350 руб
Журнал «Вопросы биологической, медицинской и фармацевтической химии» №3 за 2014 г.
Статья в номере:
Эпигенетические механизмы регуляции экспрессии генов в химическом канцерогенезе
Авторы:
Н.А. Шалгинских - лаборант-исследователь, отдел химического канцерогенеза, ФГБУ «РОНЦ им. Н.Н.Блохина» РАМН (Москва). E-mail: nshalgin@mail.ru Н.Ю. Карпеченко - мл. науч. сотрудник, отдел химического канцерогенеза, ФГБУ «РОНЦ им. Н.Н.Блохина» РАМН (Москва). E-mail: nojadg@mail.ru А.М. Оглоблина - мл. науч. сотрудник, отдел химического канцерогенеза, ФГБУ «РОНЦ им. Н.Н.Блохина» РАМН (Москва). E-mail: globbi@mail.ru Е.А. Лесовая - к.б.н., науч. сотрудник, отдел химического канцерогенеза, ФГБУ «РОНЦ им. Н.Н.Блохина» РАМН (Москва). E-mail: lesovenok@yandex.ru К.И. Кирсанов - к.б.н., ст. науч. сотрудник, отдел химического канцерогенеза, ФГБУ «РОНЦ им. Н.Н.Блохина» РАМН (Москва). E-mail: mrkir@rambler.ru Д.С. Набережнов - аспирант, отдел химического канцерогенеза, ФГБУ «РОНЦ им. Н.Н.Блохина» РАМН (Москва). E-mail: n41d23s05@gmail.com Г.А. Белицкий - д.м.н., профессор, вед. науч. сотрудник, отдел химического канцерогенеза, ФГБУ «РОНЦ им. Н.Н.Блохина» РАМН (Москва). E-mail: belitsga@mail.ru М.Г. Якубовская - д.м.н., руководитель отдела химического канцерогенеза, ФГБУ «РОНЦ им. Н.Н.Блохина» РАМН (Москва). E-mail: mgyakubovskaya@mail.ru
Аннотация:
Рассмотрены исследования в области химического канцерогенеза, направленные на изучение молекулярных механизмов действия негенотоксических канцерогенов, промоторов канцерогенеза и антиканцерогенов. Показано, что данные по участию ксенобиотиков в процессах регуляции экспрессии генов делают актуальными вопросы тестирования соединений на эпигенетическую активность, первичной профилактики онкологических заболеваний, определения риска их развития после экспозиции к эпигенетическим канцерогенам.
Страницы: 46-64
Список источников

 

  1. Crick F. Central dogma of molecular biology // Nature. 1970. V. 227. № 5258. Р. 561-563.
  2. Knudson A.G. Two genetic hits (more or less) to cancer // Nat Rev Cancer. 2001. V. 1. № 2. Р. 157-62.
  3. Boveri T. Zur Frage der Entstehung maligner Tumoren. 1914.
  4. Boveri T. The Origin of Malignant Tumors. Translated by Marcella Boveri. Baltimore, The Williams & Wilkins Company. 1929.
  5. McCann J., Spingarn N.E., Kobori J., Ames B.N. Detection of carcinogens as mutagens: bacterial tester strains with R factor plasmids // Proc Natl Acad Sci U S A. 1975. V. 72. № 3. Р. 979-983.
  6. Арзамасцев Е.В., Абилев С.К., Белицкий Г.А. Прогноз канцерогенности фармакологических средств и вспомогательных веществ в краткосрочных тестах // Ведомости фармакологического комитета. 1999. № 1. Р. 18-31.
  7. Miller J.A. Carcinogenesis by chemicals: an overview--G. H. A. Clowes memorial lecture // Cancer Res. 1970. V. 30. № 3. Р. 559-576.
  8. Slaga T.J. Cellular and molecular mechanisms involved in multistage skin carcinogenesis // Carcinog Compr Surv. 1989. № 11. Р. 1-18.
  9. Wilson G.G., Murray N.E. Restriction and modification systems // Annu Rev Genet. 1991. № 25. Р. 585-627.
  10. Bird A.P., Wolffe A.P. Methylation-induced repression--belts, braces, and chromatin // Cell. 1999. V. 99. № 5. Р. 451-454.
  11. Suzuki M.M., Bird A. DNA methylation landscapes: provocative insights from epigenomics // Nat Rev Genet. 2008. V. 9. № 6. Р. 465-476.
  12. Bestor T.H., Bourc'his D. Transposon silencing and imprint establishment in mammalian germ cells // Cold Spring Harb Symp Quant Biol. 2004. V. 69. Р. 381-387.
  13. Razin A., Kantor B. DNA methylation in epigenetic control of gene expression // Prog Mol Subcell Biol. 2005. 38: 151-67.
  14. Bird A. DNA methylation patterns and epigenetic memory // Genes Dev. 2002. V. 16. № 1. Р. 6-21.
  15. Bird A.P. CpG-rich islands and the function of DNA methylation // Nature. 1986. V. 321. № 6067. Р. 209-213.
  16. Majumder S., Kutay H., Datta J., Summers D., Jacob S.T., Ghoshal K. Epigenetic regulation of metallothionein-i gene expression: differential regulation of methylated and unmethylated promoters by DNA methyltransferases and methyl CpG binding proteins // J Cell Biochem. 2006. V. 97. № 6. Р. 1300-1316.
  17. N.E. Fomchenko E.V.V. Biological aspects of DNA methylation (literature review // Problems Of Health And Ecology. 2004. V. 3. № 33. Р. 55-58.
  18. Baylin S.B. DNA methylation and gene silencing in cancer // Nat Clin Pract Oncol. 2005. 2 Suppl 1. S4-11.
  19. Baylin S.B. Reversal of gene silencing as a therapeutic target for cancer--roles for DNA methylation and its interdigitation with chromatin // Novartis Found Symp. 2004. V. 259. Р. 226-233; discussion Р. 234-237, 285-288.
  20. Fraga M.F., Esteller M. Epigenetics and aging: the targets and the marks // Trends Genet. 2007. V. 23. № 8. Р. 413-418.
  21. Klose R.J., Bird A.P. Genomic DNA methylation: the mark and its mediators // Trends Biochem Sci. 2006. V. 31. № 2. Р. 89-97.
  22. Watt F., Molloy P.L. Cytosine methylation prevents binding to DNA of a HeLa cell transcription factor required for optimal expression of the adenovirus major late promoter // Genes Dev. 1988. 2(9): 1136-43.
  23. Fatemi M., Wade P.A. MBD family proteins: reading the epigenetic code // J Cell Sci. 2006. V. 119(Pt. 15). Р. 3033-3037.
  24. Jones P.L., Veenstra G.J., Wade P.A., Vermaak D., Kass S.U., Landsberger N., Strouboulis J., Wolffe A.P. Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription // Nat Genet. 1998. V. 19. № 2. Р. 187-191.
  25. Nan X., Ng H.H., Johnson C.A., Laherty C.D., Turner B.M., Eisenman R.N., Bird A. Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex // Nature. 1998. V. 393. № 6683. Р. 386-389.
