메뉴 건너뛰기




Volumn 4, Issue 5, 2012, Pages

Pharmacogene regulatory elements: From discovery to applications

Author keywords

CHiP Seq; Enhancers; Mirna; Next generation sequencing; Pharmacogenomics; Promoters; RNA Seq

Indexed keywords

ALCOHOL DEHYDROGENASE; ANTICOAGULANT AGENT; CARBOPLATIN; CISPLATIN; COMPLEMENTARY DNA; CYCLOPHOSPHAMIDE; CYTOCHROME P450 1A1; CYTOCHROME P450 1B1; CYTOCHROME P450 2B6; CYTOCHROME P450 2E1; DEOXYRIBONUCLEASE; DOXORUBICIN; ETOPOSIDE; FIRTECAN; GLUCURONOSYLTRANSFERASE 1A1; IRINOTECAN; ISOPROTEIN; MESSENGER RNA; METHOTREXATE; MICRORNA; RIFAMPICIN; STAT PROTEIN; STAT3 PROTEIN; TEMOZOLOMIDE; TRANSCRIPTION FACTOR AP 2; WARFARIN;

EID: 84861335436     PISSN: None     EISSN: 1756994X     Source Type: Journal    
DOI: 10.1186/gm344     Document Type: Review
Times cited : (21)

References (145)
  • 2
    • 51649084617 scopus 로고    scopus 로고
    • Regulatory polymorphism in vitamin K epoxide reductase complex subunit 1 (VKORC1) affects gene expression and warfarin dose requirement.
    • Wang D, Chen H, Momary KM, Cavallari LH, Johnson JA, Sadee W. Regulatory polymorphism in vitamin K epoxide reductase complex subunit 1 (VKORC1) affects gene expression and warfarin dose requirement. Blood 2008, 112:1013-1021.
    • (2008) Blood , vol.112 , pp. 1013-1021
    • Wang, D.1    Chen, H.2    Momary, K.M.3    Cavallari, L.H.4    Johnson, J.A.5    Sadee, W.6
  • 4
    • 83355168713 scopus 로고    scopus 로고
    • A miR-200b/200c/429-binding site polymorphism in the 3' untranslated region of the AP-2alpha gene is associated with cisplatin resistance.
    • Wu Y, Xiao Y, Ding X, Zhuo Y, Ren P, Zhou C, Zhou J. A miR-200b/200c/429-binding site polymorphism in the 3' untranslated region of the AP-2alpha gene is associated with cisplatin resistance. PLoS One 2011, 6:e29043.
    • (2011) PLoS One , vol.6
    • Wu, Y.1    Xiao, Y.2    Ding, X.3    Zhuo, Y.4    Ren, P.5    Zhou, C.6    Zhou, J.7
  • 6
    • 3142718711 scopus 로고    scopus 로고
    • Human epigenome project - up and running.
    • Bradbury J. Human epigenome project - up and running. PLoS Biol 2003, 1:E82.
    • (2003) PLoS Biol , vol.1
    • Bradbury, J.1
  • 16
    • 0043269205 scopus 로고    scopus 로고
    • The RNA polymerase II core promoter.
    • Smale ST, Kadonaga JT. The RNA polymerase II core promoter. Annu Rev Biochem 2003, 72:449-479.
    • (2003) Annu Rev Biochem , vol.72 , pp. 449-479
    • Smale, S.T.1    Kadonaga, J.T.2
  • 17
    • 0037108159 scopus 로고    scopus 로고
    • The RNA polymerase II core promoter: a key component in the regulation of gene expression.
    • Butler JE, Kadonaga JT. The RNA polymerase II core promoter: a key component in the regulation of gene expression. Genes Dev 2002, 16:2583-2592.
    • (2002) Genes Dev , vol.16 , pp. 2583-2592
    • Butler, J.E.1    Kadonaga, J.T.2
  • 18
    • 0037047146 scopus 로고    scopus 로고
    • Promoter-proximal tethering elements regulate enhancer-promoter specificity in the Drosophila Antennapedia complex.
