no. 12, pp. 2491–2501, 2004.
[66]  T. Jenuwein, “The epigenetic magic of histone lysine methylation: delivered on 6 July 2005 at the 30th FEBS Congress in Budapest, Hungary,” FEBS Journal, vol. 273, no. 14, pp. 3121–3135, 2006.
[67]  R. J. Sims III, K. Nishioka, and D. Reinberg, “Histone lysine methylation: a signature for chromatin function,” Trends in Genetics, vol. 19, no. 11, pp. 629–639, 2003.
[68]  E. Kim, M. Napierala, and S. Y. Dent, “Hyperexpansion of GAA repeats affects post-initiation steps of FXN transcription in Friedreich's ataxia,” Nucleic Acids Research, vol. 39, no. 19, pp. 8366–8377, 2011.
[69]  D. Kumari, R. E. Biacsi, and K. Usdin, “Repeat expansion affects both transcription initiation and elongation in Friedreich ataxia cells,” Journal of Biological Chemistry, vol. 286, no. 6, pp. 4209–4215, 2011.
[70]  F. Acquaviva, I. Castaldo, A. Filla et al., “Recombinant human erythropoietin increases frataxin protein expression without increasing mRNA expression,” Cerebellum, vol. 7, no. 3, pp. 360–365, 2008.
[71]  C. Sandi, R. M. Pinto, S. Al-Mahdawi et al., “Prolonged treatment with pimelic o-aminobenzamide HDAC inhibitors ameliorates the disease phenotype of a Friedreich ataxia mouse model,” Neurobiology of Disease, vol. 42, no. 3, pp. 496–505, 2011.
[72]  B. E. Bernstein, E. Birney, I. Dunham, et al., “An integrated encyclopedia of DNA elements in the human genome,” Nature, vol. 489, no. 7414, pp. 57–74, 2012.
[73]  J. Harrow, A. Frankish, J. M. Gonzalez, et al., “GENCODE: the reference human genome annotation for The ENCODE Project,” Genome Research, vol. 22, no. 9, pp. 1760–1774, 2012.
[74]  P. Qi and X. Du, “The long non-coding RNAs, a new cancer diagnostic and therapeutic gold mine,” Modern Pathology, vol. 26, no. 2, pp. 155–165, 2013.
[75]  C. P. Ponting and T. G. Belgard, “Transcribed dark matter: meaning or myth?” Human Molecular Genetics, vol. 19, no. R2, pp. R162–R168, 2010.
[76]  S. Djebali, C. A. Davis, A. Merkel, et al., “Landscape of transcription in human cells,” Nature, vol. 489, no. 7414, pp. 101–108, 2012.
[77]  G. Lavorgna, D. Dahary, B. Lehner, R. Sorek, C. M. Sanderson, and G. Casari, “In search of antisense,” Trends in Biochemical Sciences, vol. 29, no. 2, pp. 88–94, 2004.
[78]  A. E. Pasquinelli and G. Ruvkun, “Control of developmental timing by microRNAs and their targets,” Annual Review of Cell and Developmental Biology, vol. 18, pp. 495–513, 2002.
[79]  K. Delaval and R. Feil, “Epigenetic regulation of mammalian genomic imprinting,” Current Opinion in Genetics and Development, vol. 14, no. 2, pp. 188–195, 2004.
[80]  K. V. Morris, S. Santoso, A. M. Turner, C. Pastori, and P. G. Hawkins, “Bidirectional transcription directs both transcriptional gene activation and suppression in human cells,” PLoS Genetics, vol. 4, no. 11, Article ID e1000258, 2008.
[81]  S. H. Munroe and M. A. Lazar, “Inhibition of c-erbA mRNA splicing by a naturally occurring antisense RNA,” Journal of Biological Chemistry, vol. 266, no. 33, pp. 22083–22086, 1991.
[82]  D. W. Chung, D. D. Rudnicki, L. Yu, and L. Margolis, “A natural antisense transcript at the Huntington's disease repeat locus regulates HTT expression,” Human Molecular Genetics, vol. 20, no. 17, pp. 3467–3477, 2011.
[83]  A. M. Khalil, M. A. Faghihi, F. Modarresi, S. P. Brothers, and C. Wahlestedt, “A novel RNA transcript with antiapoptotic function is silenced in fragile X syndrome,” PLoS One, vol. 3, no. 1, Article ID e1486, 2008.
[84]  P. D. Ladd, L. E. Smith, N. A. Rabaia et al., “An antisense transcript spanning the CGG repeat region of FMR1 is upregulated in premutation carriers but silenced in full mutation individuals,” Human Molecular Genetics, vol. 16, no. 24, pp. 3174–3187, 2007.