  26. Sarraf S.A., Stancheva I. Methyl-CpG binding protein MBD1 couples histone H3 methylation at lysine 9 by SETDB1 to DNA replication and chromatin assembly // Mol Cell. 2004. V. 15. № 4. Р. 595-605.
  27. Boyes J., Bird A. DNA methylation inhibits transcription indirectly via a methyl-CpG binding protein // Cell. 1991. V. 64. № 6. Р. 1123-1134.
  28. Hendrich B., Bird A. Identification and characterization of a family of mammalian methyl-CpG binding proteins // Mol Cell Biol. 1998. V. 18. № 11. Р. 6538-6547.
  29. Hsieh C.L. Stability of patch methylation and its impact in regions of transcriptional initiation and elongation. // Mol Cell Biol. 1997. V. 17. № 10. Р. 5897-5904.
  30. Lorincz M.C., Dickerson D.R., Schmitt M., Groudine M. Intragenic DNA methylation alters chromatin structure and elongation efficiency in mammalian cells // Nat. Struct. Mol. Biol. 2004. V. 11. № 11. Р. 1068-1075.
  31. Lewandowska J., Bartoszek A. DNA methylation in cancer development, diagnosis and therapy--multiple opportunities for genotoxic agents to act as methylome disruptors or remediators // Mutagenesis. 2011. V. 26. № 4. Р. 475-487.
  32. Greger V., Passarge E., Hopping W., Messmer E., Horsthemke B. Epigenetic changes may contribute to the formation and spontaneous regression of retinoblastoma // Hum Genet. 1989. 83(2): 155-8.
  33. Jones P.A., Baylin S.B. The fundamental role of epigenetic events in cancer // Nat. Rev. Genet. 2002. V. 3. № 6. Р. 415-428.
  34. Herman J.G., Jen J., Merlo A., Baylin S.B.Hypermethylation-associated inactivation indicates a tumor suppressor role for p15INK4B // Cancer Res. 1996. V. 56. № 4. Р. 722-727.
  35. Herman J.G., Merlo A., Mao L., Lapidus R.G., Issa J.P., Davidson N.E., Sidransky D., Baylin S.B. Inactivation of the CDKN2/p16/MTS1 gene is frequently associated with aberrant DNA methylation in all common human cancers // Cancer Res. 1995. V. 55. № 20. Р. 4525-4530.
  36. Esteller M., Silva J.M., Dominguez G., Bonilla F., Matias-Guiu X., Lerma E., Bussaglia E., Prat J., Harkes I.C., Repasky E.A., Gabrielson E., Schutte M., Baylin S.B., Herman J.G. Promoter hypermethylation and BRCA1 inactivation in sporadic breast and ovarian tumors // J. Natl. Cancer. Inst. 2000. V. 92. № 7. Р. 564-569.
  37. Herman J.G., Umar A., Polyak K., Graff J.R., Ahuja N., Issa J.P., Markowitz S., Willson J.K., Hamilton S.R., Kinzler K.W., Kane M.F., Kolodner R.D., Vogelstein B., Kunkel T.A., Baylin S.B. Incidence and functional consequences of hMLH1 promoter hypermethylation in colorectal carcinoma // Proc. Natl. Acad. Sci. USA. 1998. V. 95. № 12. Р. 6870-6875.
  38. Issa J.P., Ottaviano Y.L., Celano P., Hamilton S.R., Davidson N.E., Baylin S.B. Methylation of the oestrogen receptor CpG island links ageing and neoplasia in human colon // Nat Genet. 1994. V. 7. № 4. Р. 536-540.
  39. Hahnel R., Harvey J., Kay P. Hypermethylation of the myogenic gene Myf-3 in human breast carcinomas // Anticancer Res. 1996. V. 16. № 4A. Р. 2111-2115.
  40. Yoshiura K., Kanai Y., Ochiai A., Shimoyama Y., Sugimura T., Hirohashi S. Silencing of the E-cadherin invasion-suppressor gene by CpG methylation in human carcinomas // Proc. Natl. Acad. Sci. USA. 1995. V. 92. № 16. Р. 7416-7419.
  41. Zion M., Ben-Yehuda D., Avraham A., Cohen O., Wetzler M., Melloul D., Ben-Neriah Y. Progressive de novo DNA methylation at the bcr-abl locus in the course of chronic myelogenous leukemia // Proc. Natl. Acad. Sci. USA. 1994. V. 91. № 22. P. 10722-6.
  42. Esteller M. Relevance of DNA methylation in the management of cancer. // Lancet Oncol. 2003. V. 4. № 6. Р.351-358.
  43. Dimitrijevic B. Cancer epigenome: A review // Arch Oncol. 2005. V. 13. № 3-4. Р. 108-114.
  44. Smith S.S., Crocitto L. DNA methylation in eukaryotic chromosome stability revisited: DNA methyltransferase in the management of DNA conformation space // Mol. Carcinog. 1999. V. 26. № 1. Р. 1-9.
  45. Liang G., Chan M.F., Tomigahara Y., Tsai Y.C., Gonzales F.A., Li E., Laird P.W., Jones P.A. Cooperativity between DNA methyltransferases in the maintenance methylation of repetitive elements // Mol. Cell. Biol. 2002. V. 22. №2. Р. 480-491.
  46. Bestor T.H., Verdine G.L. DNA methyltransferases // Curr. Opin. Cell. Biol. 1994. V. 6. № 3. Р. 380-389.
  47. Xiao R., Ding Y., Lu Q.J., Li Y.P., Li Y.J., Yang X.J., Su Y.W., Liang Y.S., Zhang G.Y., Wen H.Q. Effects of 5-azaC on methylation pattern of the perforin promoter of the perforin gene in normal human T cells // Zhong Nan Da Xue Xue Bao Yi Xue Ban. 2006. V. 31. № 6. 843-847.
  48. Kriaucionis S., Heintz N. The nuclear DNA base 5-hydroxymethylcytosine is present in Purkinje neurons and the brain // Science. 2009. V. 324. № 5929. Р. 929-930.
  49. Stroud H., Feng S., Morey Kinney S., Pradhan S., Jacobsen S.E. 5-Hydroxymethylcytosine is associated with enhancers and gene bodies in human embryonic stem cells // Genome. Biol. V. 12. № 6. R54.
  50. Киселева Н.П. К.Ф.Л. Деметилирование ДНК и канцерогенез (обзор) // Биохимия. 2005. V. 70. № 7. С. 900-911.
  51. Лихтенштейн А.В. Киселева Н.П. Метилирование ДНК и канцерогенез // Биохимия. 2001. V. 66. № 3. С. 293 - 317.
  52. Залетаев Д.В. Н.М.В., Стрельников В.В., Бабенко О.В., Пальцева Е.М., Землякова В.В., Кузнецова, Кекеева Т.В., Михайленко Д.С., Манохина И.К. Эпигенетическая регуляция экспрессии генов в процессах канцерогенеза // Молекулярная медицина. 2008. № 4. Р. 46-51.