    • Calhoun VC, Stathopoulos A, Levine M. Promoter-proximal tethering elements regulate enhancer-promoter specificity in the Drosophila Antennapedia complex. Proc Natl Acad Sci USA 2002, 99:9243-9247.
    • (2002) Proc Natl Acad Sci USA , vol.99 , pp. 9243-9247
    • Calhoun, V.C.1    Stathopoulos, A.2    Levine, M.3
  • 19
    • 0023216891 scopus 로고
    • CpG islands in vertebrate genomes.
    • Gardiner-Garden M, Frommer M. CpG islands in vertebrate genomes. J Mol Biol 1987, 196:261-282.
    • (1987) J Mol Biol , vol.196 , pp. 261-282
    • Gardiner-Garden, M.1    Frommer, M.2
  • 20
    • 79956330964 scopus 로고    scopus 로고
    • CpG islands and the regulation of transcription.
    • Deaton AM, Bird A. CpG islands and the regulation of transcription. Genes Dev 2011, 25:1010-1022.
    • (2011) Genes Dev , vol.25 , pp. 1010-1022
    • Deaton, A.M.1    Bird, A.2
  • 21
    • 67349255210 scopus 로고    scopus 로고
    • CpG islands - 'a rough guide'.
    • Illingworth RS, Bird AP. CpG islands - 'a rough guide'. FEBS Lett 2009, 583:1713-1720.
    • (2009) FEBS Lett , vol.583 , pp. 1713-1720
    • Illingworth, R.S.1    Bird, A.P.2
  • 28
    • 0032519431 scopus 로고    scopus 로고
    • Genetic predisposition to the metabolism of irinotecan (CPT-11). Role of uridine diphosphate glucuronosyltransferase isoform 1A1 in the glucuronidation of its active metabolite (SN-38) in human liver microsomes.
    • Iyer L, King CD, Whitington PF, Green MD, Roy SK, Tephly TR, Coffman BL, Ratain MJ. Genetic predisposition to the metabolism of irinotecan (CPT-11). Role of uridine diphosphate glucuronosyltransferase isoform 1A1 in the glucuronidation of its active metabolite (SN-38) in human liver microsomes. J Clin Invest 1998, 101:847-854.
    • (1998) J Clin Invest , vol.101 , pp. 847-854
    • Iyer, L.1    King, C.D.2    Whitington, P.F.3    Green, M.D.4    Roy, S.K.5    Tephly, T.R.6    Coffman, B.L.7    Ratain, M.J.8
  • 35
    • 0030783250 scopus 로고    scopus 로고
    • A regulatory element within a coding exon modulates keratin 18 gene expression in transgenic mice.
    • Neznanov N, Umezawa A, Oshima RG. A regulatory element within a coding exon modulates keratin 18 gene expression in transgenic mice. J Biol Chem 1997, 272:27549-27557.
    • (1997) J Biol Chem , vol.272 , pp. 27549-27557
    • Neznanov, N.1    Umezawa, A.2    Oshima, R.G.3
  • 40
    • 79955019510 scopus 로고    scopus 로고
    • Cis-regulatory mutations are a genetic cause of human limb malformations.
    • VanderMeer JE, Ahituv N. cis-regulatory mutations are a genetic cause of human limb malformations. Dev Dyn 2011, 240:920-930.
    • (2011) Dev Dyn , vol.240 , pp. 920-930
    • VanderMeer, J.E.1    Ahituv, N.2
  • 41
    • 11144339384 scopus 로고    scopus 로고
    • Long-range control of gene expression: emerging mechanisms and disruption in disease.
    • Kleinjan DA, van Heyningen V. Long-range control of gene expression: emerging mechanisms and disruption in disease. Am J Hum Genet 2005, 76:8-32.