[85]  B. L. Sopher, P. D. Ladd, V. V. Pineda et al., “CTCF regulates ataxin-7 expression through promotion of a convergently transcribed, antisense noncoding RNA,” Neuron, vol. 70, no. 6, pp. 1071–1084, 2011.
[86]  M. L. Moseley, T. Zu, Y. Ikeda et al., “Bidirectional expression of CUG and CAG expansion transcripts and intranuclear polyglutamine inclusions in spinocerebellar ataxia type 8,” Nature Genetics, vol. 38, no. 7, pp. 758–769, 2006.
[87]  Z. Yu, X. Teng, and N. M. Bonini, “Triplet repeat-derived siRNAs enhance RNA-mediated toxicity in a drosophila model for myotonic dystrophy,” PLoS Genetics, vol. 7, no. 3, Article ID e1001340, 2011.
[88]  J. Chen, M. Sun, W. J. Kent et al., “Over 20% of human transcripts might form sense-antisense pairs,” Nucleic Acids Research, vol. 32, no. 16, pp. 4812–4820, 2004.
[89]  X. J. Wang, T. Gaasterland, and N. H. Chua, “Genome-wide prediction and identification of cis-natural antisense transcripts in Arabidopsis thaliana,” Genome Biology, vol. 6, no. 4, article R30, 2005.
[90]  G. G. Carmichael, “Antisense starts making more sense,” Nature Biotechnology, vol. 21, no. 4, pp. 371–372, 2003.
[91]  V. V. Lobanenkov, R. H. Nicolas, V. V. Adler et al., “A novel sequence-specific DNA binding protein which interacts with three regularly spaced direct repeats of the CCCTC-motif in the 5′-flanking sequence of the chicken c-myc gene,” Oncogene, vol. 5, no. 12, pp. 1743–1753, 1990.
[92]  J. E. Phillips and V. G. Corces, “CTCF: master weaver of the genome,” Cell, vol. 137, no. 7, pp. 1194–1211, 2009.
[93]  S. Shukla, E. Kavak, M. Gregory, et al., “CTCF-promoted RNA polymerase II pausing links DNA methylation to splicing,” Nature, vol. 479, no. 7371, pp. 74–79, 2011.
[94]  N. Engel, J. L. Thorvaldsen, and M. S. Bartolomei, “CTCF binding sites promote transcription initiation and prevent DNA methylation on the maternal allele at the imprinted H19/Igf2 locus,” Human Molecular Genetics, vol. 15, no. 19, pp. 2945–2954, 2006.
[95]  G. N. Filippova, M. K. Cheng, J. M. Moore et al., “Boundaries between chromosomal domains of X inactivation and escape bind CTCF and lack CpG methylation during early development,” Developmental Cell, vol. 8, no. 1, pp. 31–42, 2005.
[96]  G. N. Filippova, C. P. Thienes, B. H. Penn et al., “CTCF-binding sites flank CTG/CAG repeats and form a methylation-sensitive insulator at the DM1 locus,” Nature Genetics, vol. 28, no. 4, pp. 335–343, 2001.
[97]  C. B. Yoo and P. A. Jones, “Epigenetic therapy of cancer: past, present and future,” Nature Reviews Drug Discovery, vol. 5, no. 1, pp. 37–50, 2006.
[98]  N. Jain, A. Rossi, and G. Garcia-Manero, “Epigenetic therapy of leukemia: an update,” International Journal of Biochemistry and Cell Biology, vol. 41, no. 1, pp. 72–80, 2009.
[99]  J. P. J. Issa, G. Garcia-Manero, F. J. Giles et al., “Phase 1 study of low-dose prolonged exposure schedules of the hypomethylating agent 5-aza-2′-deoxycytidine (decitabine) in hematopoietic malignancies,” Blood, vol. 103, no. 5, pp. 1635–1640, 2004.
[100]  H. I. Saba, “Decitabine in the treatment of myelodysplastic syndromes,” Therapeutics and Clinical Risk Management, vol. 3, no. 5, pp. 807–817, 2007.
[101]  H. I. Saba and P. W. Wijermans, “Decitabine in myelodysplastic syndromes,” Seminars in Hematology, vol. 42, no. 3, supplement 2, pp. S23–S31, 2005.
[102]  P. Chiurazzi, M. G. Pomponi, R. Willemsen, B. A. Oostra, and G. Neri, “In vitro reactivation of the FMR1 gene involved in fragile X syndrome,” Human Molecular Genetics, vol. 7, no. 1, pp. 109–113, 1998.
[103]  P. Chiurazzi, M. G. Pomponi, R. Pietrobono, C. E. Bakker, G. Neri, and B. A. Oostra, “Synergistic effect of histone hyperacetylation and DNA demethylation in the reactivation of the FMR1 gene,” Human Molecular Genetics, vol. 8, no. 12, pp. 2317–2323, 1999.