  53. Francis N.J., Kingston R.E., Woodcock C.L. Chromatin compaction by a polycomb group protein complex // Science. 2004. V. 306. № 5701. Р. 1574-1577.
  54. Suto R.K., Clarkson M.J., Tremethick D.J., Luger K. Crystal structure of a nucleosome core particle containing the variant histone H2A.Z // Nat Struct Biol. 2000. V. 7. № 12. Р. 1121-1124.
  55. Luger K., Mader A.W., Richmond R.K., Sargent D.F., Richmond T.J. Crystal structure of the nucleosome core particle at 2.8 A resolution. // Nature. 1997. V. 389. № 6648. Р. 251-260.
  56. Hayes J.J., Clark D.J., Wolffe A.P. Histone contributions to the structure of DNA in the nucleosome // Proc. Natl. Acad. Sci. USA. 1991. V. 88. № 15. Р. 6829-6833.
  57. Kouzarides T. Chromatin modifications and their function // Cell. 2007. V. 128. № 4. Р. 693-705.
  58. Barski A., Cuddapah S., Cui K., Roh T.Y., Schones D.E., Wang Z., Wei G., Chepelev I., Zhao K. High-resolution profiling of histone methylations in the human genome // Cell. 2007. V. 129. № 4. Р. 823-837.
  59. Marks P., Rifkind R.A., Richon V.M., Breslow R., Miller T., Kelly W.K. Histone deacetylases and cancer: causes and therapies // Nat. Rev. Cancer. 2001. V. 1. № 3. Р. 194-202.
  60. Turner B.M. Cellular memory and the histone code // Cell. 2002. V. 111. № 3. Р. 285-291.
  61. Jenuwein T., Allis C.D. Translating the histone code // Science. 2001. V. 293. № 5532. Р. 1074-1080.
  62. Wolffe A.P., Hayes J.J. Chromatin disruption and modification // Nucleic Acids Res. 1999. V. 27. № 3. Р. 711-720.
  63. Grunstein M. Histone acetylation in chromatin structure and transcription // Nature. 1997. V. 389. № 6649. Р. 349-352.
  64. Sterner D.E., Berger S.L. Acetylation of histones and transcription-related factors // Microbiol. Mol. Biol. Rev. 2000. V. 64. № 2. Р. 435-459.
  65. Lee K.K., Workman J.L. Histone acetyltransferase complexes: one size doesn't fit all. // Nat. Rev. Mol. Cell. Biol. 2007. V. 8. № 4. Р. 284-295.
  66. Fraga M.F., Ballestar E., Villar-Garea A., Boix-Chornet M., Espada J., Schotta G., Bonaldi T., Haydon C., Ropero S., Petrie K., Iyer N.G., Perez-Rosado A., Calvo E., Lopez J.A., Cano A., Calasanz M.J., Colomer D., Piris M.A., Ahn N., Imhof A., Caldas C., Jenuwein T., Esteller M. Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer // Nat Genet. 2005. V. 37. № 4. Р. 391-400.
  67. Shogren-Knaak M., Ishii H., Sun J.M., Pazin M.J., Davie J.R., Peterson C.L. Histone H4-K16 acetylation controls chromatin structure and protein interactions. // Science. 2006. V. 311. № 5762. Р. 844-847.
  68. de Ruijter A.J., van Gennip A.H., Caron H.N., Kemp S., van Kuilenburg A.B. Histone deacetylases (HDACs): characterization of the classical HDAC family // Biochem. J. 2003. V. 370 (Pt. 3). Р. 737-749.
  69. van der Heijden G.W., Dieker J.W., Derijck A.A., Muller S., Berden J.H., Braat D.D., van der Vlag J., de Boer P. Asymmetry in histone H3 variants and lysine methylation between paternal and maternal chromatin of the early mouse zygote // Mech. Dev. 2005. V. 122. № 9. Р. 1008-1022.
  70. Guenther M.G., Levine S.S., Boyer L.A., Jaenisch R., Young R.A. A chromatin landmark and transcription initiation at most promoters in human cells // Cell. 2007. V. 130. № 1. Р. 77-88.
  71. Bannister A.J., Zegerman P., Partridge J.F., Miska E.A., Thomas J.O., Allshire R.C., Kouzarides T. Selective recognition of methylated lysine 9 on histone H3 by the HP1 chromo domain // Nature. 2001. V. 410. № 6824. Р. 120-124.
  72. Vakoc C.R., Sachdeva M.M., Wang H., Blobel G.A. Profile of histone lysine methylation across transcribed mammalian chromatin // Mol. Cell. Biol. 2006. V. 26. № 24. Р. 9185-9195.
  73. Sims J.K., Houston S.I., Magazinnik T., Rice J.C. A trans-tail histone code defined by monomethylated H4 Lys-20 and H3 Lys-9 demarcates distinct regions of silent chromatin // J. Biol. Chem. 2006. V. 281. № 18. Р. 12760-12766.
  74. Yamane K., Toumazou C., Tsukada Y., Erdjument-Bromage H., Tempst P., Wong J., Zhang Y. JHDM2A, a JmjC-containing H3K9 demethylase, facilitates transcription activation by androgen receptor // Cell. 2006. V. 125. № 3. Р. 483-495.
  75. Metzger E., Schule R. The expanding world of histone lysine demethylases // Nat. Struct. Mol. Biol. 2007. V. 14. № 4. Р. 252-254.
  76. Nowak S.J., Corces V.G. Phosphorylation of histone H3 correlates with transcriptionally active loci // Genes Dev. 2000. V. 14. № 23. Р. 3003-3013.
  77. Wang H., Wang L., Erdjument-Bromage H., Vidal M., Tempst P., Jones R.S., Zhang Y. Role of histone H2A ubiquitination in Polycomb silencing // Nature. 2004. V. 431. № 7010. 873-878.
  78. Zhu B., Zheng Y., Pham A.D., Mandal S.S., Erdjument-Bromage H., Tempst P., Reinberg D. Monoubiquitination of human histone H2B: the factors involved and their roles in HOX gene regulation // Mol. Cell. 2005. V. 20. № 4. Р. 601-611.
  79. Nathan D., Ingvarsdottir K., Sterner D.E., Bylebyl G.R., Dokmanovic M., Dorsey J.A., Whelan K.A., Krsmanovic M., Lane W.S., Meluh P.B., Johnson E.S., Berger S.L. Histone sumoylation is a negative regulator in Saccharomyces cerevisiae and shows dynamic interplay with positive-acting histone modifications // Genes Dev. 2006. V. 20. № 8. Р. 966-976.
  80. Cuthbert G.L., Daujat S., Snowden A.W., Erdjument-Bromage H., Hagiwara T., Yamada M., Schneider R., Gregory P.D., Tempst P., Bannister A.J., Kouzarides T. Histone deimination antagonizes arginine methylation // Cell. 2004. V. 118. № 5. Р. 545-553.