    • (2005) Am J Hum Genet , vol.76 , pp. 8-32
    • Kleinjan, D.A.1    van Heyningen, V.2
  • 43
    • 0026496950 scopus 로고
    • The role of individual human cytochromes P450 in drug metabolism and clinical response.
    • Cholerton S, Daly AK, Idle JR. The role of individual human cytochromes P450 in drug metabolism and clinical response. Trends Pharmacol Sci 1992, 13:434-439.
    • (1992) Trends Pharmacol Sci , vol.13 , pp. 434-439
    • Cholerton, S.1    Daly, A.K.2    Idle, J.R.3
  • 44
    • 0037251812 scopus 로고    scopus 로고
    • Cytochrome P450 3A pharmacogenetics: the road that needs traveled.
    • Flockhart DA, Rae JM. Cytochrome P450 3A pharmacogenetics: the road that needs traveled. Pharmacogenomics J 2003, 3:3-5.
    • (2003) Pharmacogenomics J , vol.3 , pp. 3-5
    • Flockhart, D.A.1    Rae, J.M.2
  • 45
    • 0035197548 scopus 로고    scopus 로고
    • Identification of variants of CYP3A4 and characterization of their abilities to metabolize testosterone and chlorpyrifos.
    • Dai D, Tang J, Rose R, Hodgson E, Bienstock RJ, Mohrenweiser HW, Goldstein JA. Identification of variants of CYP3A4 and characterization of their abilities to metabolize testosterone and chlorpyrifos. J Pharmacol Exp Ther 2001, 299:825-831.
    • (2001) J Pharmacol Exp Ther , vol.299 , pp. 825-831
    • Dai, D.1    Tang, J.2    Rose, R.3    Hodgson, E.4    Bienstock, R.J.5    Mohrenweiser, H.W.6    Goldstein, J.A.7
  • 47
    • 0032702726 scopus 로고    scopus 로고
    • The orphan human pregnane × receptor mediates the transcriptional activation of CYP3A4 by rifampicin through a distal enhancer module.
    • Goodwin B, Hodgson E, Liddle C. The orphan human pregnane × receptor mediates the transcriptional activation of CYP3A4 by rifampicin through a distal enhancer module. Mol Pharmacol 1999, 56:1329-1339.
    • (1999) Mol Pharmacol , vol.56 , pp. 1329-1339
    • Goodwin, B.1    Hodgson, E.2    Liddle, C.3
  • 49
    • 77957242787 scopus 로고    scopus 로고
    • Role of epigenetic mechanisms in differential regulation of the dioxin-inducible human CYP1A1 and CYP1B1 genes.
    • Beedanagari SR, Taylor RT, Bui P, Wang F, Nickerson DW, Hankinson O. Role of epigenetic mechanisms in differential regulation of the dioxin-inducible human CYP1A1 and CYP1B1 genes. Mol Pharmacol 2010, 78:608-616.
    • (2010) Mol Pharmacol , vol.78 , pp. 608-616
    • Beedanagari, S.R.1    Taylor, R.T.2    Bui, P.3    Wang, F.4    Nickerson, D.W.5    Hankinson, O.6
  • 51
    • 0037515661 scopus 로고    scopus 로고
    • A novel distal enhancer module regulated by pregnane × receptor/constitutive androstane receptor is essential for the maximal induction of CYP2B6 gene expression.
    • Wang H, Faucette S, Sueyoshi T, Moore R, Ferguson S, Negishi M, LeCluyse EL. A novel distal enhancer module regulated by pregnane × receptor/constitutive androstane receptor is essential for the maximal induction of CYP2B6 gene expression. J Biol Chem 2003, 278:14146-14152.
    • (2003) J Biol Chem , vol.278 , pp. 14146-14152
    • Wang, H.1    Faucette, S.2    Sueyoshi, T.3    Moore, R.4    Ferguson, S.5    Negishi, M.6    LeCluyse, E.L.7
  • 52
    • 77953169448 scopus 로고    scopus 로고
    • Identification of a FOXA-dependent enhancer of human alcohol dehydrogenase 4 (ADH4).