[104]  D. J. Stewart, R. C. Donehower, E. A. Eisenhauer et al., “A phase I pharmacokinetic and pharmacodynamic study of the DNA methyltransferase 1 inhibitor MG98 administered twice weekly,” Annals of Oncology, vol. 14, no. 5, pp. 766–774, 2003.
[105]  E. Winquist, J. Knox, J. P. Ayoub et al., “Phase II trial of DNA methyltransferase 1 inhibition with the antisense oligonucleotide MG98 in patients with metastatic renal carcinoma: a National Cancer Institute of Canada Clinical Trials Group investigational new drug study,” Investigational New Drugs, vol. 24, no. 2, pp. 159–167, 2006.
[106]  R. J. Amato, “Inhibition of DNA methylation by antisense oligonucleotide MG98 as cancer therapy,” Clinical Genitourinary Cancer, vol. 5, no. 7, pp. 422–426, 2007.
[107]  R. Plummer, L. Vidal, M. Griffin et al., “Phase I study of MG98, an oligonucleotide antisense inhibitor of human DNA methyltransferase 1, given as a 7-day infusion in patients with advanced solid tumors,” Clinical Cancer Research, vol. 15, no. 9, pp. 3177–3183, 2009.
[108]  K. Kato, N. K. Long, H. Makita et al., “Effects of green tea polyphenol on methylation status of RECK gene and cancer cell invasion in oral squamous cell carcinoma cells,” British Journal of Cancer, vol. 99, no. 4, pp. 647–654, 2008.
[109]  Q. P. Dou, “Molecular mechanisms of green tea polyphenols,” Nutrition and Cancer, vol. 61, no. 6, pp. 827–835, 2009.
[110]  Y. Yuasa, H. Nagasaki, Y. Akiyama et al., “DNA methylation status is inversely correlated with green tea intake and physical activity in gastric cancer patients,” International Journal of Cancer, vol. 124, no. 11, pp. 2677–2682, 2009.
[111]  M. Pandey, S. Shukla, and S. Gupta, “Promoter demethylation and chromatin remodeling by green tea polyphenols leads to re-expression of GSTP1 in human prostate cancer cells,” International Journal of Cancer, vol. 126, no. 11, pp. 2520–2533, 2010.
[112]  R. Butler and G. P. Bates, “Histone deacetylase inhibitors as therapeutics for polyglutamine disorders,” Nature Reviews Neuroscience, vol. 7, no. 10, pp. 784–796, 2006.
[113]  R. Festenstein, “Breaking the silence in Friedreich's ataxia,” Nature Chemical Biology, vol. 2, no. 10, pp. 512–513, 2006.
[114]  J. P. Sarsero, L. Li, H. Wardan, K. Sitte, R. Williamson, and P. A. Ioannou, “Upregulation of expression from the FRDA genomic locus for the therapy of Friedreich ataxia,” Journal of Gene Medicine, vol. 5, no. 1, pp. 72–81, 2003.
[115]  M. Rai, E. Soragni, C. J. Chou et al., “Two new pimelic diphenylamide HDAC inhibitors induce sustained frataxin upregulation in cells from Friedreich's ataxia patients and in a mouse model,” PloS One, vol. 5, no. 1, Article ID e8825, 2010.
[116]  C. J. Chou, D. Herman, and J. M. Gottesfeld, “Pimelic diphenylamide 106 is a slow, tight-binding inhibitor of class I histone deacetylases,” Journal of Biological Chemistry, vol. 283, no. 51, pp. 35402–35409, 2008.
[117]  E. Soragni, D. Herman, S. Y. R. Dent, J. M. Gottesfeld, R. D. Wells, and M. Napierala, “Long intronic GAA?TTC repeats induce epigenetic changes and reporter gene silencing in a molecular model of Friedreich ataxia,” Nucleic Acids Research, vol. 36, no. 19, pp. 6056–6065, 2008.
[118]  C. Xu, E. Soragni, C. J. Chou et al., “Chemical probes identify a role for histone deacetylase 3 in Friedreich's ataxia gene silencing,” Chemistry and Biology, vol. 16, no. 9, pp. 980–989, 2009.
[119]  S. Ghosh and M. B. Feany, “Comparison of pathways controlling toxicity in the eye and brain in Drosophila models of human neurodegenerative diseases,” Human Molecular Genetics, vol. 13, no. 18, pp. 2011–2018, 2004.
[120]  H. Ota, E. Tokunaga, K. Chang et al., “Sirt1 inhibitor, Sirtinol, induces senescence-like growth arrest with attenuated Ras-MAPK signaling in human cancer cells,” Oncogene, vol. 25, no. 2, pp. 176–185, 2006.