  81. Ju B.G., Lunyak V.V., Perissi V., Garcia-Bassets I., Rose D.W., Glass C.K., Rosenfeld M.G. A topoisomerase IIbeta-mediated dsDNA break required for regulated transcription // Science. 2006. V. 312. № 5781. Р. 1798-1802.
  82. Chen Z., Zang J., Whetstine J., Hong X., Davrazou F., Kutateladze T.G., Simpson M., Mao Q., Pan C.H., Dai S., Hagman J., Hansen K., Shi Y., Zhang G. Structural insights into histone demethylation by JMJD2 family members // Cell. 2006. V. 125. № 4. Р. 691-702.
  83. Fire A., Albertson D., Harrison S.W., Moerman D.G. Production of antisense RNA leads to effective and specific inhibition of gene expression in C. elegans muscle // Development. 1991. V. 113. № 2. Р. 503-514.
  84. Fire A., Xu S., Montgomery M.K., Kostas S.A., Driver S.E., Mello C.C. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans // Nature. 1998. V. 391. № 6669. Р. 806-811.
  85. Tijsterman M., Ketting R.F., Plasterk R.H. The genetics of RNA silencing // Annu. Rev. Genet. 2002. V. 36. Р. 489-519.
  86. Ullu E., Tschudi C., Chakraborty T. RNA interference in protozoan parasites // Cell Microbiol. 2004. V. 6. №. 6. Р509-519.
  87. Waterhouse P.M., Wang M.B., Lough T. Gene silencing as an adaptive defence against viruses // Nature. 2001. V. 411. Р. 6839. Р. 834-842.
  88. Bartel D.P. MicroRNAs: genomics, biogenesis, mechanism, and function // Cell. 2004. V. 116. № 2. Р. 281-297.
  89. Meister G., Tuschl T. Mechanisms of gene silencing by double-stranded RNA // Nature. 2004. V. 431. № 7006. Р. 343-349.
  90. Ryan B.M., Robles A.I., Harris C.C. Genetic variation in microRNA networks: the implications for cancer research // Nat. Rev. Cancer. 2010. V. 10. № 6. Р. 389-402.
  91. Lee Y., Ahn C., Han J., Choi H., Kim J., Yim J., Lee J., Provost P., Radmark O., Kim S., Kim V.N. The nuclear RNase III Drosha initiates microRNA processing // Nature. 2003. V. 425. № 6956. Р. 415-419.
  92. Lee Y., Jeon K., Lee J.T., Kim S., Kim V.N. MicroRNA maturation: stepwise processing and subcellular localization // EMBO J. 2002. V. 21. № 17. Р. 4663-4670.
  93. Bohnsack M.T., Czaplinski K., Gorlich D. Exportin 5 is a RanGTP-dependent dsRNA-binding protein that mediates nuclear export of pre-miRNAs // RNA. 2004. V. 10. № 2. Р. 185-191.
  94. Lund E., Guttinger S., Calado A., Dahlberg J.E., Kutay U. Nuclear export of microRNA precursors // Science. 2004. V. 303. № 5654. 95-98.
  95. Yi R., Qin Y., Macara I.G., Cullen B.R. Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs // Genes Dev. 2003. V. 17. № 24. Р. 3011-3016.
  96. Bernstein E., Caudy A.A., Hammond S.M., Hannon G.J. Role for a bidentate ribonuclease in the initiation step of RNA interference. // Nature. 2001. V. 409. № 6818. Р. 363-366.
  97. Hutvagner G., McLachlan J., Pasquinelli A.E., Balint E., Tuschl T., Zamore P.D. A cellular function for the RNA-interference enzyme Dicer in the maturation of the let-7 small temporal RNA // Science. 2001. V. 293. № 5531. Р. 834-838.
  98. Lee Y.S., Nakahara K., Pham J.W., Kim K., He Z., Sontheimer E.J., Carthew R.W. Distinct roles for Drosophila Dicer-1 and Dicer-2 in the siRNA/miRNA silencing pathways // Cell. 2004. V. 117. № 1. Р. 69-81.
  99. Hammond S.M., Bernstein E., Beach D., Hannon G.J. An RNA-directed nuclease mediates post-transcriptional gene silencing in Drosophila cells // Nature. 2000. V. 404. № 6775. Р. 293-296.
  100. Griffiths-Jones S., Grocock R.J., van Dongen S., Bateman A., Enright A.J. miRBase: microRNA sequences, targets and gene nomenclature // Nucleic Acids Res. 2006. 34(Database issue): D140-4.
  101. O'Toole A.S., Miller S., Haines N., Zink M.C., Serra M.J. Comprehensive thermodynamic analysis of 3' double-nucleotide overhangs neighboring Watson-Crick terminal base pairs // Nucleic Acids Res. 2006. V. 34. № 11. Р. 3338-3344.
  102. Martinez J., Patkaniowska A., Urlaub H., Luhrmann R., Tuschl T. Single-stranded antisense siRNAs guide target RNA cleavage in RNAi // Cell. 2002. V. 110. № 5. Р. 563-574.
  103. Martinez J., Tuschl T. RISC is a 5' phosphomonoester-producing RNA endonuclease // Genes Dev. 2004. V. 18. № 9. Р. 975-80.
  104. Pillai R.S., Bhattacharyya S.N., Filipowicz W. Repression of protein synthesis by miRNAs: how many mechanisms - // Trends. Cell. Biol. 2007. V. 17. № 3. Р. 118-126.
  105. Boeger H., Bushnell D.A., Davis R., Griesenbeck J., Lorch Y., Strattan J.S., Westover K.D., Kornberg R.D. Structural basis of eukaryotic gene transcription. // FEBS Lett. 2005. V. 579. № 4. Р. 899-903.
  106. Morris K.V. siRNA-mediated transcriptional gene silencing: the potential mechanism and a possible role in the histone code // Cell. Mol. Life Sci. 2005. V. 62. № 24. Р. 3057-3066.
  107. Jeffery L., Nakielny S. Components of the DNA methylation system of chromatin control are RNA-binding proteins // J. Biol. Chem. 2004 V. 279. № 47. Р. 49479-49487.
  108. Kim D.H., Villeneuve L.M., Morris K.V., Rossi J.J. Argonaute-1 directs siRNA-mediated transcriptional gene silencing in human cells. // Nat Struct Mol Biol. 2006 V. 13. № 9. Р. 793-797.
  109. Weinberg M.S., Villeneuve L.M., Ehsani A., Amarzguioui M., Aagaard L., Chen Z.X., Riggs A.D., Rossi J.J., Morris K.V. The antisense strand of small interfering RNAs directs histone methylation and transcriptional gene silencing in human cells // RNA. 2006. V. 12. № 2. Р. 256-262.
  110. Lim L.P., Lau N.C., Garrett-Engele P., Grimson A., Schelter J.M., Castle J., Bartel D.P., Linsley P.S., Johnson J.M. Microarray analysis shows that some microRNAs downregulate large numbers of target mRNAs // Nature. 2005. V. 433. № 7027. Р. 769-773.