    • Pochareddy S, Edenberg HJ. Identification of a FOXA-dependent enhancer of human alcohol dehydrogenase 4 (ADH4). Gene 2010, 460:1-7.
    • (2010) Gene , vol.460 , pp. 1-7
    • Pochareddy, S.1    Edenberg, H.J.2
  • 55
    • 0347361541 scopus 로고    scopus 로고
    • Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs.
    • Yi R, Qin Y, Macara IG, Cullen BR. Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs. Genes Dev 2003, 17:3011-3016.
    • (2003) Genes Dev , vol.17 , pp. 3011-3016
    • Yi, R.1    Qin, Y.2    Macara, I.G.3    Cullen, B.R.4
  • 56
    • 0035887005 scopus 로고    scopus 로고
    • Dicer functions in RNA interference and in synthesis of small RNA involved in developmental timing in C. elegans.
    • Ketting RF, Fischer SE, Bernstein E, Sijen T, Hannon GJ, Plasterk RH. Dicer functions in RNA interference and in synthesis of small RNA involved in developmental timing in C. elegans. Genes Dev 2001, 15:2654-2659.
    • (2001) Genes Dev , vol.15 , pp. 2654-2659
    • Ketting, R.F.1    Fischer, S.E.2    Bernstein, E.3    Sijen, T.4    Hannon, G.J.5    Plasterk, R.H.6
  • 57
    • 36849048778 scopus 로고    scopus 로고
    • Dual role for argonautes in microRNA processing and posttranscriptional regulation of microRNA expression.
    • Diederichs S, Haber DA. Dual role for argonautes in microRNA processing and posttranscriptional regulation of microRNA expression. Cell 2007, 131:1097-1108.
    • (2007) Cell , vol.131 , pp. 1097-1108
    • Diederichs, S.1    Haber, D.A.2
  • 61
    • 77954757866 scopus 로고    scopus 로고
    • Short interfering RNA-mediated knockdown of drosha and pasha in undifferentiated Meloidogyne incognita eggs leads to irregular growth and embryonic lethality.
    • Dalzell JJ, Warnock ND, Stevenson MA, Mousley A, Fleming CC, Maule AG. Short interfering RNA-mediated knockdown of drosha and pasha in undifferentiated Meloidogyne incognita eggs leads to irregular growth and embryonic lethality. Int J Parasitol 2010, 40:1303-1310.
    • (2010) Int J Parasitol , vol.40 , pp. 1303-1310
    • Dalzell, J.J.1    Warnock, N.D.2    Stevenson, M.A.3    Mousley, A.4    Fleming, C.C.5    Maule, A.G.6
  • 64
    • 77952840905 scopus 로고    scopus 로고
    • MicroRNAs - targeting and target prediction.
    • Saito T, Saetrom P. MicroRNAs - targeting and target prediction. Nat Biotechnol 2010, 27:243-249.
    • (2010) Nat Biotechnol , vol.27 , pp. 243-249
    • Saito, T.1    Saetrom, P.2
  • 65
    • 84873516641 scopus 로고    scopus 로고
    • MiRBase: the microRNA database
    • miRBase: the microRNA database. , http://www.mirbase.org/
  • 67
    • 80053591724 scopus 로고    scopus 로고
    • MicroRNAs as post-transcriptional machines and their interplay with cellular networks.
    • Janga SC, Vallabhaneni S. MicroRNAs as post-transcriptional machines and their interplay with cellular networks. Adv 2011, 722:59-74.
    • (2011) Adv , vol.722 , pp. 59-74
    • Janga, S.C.1    Vallabhaneni, S.2
  • 68
    • 4644309196 scopus 로고    scopus 로고
    • The functions of animal microRNAs.
    • Ambros V. The functions of animal microRNAs. Nature 2004, 431:350-355.