[121]  R. Biacsi, D. Kumari, and K. Usdin, “SIRT1 inhibition alleviates gene silencing in Fragile X mental retardation syndrome,” PLoS Genetics, vol. 4, no. 3, Article ID e1000017, 2008.
[122]  L. Garbes, M. Riessland, I. H?lker et al., “LBH589 induces up to 10-fold SMN protein levels by several independent mechanisms and is effective even in cells from SMA patients non-responsive to valproate,” Human Molecular Genetics, vol. 18, no. 19, pp. 3645–3658, 2009.
[123]  Y. B. Kim, K. H. Lee, K. Sugita, M. Yoshida, and S. Horinouchi, “Oxamflatin is a novel antitumor compound that inhibits mammalian histone deacetylase,” Oncogene, vol. 18, no. 15, pp. 2461–2470, 1999.
[124]  D. Cecconi, M. Donadelli, E. Dalla Pozza et al., “Synergistic effect of trichostatin A and 5-aza-2′-deoxycytidine on growth inhibition of pancreatic endocrine tumour cell lines: a proteomic study,” Proteomics, vol. 9, no. 7, pp. 1952–1966, 2009.
[125]  W. Luszczek, V. Cheriyath, T. M. Mekhail, and E. C. Borden, “Combinations of DNA methyltransferase and histone deacetylase inhibitors induce DNA damage in small cell lung cancer cells: correlation of resistance with IFN-stimulated gene expression,” Molecular Cancer Therapeutics, vol. 9, no. 8, pp. 2309–2321, 2010.
[126]  J. P. Cogswell, J. Ward, I. A. Taylor et al., “Identification of miRNA changes in Alzheimer's disease brain and CSF yields putative biomarkers and insights into disease pathways,” Journal of Alzheimer's Disease, vol. 14, no. 1, pp. 27–41, 2008.
[127]  S. T. Lee, K. Chu, W. S. Im, et al., “Altered microRNA regulation in Huntington's disease models,” Experimental Neurology, vol. 227, no. 1, pp. 172–179, 2011.
[128]  L. H. Mahishi, R. P. Hart, D. R. Lynch, and R. R. Ratan, “miR-886-3p levels are elevated in Friedreich Ataxia,” The Journal of Neuroscience, vol. 32, no. 27, pp. 9369–9373, 2012.
[129]  G. J. Hannon, “RNA interference,” Nature, vol. 418, no. 6894, pp. 244–251, 2002.
[130]  B. A. Janowski, J. Hu, and D. R. Corey, “Silencing gene expression by targeting chromosomal DNA with antigene peptide nucleic acids and duplex RNAs,” Nature Protocols, vol. 1, no. 1, pp. 436–443, 2006.
[131]  B. A. Janowski, K. E. Huffman, J. C. Schwartz et al., “Inhibiting gene expression at transcription start sites in chromosomal DNA with antigene RNAs,” Nature Chemical Biology, vol. 1, no. 4, pp. 216–222, 2005.
[132]  K. V. Morris, S. W. L. Chan, S. E. Jacobsen, and D. J. Looney, “Small interfering RNA-induced transcriptional gene silencing in human cells,” Science, vol. 305, no. 5688, pp. 1289–1292, 2004.
[133]  B. A. Janowski, S. T. Younger, D. B. Hardy, R. Ram, K. E. Huffman, and D. R. Corey, “Activating gene expression in mammalian cells with promoter-targeted duplex RNAs,” Nature Chemical Biology, vol. 3, no. 3, pp. 166–173, 2007.
[134]  J. K. Watts, D. Yu, K. Charisse et al., “Effect of chemical modifications on modulation of gene expression by duplex antigene RNAs that are complementary to non-coding transcripts at gene promoters,” Nucleic Acids Research, vol. 38, no. 15, pp. 5242–5259, 2010.
[135]  L. C. Li, S. T. Okino, H. Zhao et al., “Small dsRNAs induce transcriptional activation in human cells,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, no. 46, pp. 17337–17342, 2006.
[136]  S. Kriaucionis and N. Heintz, “The nuclear DNA base 5-hydroxymethylcytosine is present in purkinje neurons and the brain,” Science, vol. 324, no. 5929, pp. 929–930, 2009.
[137]  M. Tahiliani, K. P. Koh, Y. Shen et al., “Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1,” Science, vol. 324, no. 5929, pp. 930–935, 2009.
[138]  A. Martelli, M. Napierala, and H. Puccio, “Understanding the genetic and molecular pathogenesis of Friedreich’s ataxia through animal and cellular models,” Disease Models & Mechanisms, vol. 5, no. 2, pp. 165–176, 2012.
[139]  M. A. Pook, “DNA methylation and trinucleotide repeat expansion diseases,” in DNA Methylation—From Genomics to Technology, T. Tatarinova and O. Kerton, Eds., InTech, 2012.

Full-Text

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133