  111. Bommer G.T., Gerin I., Feng Y., Kaczorowski A.J., Kuick R., Love R.E., Zhai Y., Giordano T.J., Qin Z.S., Moore B.B., MacDougald O.A., Cho K.R., Fearon E.R. p53-mediated activation of miRNA34 candidate tumor-suppressor genes // Curr. Biol. 2007. V. 17. № 15. Р. 1298-1307.
  112. Chang T.C., Wentzel E.A., Kent O.A., Ramachandran K., Mullendore M., Lee K.H., Feldmann G., Yamakuchi M., Ferlito M., Lowenstein C.J., Arking D.E., Beer M.A., Maitra A., Mendell J.T. Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis // Mol. Cell. 2007. V. 26. № 5. Р. 745-752.
  113. Tazawa H., Tsuchiya N., Izumiya M., Nakagama H. Tumor-suppressive miR-34a induces senescence-like growth arrest through modulation of the E2F pathway in human colon cancer cells // Proc. Natl. Acad. Sci. USA. 2007. V. 104. № (39). Р. 15472-15477.
  114. Frank-Kamenetskii M.D., Mirkin S.M. Triplex DNA structures. // Annu Rev Biochem. 1995. V. 64. Р. 65-95.
  115. Saini N., Zhang Y., Usdin K., Lobachev K.S. When secondary comes first--the importance of non-canonical DNA structures // Biochimie. 2013. V. 95. 2): 117-123.
  116. Zhao J., Bacolla A., Wang G., Vasquez K.M. Non-B DNA structure-induced genetic instability and evolution. // Cell. Mol. LifeSci. 2010. 67(1): 43-62.
  117. Никитина Т.В. Н.С.А. Микросателлитные последовательности ДНК человека: мутационный процесс и эволюция // Генетика. 2004. V. 4. № 10. Р. 1301-1318.
  118. Verma A., Halder K., Halder R., Yadav V.K., Rawal P., Thakur R.K., Mohd F., Sharma A., Chowdhury S. Genome-wide computational and expression analyses reveal G-quadruplex DNA motifs as conserved cis-regulatory elements in human and related species // J. Med. Chem. 2008. V. 51. № 18. Р. 5641-5649.
  119. Huppert J.L. Structure, location and interactions of G-quadruplexes // FEBS J. 2010. V. 277. № 17. Р. 3452-3458.
  120. Ambrus A., Chen D., Dai J., Jones R.A., Yang D. Solution structure of the biologically relevant G-quadruplex element in the human c-MYC promoter. Implications for G-quadruplex stabilization // Biochemistry. 2005. V. 44. № 6. Р. 2048-2058.
  121. Phan A.T., Kuryavyi V., Gaw H.Y., Patel D.J. Small-molecule interaction with a five-guanine-tract G-quadruplex structure from the human MYC promoter. // Nat. Chem. Biol. 2005. V. 1. № 3. Р. 167-173.
  122. Phan A.T., Modi Y.S., Patel D.J. Propeller-type parallel-stranded G-quadruplexes in the human c-myc promoter // J. Am. Chem. Soc. 2004. V. 126. № 28. Р. 8710-8716.
  123. Wang X.D., Ou T.M., Lu Y.J., Li Z., Xu Z., Xi C., Tan J.H., Huang S.L., An L.K., Li D., Gu L.Q., Huang Z.S. Turning off transcription of the bcl-2 gene by stabilizing the bcl-2 promoter quadruplex with quindoline derivatives // J Med Chem. 2010. V. 53. № 11. Р. 4390-4398.
  124. Fernando H., Reszka A.P., Huppert J., Ladame S., Rankin S., Venkitaraman A.R., Neidle S., Balasubramanian S. A conserved quadruplex motif located in a transcription activation site of the human c-kit oncogene // Biochemistry. 2006. V. 45. № 25. Р. 7854-7860.
  125. Hsu S.T., Varnai P., Bugaut A., Reszka A.P., Neidle S., Balasubramanian S. A G-rich sequence within the c-kit oncogene promoter forms a parallel G-quadruplex having asymmetric G-tetrad dynamics // J Am Chem Soc. 2009. V. 131. № 37. 13399-13409.
  126. McLuckie K.I., Waller Z.A., Sanders D.A., Alves D., Rodriguez R., Dash J., McKenzie G.J., Venkitaraman A.R., Balasubramanian S. G-quadruplex-binding benzo[a]phenoxazines down-regulate c-KIT expression in human gastric carcinoma cells // J. Am. Chem. Soc. 2011. V. 133. № 8. Р. 2658-2663.
  127. Phan A.T., Kuryavyi V., Burge S., Neidle S., Patel D.J. Structure of an unprecedented G-quadruplex scaffold in the human c-kit promoter // J. Am. Chem. Soc. 2007. V. 129. № 14. Р. 4386-4392.
  128. Sun D., Liu W.J., Guo K., Rusche J.J., Ebbinghaus S., Gokhale V., Hurley L.H. The proximal promoter region of the human vascular endothelial growth factor gene has a G-quadruplex structure that can be targeted by G-quadruplex-interactive agents // Mol. Cancer. Ther. 2008. V. 7. № 4. Р. 880-889.
  129. Cogoi S., Xodo L.E. G-quadruplex formation within the promoter of the KRAS proto-oncogene and its effect on transcription // Nucleic Acids Res. 2006. V. 34. № 9. Р. 2536-2549.
  130. Membrino A., Cogoi S., Pedersen E.B., Xodo L.E. G4-DNA formation in the HRAS promoter and rational design of decoy oligonucleotides for cancer therapy // PLoS One. 2011. V. 6. № 9. e24421.
  131. Qin Y., Fortin J.S., Tye D., Gleason-Guzman M., Brooks T.A., Hurley L.H. Molecular cloning of the human platelet-derived growth factor receptor beta (PDGFR-beta) promoter and drug targeting of the G-quadruplex-forming region to repress PDGFR-beta expression // Biochemistry. 2010. V. 49. № 19. Р. 4208-4219.
  132. Zhang R., Lin Y., Zhang C.T. Greglist: a database listing potential G-quadruplex regulated genes // Nucleic Acids Res. 2008. 36(Database issue). D372-6.
  133. Yadav V.K., Abraham J.K., Mani P., Kulshrestha R., Chowdhury S. QuadBase: genome-wide database of G4 DNA--occurrence and conservation in human, chimpanzee, mouse and rat promoters and 146 microbes. // Nucleic Acids Res. 2008. 36(Database issue). D381-5.
  134. Kikin O., Zappala Z., D'Antonio L., Bagga P.S. GRSDB2 and GRS_UTRdb: databases of quadruplex forming G-rich sequences in pre-mRNAs and mRNAs // Nucleic Acids Res. 2008. 36(Database issue). D141-8.
  135. Kostadinov R., Malhotra N., Viotti M., Shine R., D'Antonio L., Bagga P. GRSDB: a database of quadruplex forming G-rich sequences in alternatively processed mammalian pre-mRNA sequences // Nucleic Acids Res. 2006. 34(Database issue). D119-24.