    • (2004) Nature , vol.431 , pp. 350-355
    • Ambros, V.1
  • 69
    • 33749487425 scopus 로고    scopus 로고
    • MicroRNA regulates the expression of human cytochrome P450 1B1.
    • Tsuchiya Y, Nakajima M, Takagi S, Taniya T, Yokoi T. MicroRNA regulates the expression of human cytochrome P450 1B1. Cancer Res 2006, 66:9090-9098.
    • (2006) Cancer Res , vol.66 , pp. 9090-9098
    • Tsuchiya, Y.1    Nakajima, M.2    Takagi, S.3    Taniya, T.4    Yokoi, T.5
  • 70
    • 70349268327 scopus 로고    scopus 로고
    • MicroRNAs regulate CYP3A4 expression via direct and indirect targeting.
    • Pan YZ, Gao W, Yu AM. MicroRNAs regulate CYP3A4 expression via direct and indirect targeting. Drug Metab Dispos 2009, 37:2112-2117.
    • (2009) Drug Metab Dispos , vol.37 , pp. 2112-2117
    • Pan, Y.Z.1    Gao, W.2    Yu, A.M.3
  • 71
    • 49149114863 scopus 로고    scopus 로고
    • Role of MicroRNA miR-27a and miR-451 in the regulation of MDR1/P-glycoprotein expression in human cancer cells.
    • Zhu H, Wu H, Liu X, Evans BR, Medina DJ, Liu CG, Yang JM. Role of MicroRNA miR-27a and miR-451 in the regulation of MDR1/P-glycoprotein expression in human cancer cells. Biochem Pharmacol 2008, 76:582-588.
    • (2008) Biochem Pharmacol , vol.76 , pp. 582-588
    • Zhu, H.1    Wu, H.2    Liu, X.3    Evans, B.R.4    Medina, D.J.5    Liu, C.G.6    Yang, J.M.7
  • 72
    • 84867996343 scopus 로고    scopus 로고
    • Down-regulation of microRNA-200c is associated with drug resistance in human breast cancer.
    • doi: 10.1007/s12032-011-0117-4
    • Chen J, Tian W, Cai H, He H, Deng Y. Down-regulation of microRNA-200c is associated with drug resistance in human breast cancer. Med Oncol 2011, doi: 10.1007/s12032-011-0117-4.
    • (2011) Med Oncol
    • Chen, J.1    Tian, W.2    Cai, H.3    He, H.4    Deng, Y.5
  • 73
    • 34548059198 scopus 로고    scopus 로고
    • A miR-24 microRNA binding-site polymorphism in dihydrofolate reductase gene leads to methotrexate resistance.
    • Mishra PJ, Humeniuk R, Longo-Sorbello GS, Banerjee D, Bertino JR. A miR-24 microRNA binding-site polymorphism in dihydrofolate reductase gene leads to methotrexate resistance. Proc Natl Acad Sci USA 2007, 104:13513-13518.
    • (2007) Proc Natl Acad Sci USA , vol.104 , pp. 13513-13518
    • Mishra, P.J.1    Humeniuk, R.2    Longo-Sorbello, G.S.3    Banerjee, D.4    Bertino, J.R.5
  • 75
    • 80053247020 scopus 로고    scopus 로고
    • GSTP1 DNA methylation and expression status is indicative of 5-aza-2'-deoxycytidine efficacy in human prostate cancer cells.
    • Chiam K, Centenera MM, Butler LM, Tilley WD, Bianco-Miotto T. GSTP1 DNA methylation and expression status is indicative of 5-aza-2'-deoxycytidine efficacy in human prostate cancer cells. PLoS One 2011, 6:e25634.
    • (2011) PLoS One , vol.6
    • Chiam, K.1    Centenera, M.M.2    Butler, L.M.3    Tilley, W.D.4    Bianco-Miotto, T.5
  • 77
    • 84859256220 scopus 로고    scopus 로고
    • Targeting genetic and epigenetic alterations in the treatment of serous ovarian cancer.