  136. Brooks T.A., Kendrick S., Hurley L. Making sense of G-quadruplex and i-motif functions in oncogene promoters // FEBS J. 2010. V. 277. № 17. Р. 3459-3469.
  137. Fernando H., Sewitz S., Darot J., Tavare S., Huppert J.L., Balasubramanian S. Genome-wide analysis of a G-quadruplex-specific single-chain antibody that regulates gene expression // Nucleic Acids Res. 2009. V. 37. № 20. Р. 6716-6722.
  138. Sarkies P., Reams C., Simpson L.J., Sale J.E. Epigenetic instability due to defective replication of structured DNA // Mol Cell. 2010. V. 40. № 5. Р. 703-713.
  139. Sarkies P., Murat P., Phillips L.G., Patel K.J., Balasubramanian S., Sale J.E. FANCJ coordinates two pathways that maintain epigenetic stability at G-quadruplex DNA // Nucleic Acids Res. 2012. V. 40. № 4. Р. 1485-1498.
  140. Halder R., Halder K., Sharma P., Garg G., Sengupta S., Chowdhury S. Guanine quadruplex DNA structure restricts methylation of CpG dinucleotides genome-wide // Mol Biosyst. 2010. V. 6. № 12. Р. 2439-2447.
  141. Ray B.K., Dhar S., Shakya A., Ray A. Z-DNA-forming silencer in the first exon regulates human ADAM-12 gene expression // Proc. Natl. Acad. Sci. USA. 2011. V. 108. № 1. Р. 103-108.
  142. Ray B.K., Dhar S., Henry C., Rich A., Ray A. Epigenetic regulation by Z-DNA silencer function controls cancer-associated ADAM-12 expression in breast cancer: cross-talk between MeCP2 and NF1 transcription factor family // Cancer Res. 2013. V. 73. № 2. Р. 736-744.
  143. Davis T.L., Firulli A.B., Kinniburgh A.J. Ribonucleoprotein and protein factors bind to an H-DNA-forming c-myc DNA element: possible regulators of the c-myc gene // Proc. Natl. Acad. Sci. USA. 1989. V. 86. № 24. Р. 9682-9686.
  144. Shiraga T., Smith D., Nuthall H.N., Hollingsworth M.A., Harris A. Identification of two novel elements involved in human MUCI gene expression in vivo // Mol. Med. 2002. V. 8. № 1. Р. 33-41.
  145. Walter K., Warnecke G., Bowater R., Deppert W., Kim E. Tumor suppressor p53 binds with high affinity to CTG.CAG trinucleotide repeats and induces topological alterations in mismatched duplexes // J. Biol. Chem. 2005. V. 280. № 52. Р. 42497-42507.
  146. Cisneros F.J., Branch S. Transplacental exposure to the DNA demethylating agent, 5-AZA-CdR, affects the sexual behavior of CD-1 male mice // Neurotoxicology. 2004. V. 25. № 3. Р. 411-417.
  147. Anway M.D., Cupp A.S., Uzumcu M., Skinner M.K. Epigenetic transgenerational actions of endocrine disruptors and male fertility // Science. 2005. V. 308. № 5727. Р. 1466-9.
  148. Wong B., Chen S., Kwon J.A., Rich A. Characterization of Z-DNA as a nucleosome-boundary element in yeast Saccharomyces cerevisiae // Proc. Natl. Acad. Sci. USA. 2007. V. 104. № 7. Р. 2229-2234.
  149. Bachman A.N., Phillips J.M., Goodman J.I. Phenobarbital induces progressive patterns of GC-rich and gene-specific altered DNA methylation in the liver of tumor-prone B6C3F1 mice // Toxicol Sci. 2006. V. 91. № 2. Р. 393-405.
  150. Pogribny I.P., Tryndyak V.P., Woods C.G., Witt S.E., Rusyn I.Epigenetic effects of the continuous exposure to peroxisome proliferator WY-14,643 in mouse liver are dependent upon peroxisome proliferator activated receptor alpha // Mutat Res. 2007. V. 625. № 1-2. Р. 62-71.
  151. Hu W., Feng Z., Tang M.S. Preferential carcinogen-DNA adduct formation at codons 12 and 14 in the human K-ras gene and their possible mechanisms // Biochemistry. 2003. V. 42. № 33. Р. 10012-10023.
  152. Yoon J.H., Smith L.E., Feng Z., Tang M., Lee C.S., Pfeifer G.P. Methylated CpG dinucleotides are the preferential targets for G-to-T transversion mutations induced by benzo[a]pyrene diol epoxide in mammalian cells: similarities with the p53 mutation spectrum in smoking-associated lung cancers // Cancer Res. 2001. V. 61. № 19. Р. 7110-7117.
  153. Matter B., Wang G., Jones R., Tretyakova N. Formation of diastereomeric benzo[a]pyrene diol epoxide-guanine adducts in p53 gene-derived DNA sequences // Chem. Res. Toxicol. 2004. V. 17. № 6. Р. 731-741.
  154. Guza R., Kotandeniya D., Murphy K., Dissanayake T., Lin C., Giambasu G.M., Lad R.R., Wojciechowski F., Amin S., Sturla S.J., Hudson R.H., York D.M., Jankowiak R., Jones R., Tretyakova N.Y. Influence of C-5 substituted cytosine and related nucleoside analogs on the formation of benzo[a]pyrene diol epoxide-dG adducts at CG base pairs of DNA // Nucleic Acids Res. 2011. V. 39. № 9. Р. 3988-4006.
  155. Zhao C.Q., Young M.R., Diwan B.A., Coogan T.P., Waalkes M.P. Association of arsenic-induced malignant transformation with DNA hypomethylation and aberrant gene expression // Proc. Natl. Acad. Sci. USA. 1997. V. 94. № 20. Р. 10907-10912.
  156. Zheng H., Shank R.C. Changes in methyl-sensitive restriction sites of liver DNA from hamsters chronically exposed to hydrazine sulfate // Carcinogenesis. 1996. V. 17. № 12. Р. 2711-2717.
  157. Liu W.B., Liu J.Y., Ao L., Zhou Z.Y., Zhou Y.H., Cui Z.H., Yang H., Cao J. Dynamic changes in DNA methylation during multistep rat lung carcinogenesis induced by 3-methylcholanthrene and diethylnitrosamine // Toxicol Lett. 2009. V. 189. № 1. Р. 5-13.
  158. Belinsky S.A., Klinge D.M., Liechty K.C., March T.H., Kang T., Gilliland F.D., Sotnic N., Adamova G., Rusinova G., Telnov V. Plutonium targets the p16 gene for inactivation by promoter hypermethylation in human lung adenocarcinoma // Carcinogenesis. 2004. V. 25. № 6. Р. 1063-1067.
  159. Salnikow K., Zhitkovich A. Genetic and epigenetic mechanisms in metal carcinogenesis and cocarcinogenesis: nickel, arsenic, and chromium // Chem. Res. Toxicol. 2008. V. 21. № 1. Р. 28-44.