    • Weberpals JI, Koti M, Squire JA. Targeting genetic and epigenetic alterations in the treatment of serous ovarian cancer. Cancer 2011, 204:525-535.
    • (2011) Cancer , vol.204 , pp. 525-535
    • Weberpals, J.I.1    Koti, M.2    Squire, J.A.3
  • 80
    • 27144500218 scopus 로고    scopus 로고
    • Reduced representation bisulfite sequencing for comparative high-resolution DNA methylation analysis.
    • Meissner A, Gnirke A, Bell GW, Ramsahoye B, Lander ES, Jaenisch R. Reduced representation bisulfite sequencing for comparative high-resolution DNA methylation analysis. Nucleic Acids Res 2005, 33:5868-5877.
    • (2005) Nucleic Acids Res , vol.33 , pp. 5868-5877
    • Meissner, A.1    Gnirke, A.2    Bell, G.W.3    Ramsahoye, B.4    Lander, E.S.5    Jaenisch, R.6
  • 82
    • 73249152285 scopus 로고    scopus 로고
    • Immunoprecipitation of methylated DNA.
    • Sorensen AL, Collas P. Immunoprecipitation of methylated DNA. Methods Mol Biol 2009, 567:249-262.
    • (2009) Methods Mol Biol , vol.567 , pp. 249-262
    • Sorensen, A.L.1    Collas, P.2
  • 83
    • 77449127799 scopus 로고    scopus 로고
    • MBD-isolated genome sequencing provides a high-throughput and comprehensive survey of DNA methylation in the human genome.
    • Serre D, Lee BH, Ting AH. MBD-isolated genome sequencing provides a high-throughput and comprehensive survey of DNA methylation in the human genome. Nucleic Acids Res 2010, 38:391-399.
    • (2010) Nucleic Acids Res , vol.38 , pp. 391-399
    • Serre, D.1    Lee, B.H.2    Ting, A.H.3
  • 86
    • 34250159524 scopus 로고    scopus 로고
    • Genome-wide mapping of in vivo protein-DNA interactions.
    • Johnson DS, Mortazavi A, Myers RM, Wold B. Genome-wide mapping of in vivo protein-DNA interactions. Science 2007, 316:1497-1502.
    • (2007) Science , vol.316 , pp. 1497-1502
    • Johnson, D.S.1    Mortazavi, A.2    Myers, R.M.3    Wold, B.4
  • 95
    • 82455188015 scopus 로고    scopus 로고
    • Oestrogen receptor-co-factor-chromatin specificity in the transcriptional regulation of breast cancer.
    • Zwart W, Theodorou V, Kok M, Canisius S, Linn S, Carroll JS. Oestrogen receptor-co-factor-chromatin specificity in the transcriptional regulation of breast cancer. EMBO J 2011, 30:4764-4776.
    • (2011) EMBO J , vol.30 , pp. 4764-4776
    • Zwart, W.1    Theodorou, V.2    Kok, M.3    Canisius, S.4    Linn, S.5    Carroll, J.S.6
  • 97
    • 77950605769 scopus 로고    scopus 로고
    • Genome-wide tissue-specific farnesoid × receptor binding in mouse liver and intestine.
    • Thomas AM, Hart SN, Kong B, Fang J, Zhong XB, Guo GL. Genome-wide tissue-specific farnesoid × receptor binding in mouse liver and intestine. Hepatology 2010, 51:1410-1419.
    • (2010) Hepatology , vol.51 , pp. 1410-1419
    • Thomas, A.M.1    Hart, S.N.2    Kong, B.3    Fang, J.4    Zhong, X.B.5    Guo, G.L.6
  • 99
    • 56749098074 scopus 로고    scopus 로고
    • Deep surveying of alternative splicing complexity in the human transcriptome by high-throughput sequencing.