  160. Esteller M. CpG island hypermethylation and tumor suppressor genes: a booming present, a brighter future // Oncogene. 2002. V. 21. № 35. Р. 5427-5440.
  161. Herman J.G., Baylin S.B. Gene silencing in cancer in association with promoter hypermethylation // N. Engl. J. Med. 2003. V. 349. № 21. Р. 2042-2054.
  162. Belinsky S.A. Silencing of genes by promoter hypermethylation: key event in rodent and human lung cancer // Carcinogenesis. 2005. V. 26. № 9. Р. 1481-1487.
  163. Smith L.E., Denissenko M.F., Bennett W.P., Li H., Amin S., Tang M., Pfeifer G.P. Targeting of lung cancer mutational hotspots by polycyclic aromatic hydrocarbons // J. Natl. Cancer. Inst. 2000. V. 92. № 10. Р. 803-811.
  164. Denissenko M.F., Chen J.X., Tang M.S., Pfeifer G.P. Cytosine methylation determines hot spots of DNA damage in the human P53 gene // Proc. Natl. Acad. Sci. USA. 1997. V. 94. № 8. Р. 3893-3898.
  165. Nakajima M., Yokoi T. MicroRNAs from biology to future pharmacotherapy: regulation of cytochrome P450s and nuclear receptors // Pharmacol. Ther. 2011. V. 131. № 3. Р. 330-337.
  166. Arita A., Shamy M.Y., Chervona Y., Clancy H.A., Sun H., Hall M.N., Qu Q., Gamble M.V., Costa M. The effect of exposure to carcinogenic metals on histone tail modifications and gene expression in human subjects. // J. Trace. Elem. Med. Biol. 2012. V. 26. № 2-3. Р. 174-178.
  167. Rajendrasozhan S., Yao H., Rahman I. Current perspectives on role of chromatin modifications and deacetylases in lung inflammation in COPD // COPD. 2009. V. 6. № 4. Р. 291-297.
  168. Chen D., Fang L., Li H., Tang M.S., Jin C. Cigarette smoke component acrolein modulates chromatin assembly by inhibiting histone acetylation // J. Biol. Chem. 2013. V. 288. № 30. Р. 21678-21687.
  169. Wang Z., Zhao Y., Smith E., Goodall G.J., Drew P.A., Brabletz T., Yang C. Reversal and prevention of arsenic-induced human bronchial epithelial cell malignant transformation by microRNA-200b // Toxicol Sci. 2011. V. 121. № 1. Р. 110-122.
  170. Zhao Y., Liu H., Li Y., Wu J., Greenlee A.R., Yang C., Jiang Y. The role of miR-506 in transformed 16HBE cells induced by anti-benzo[a]pyrene-trans-7,8-dihydrodiol-9,10-epoxide // Toxicol Lett. 2011. V. 205. № 3. Р. 320-326.
  171. Shah Y.M., Morimura K., Yang Q., Tanabe T., Takagi M., Gonzalez F.J. Peroxisome proliferator-activated receptor alpha regulates a microRNA-mediated signaling cascade responsible for hepatocellular proliferation // Mol. Cell. Biol. 2007. V. 27. № 12. Р. 4238-4247.
  172. Kalscheuer S., Zhang X., Zeng Y., Upadhyaya P. Differential expression of microRNAs in early-stage neoplastic transformation in the lungs of F344 rats chronically treated with the tobacco carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone // Carcinogenesis. 2008. V. 29. № 12. Р. 2394-2399.
  173. Thomas T.J., Messner R.P. A left-handed (Z) conformation of poly(dA-dC).poly(dG-dT) induced by polyamines // Nucleic. Acids. Res. 1986. V. 14. № 16. Р. 6721-6733.
  174. Das S., Kundu S., Suresh Kumar G. Quinacrine and 9-amino acridine inhibit B-Z and B-H(l) form DNA conformational transitions // DNA Cell. Biol. 2011. V. 30. № 7. Р. 525-535.
  175. Du Y., Zhou X. Targeting non-B-form DNA in living cells // Chem Rec. 2013. Р. 13. № 4. Р. 371-384.
  176. Di Antonio M., Rodriguez R., Balasubramanian S. Experimental approaches to identify cellular G-quadruplex structures and functions // Methods. 2012. V. 57. № 1. Р. 84-92.
  177. Arola A., Vilar R. Stabilisation of G-quadruplex DNA by small molecules // Curr Top Med Chem. 2008. V. 8. № 15. Р. 1405-1415.
  178. Halder R., Riou J.F., Teulade-Fichou M.P., Frickey T., Hartig J.S. Bisquinolinium compounds induce quadruplex-specific transcriptome changes in HeLa S3 cell lines // BMC Res. Notes. 2012. V. 5. Р. 138.
  179. Mikami-Terao Y., Akiyama M., Yuza Y., Yanagisawa T., Yamada O., Yamada H. Antitumor activity of G-quadruplex-interactive agent TMPyP4 in K562 leukemic cells // Cancer Lett. 2008. V. 261. № 2. Р. 226-234.
  180. Postel E.H., Mango S.E., Flint S.J. A nuclease-hypersensitive element of the human c-myc promoter interacts with a transcription initiation factor // Mol. Cell. Biol. 1989. V. 9. № 11. Р. 5123-5133.
  181. Brooks T.A., Hurley L.H. Targeting MYC Expression through G-Quadruplexes // Genes. Cancer. 2010. V. 1. № 6. Р. 641-649.
  182. Dexheimer T.S., Carey S.S., Zuohe S., Gokhale V.M., Hu X., Murata L.B., Maes E.M., Weichsel A., Sun D., Meuillet E.J., Montfort W.R., Hurley L.H. NM23-H2 may play an indirect role in transcriptional activation of c-myc gene expression but does not cleave the nuclease hypersensitive element III(1) // Mol. Cancer. Ther. 2009. V. 8. № 5. Р. 1363-1377.
  183. Siddiqui-Jain A., Grand C.L., Bearss D.J., Hurley L.H. Direct evidence for a G-quadruplex in a promoter region and its targeting with a small molecule to repress c-MYC transcription // Proc. Natl. Acad. Sci. USA. 2002. V. 99. № 18. Р. 11593-11598.
  184. Ou T.M., Lu Y.J., Zhang C., Huang Z.S., Wang X.D., Tan J.H., Chen Y., Ma D.L., Wong K.Y., Tang J.C., Chan A.S., Gu L.Q. Stabilization of G-quadruplex DNA and down-regulation of oncogene c-myc by quindoline derivatives // J. Med. Chem. 2007. V. 50. № 7. Р. 1465-1474.
  185. Wu P., Ma D.L., Leung C.H., Yan S.C., Zhu N., Abagyan R., Che C.M. Stabilization of G-quadruplex DNA with platinum(II) Schiff base complexes: luminescent probe and down-regulation of c-myc oncogene expression // Chemistry. 2009. V. 15. № 47. Р. 13008-13021.
  186. Maiti S., Chaudhury N.K., Chowdhury S. Hoechst 33258 binds to G-quadruplex in the promoter region of human c-myc // Biochem. Biophys. Res. Commun. 2003. V. 310. № 2. Р. 505-512.
  187. Ma D.L., Chan D.S., Fu W.C., He H.Z., Yang H., Yan S.C., Leung C.H. Discovery of a natural product-like c-myc G-quadruplex DNA groove-binder by molecular docking // PLoS One. 2012. V. 7. № 8. e43278.
  188. Cheng M.K., Modi C., Cookson J.C., Hutchinson I., Heald R.A., McCarroll A.J., Missailidis S., Tanious F., Wilson W.D., Mergny J.L., Laughton C.A., Stevens M.F. Antitumor polycyclic acridines. 20. Search for DNA quadruplex binding selectivity in a series of 8,13-dimethylquino[4,3,2-kl]acridinium salts: telomere-targeted agents // J. Med. Chem. 2008. V. 51. № 4. Р. 963-975.
  189. Cuenca F., Moore M.J., Johnson K., Guyen B., De Cian A., Neidle S. Design, synthesis and evaluation of 4,5-di-substituted acridone ligands with high G-quadruplex affinity and selectivity, together with low toxicity to normal cells // Bioorg. Med. Chem. Lett. 2009. V. 19. № 17. Р. 5109-5113.
  190. Dash J., Shirude P.S., Balasubramanian S. G-quadruplex recognition by bis-indole carboxamides // Chem. Commun. (Camb). 2008. V. 26. Р. 3055-3057.
  191. Dash J., Shirude P.S., Hsu S.T., Balasubramanian S. Diarylethynyl amides that recognize the parallel conformation of genomic promoter DNA G-quadruplexes. // J. Am. Chem. Soc. 2008. V. 130. № 47. Р. 15950-15956.
  192. De Cian A., Delemos E., Mergny J.L., Teulade-Fichou M.P., Monchaud D. Highly efficient G-quadruplex recognition by bisquinolinium compounds // J. Am. Chem. Soc. 2007. V. 129. № 7. Р. 1856-1857.
  193. Hounsou C., Guittat L., Monchaud D., Jourdan M., Saettel N., Mergny J.L., Teulade-Fichou M.P. G-quadruplex recognition by quinacridines: a SAR, NMR, and biological study // ChemMedChem. 2007. V. 2. № 5. Р. 655-666.
  194. Kim M.Y., Gleason-Guzman M., Izbicka E., Nishioka D., Hurley L.H. The different biological effects of telomestatin and TMPyP4 can be attributed to their selectivity for interaction with intramolecular or intermolecular G-quadruplex structures // Cancer Res. 2003. Р. 63. № 12. Р. 3247-3256.
  195. Kim M.Y., Vankayalapati H., Shin-Ya K., Wierzba K., Hurley L.H. Telomestatin, a potent telomerase inhibitor that interacts quite specifically with the human telomeric intramolecular g-quadruplex // J. Am. Chem. Soc. 2002. V. 124. № 10. Р. 2098-2099.
  196. Seenisamy J., Bashyam S., Gokhale V., Vankayalapati H., Sun D., Siddiqui-Jain A., Streiner N., Shin-Ya K., White E., Wilson W.D., Hurley L.H. Design and synthesis of an expanded porphyrin that has selectivity for the c-MYC G-quadruplex structure // J. Am. Chem. Soc. 2005. V. 127. № 9. Р. 2944-2959.
  197. Waller Z.A., Sewitz S.A., Hsu S.T., Balasubramanian S. A small molecule that disrupts G-quadruplex DNA structure and enhances gene expression // J. Am. Chem. Soc. 2009. V. 131. № 35. Р. 12628-12633.
  198. Li Q., Xiang J.F., Yang Q.F., Sun H.X., Guan A.J., Tang Y.L.G4LDB: a database for discovering and studying G-quadruplex ligands // Nucleic Acids Res. 2013. 41(Database issue). D1115-23.
  199. Alcaro S., Costa G., Distinto S., Moraca F., Ortuso F., Parrotta L., Artese A. The polymorphisms of DNA G-quadruplex investigated by docking experiments with telomestatin enantiomers. // Curr. Pharm. Des. 2012. V. 18. № 14. Р. 1873-1879.
  200. Cosconati S., Marinelli L., Trotta R., Virno A., Mayol L., Novellino E., Olson A.J., Randazzo A. Tandem application of virtual screening and NMR experiments in the discovery of brand new DNA quadruplex groove binders. // J. Am. Chem. Soc. 2009. V. 131. № 45. Р. 16336-16337.
  201. Haider S., Neidle S. Molecular modeling and simulation of G-quadruplexes and quadruplex-ligand complexes // Methods Mol Biol. 2010. V. 608. Р. 17-37.
  202. Redman J.E., Granadino-Roldan J.M., Schouten J.A., Ladame S., Reszka A.P., Neidle S., Balasubramanian S. Recognition and discrimination of DNA quadruplexes by acridine-peptide conjugates // Org Biomol Chem. 2009. V. 7. № 1. Р. 76-84.
  203. Herceg Z., Lambert M.P., van Veldhoven K., Demetriou C., Vineis P., Smith M.T., Straif K., Wild C.P. Towards incorporating epigenetic mechanisms into carcinogen identification and evaluation // Carcinogenesis. 2013.
  204. Pogribny I.P. R.I., Karpf A.R. . Epigenetic alterations in oncogenesis. Preface // Adv. Exp. Med. Biol. 2013. V. 754. v-vii.
  205. Martinez-Zamudio R., Ha H.C. Environmental epigenetics in metal exposure // Epigenetics. 2011. V. 6. № 7. Р. 820-827.
  206. Kundakovic M., Gudsnuk K., Franks B., Madrid J., Miller R.L., Perera F.P., Champagne F.A. Sex-specific epigenetic disruption and behavioral changes following low-dose in utero bisphenol A exposure // Proc. Natl. Acad. Sci. USA. 2013. V. 110. № 24. 9956-9961.
  207. Monchaud D., Granzhan A., Saettel N., Guedin A., Mergny J.L., Teulade-Fichou M.P. One ring to bind them all. Part I: the efficiency of the macrocyclic scaffold for g-quadruplex DNA recognition. // J. Nucleic. Acids. 2010; 2010.
  208. Prakash A., Kieken F., Marky L.A., Borgstahl G.E. Stabilization of a G-Quadruplex from Unfolding by Replication Protein A Using Potassium and the Porphyrin TMPyP4 // J. Nucleic. Acids. 2011; 2011: 529828.
  209. Zhu L.N., Wu B., Kong D.M. Specific recognition and stabilization of monomeric and multimeric G-quadruplexes by cationic porphyrin TMPipEOPP under molecular crowding conditions // Nucleic. Acids. Res. 2013. V. 41. № 7. Р. 4324-4335.
  210. Yu Q., Liu Y., Wang C., Sun D., Yang X., Liu J. Chiral ruthenium(II) polypyridyl complexes: stabilization of g-quadruplex DNA, inhibition of telomerase activity and cellular uptake // PLoS One. 2012. V. 7. № 12. e50902.