    • Pan Q, Shai O, Lee LJ, Frey BJ, Blencowe BJ. Deep surveying of alternative splicing complexity in the human transcriptome by high-throughput sequencing. Nat Genet 2008, 40:1413-1415.
    • (2008) Nat Genet , vol.40 , pp. 1413-1415
    • Pan, Q.1    Shai, O.2    Lee, L.J.3    Frey, B.J.4    Blencowe, B.J.5
  • 101
    • 79251644774 scopus 로고    scopus 로고
    • Whole transcriptome sequencing reveals gene expression and splicing differences in brain regions affected by Alzheimer's disease.
    • Twine NA, Janitz K, Wilkins MR, Janitz M. Whole transcriptome sequencing reveals gene expression and splicing differences in brain regions affected by Alzheimer's disease. PLoS One 2011, 6:e16266.
    • (2011) PLoS One , vol.6
    • Twine, N.A.1    Janitz, K.2    Wilkins, M.R.3    Janitz, M.4
  • 106
    • 54049083902 scopus 로고    scopus 로고
    • Genetic variants associated with carboplatin-induced cytotoxicity in cell lines derived from Africans.
    • Huang RS, Duan S, Kistner EO, Hartford CM, Dolan ME. Genetic variants associated with carboplatin-induced cytotoxicity in cell lines derived from Africans. Mol Cancer Ther 2008, 7:3038-3046.
    • (2008) Mol Cancer Ther , vol.7 , pp. 3038-3046
    • Huang, R.S.1    Duan, S.2    Kistner, E.O.3    Hartford, C.M.4    Dolan, M.E.5
  • 108
    • 85017176879 scopus 로고    scopus 로고
    • PharmGKB: The Pharmacogenomics Knowledgebase
    • PharmGKB: The Pharmacogenomics Knowledgebase. , http://www.pharmgkb.org
  • 109
    • 78149332037 scopus 로고    scopus 로고
    • Complexity of the microRNA repertoire revealed by next-generation sequencing.
    • Lee LW, Zhang S, Etheridge A, Ma L, Martin D, Galas D, Wang K. Complexity of the microRNA repertoire revealed by next-generation sequencing. RNA 2010, 16:2170-2180.
    • (2010) RNA , vol.16 , pp. 2170-2180
    • Lee, L.W.1    Zhang, S.2    Etheridge, A.3    Ma, L.4    Martin, D.5    Galas, D.6    Wang, K.7
  • 111
    • 79951849971 scopus 로고    scopus 로고
    • Integrative analysis of next generation sequencing for small non-coding RNAs and transcriptional regulation in myelodysplastic syndromes.
    • Beck D, Ayers S, Wen J, Brandl MB, Pham TD, Webb P, Chang CC, Zhou X. Integrative analysis of next generation sequencing for small non-coding RNAs and transcriptional regulation in myelodysplastic syndromes. BMC Med Genomics 2011, 4:19.
    • (2011) BMC Med Genomics , vol.4 , pp. 19
    • Beck, D.1    Ayers, S.2    Wen, J.3    Brandl, M.B.4    Pham, T.D.5    Webb, P.6    Chang, C.C.7    Zhou, X.8
  • 113
    • 78649802141 scopus 로고    scopus 로고
    • Characterization of the small RNA transcriptomes of androgen dependent and independent prostate cancer cell line by deep sequencing.
    • Xu G, Wu J, Zhou L, Chen B, Sun Z, Zhao F, Tao Z. Characterization of the small RNA transcriptomes of androgen dependent and independent prostate cancer cell line by deep sequencing. PLoS One 2010, 5:e15519.
    • (2010) PLoS One , vol.5
    • Xu, G.1    Wu, J.2    Zhou, L.3    Chen, B.4    Sun, Z.5    Zhao, F.6    Tao, Z.7
  • 116
    • 80053619518 scopus 로고    scopus 로고
    • Application of next-generation sequencing technology to profile the circulating microRNAs in the serum of preeclampsia versus normal pregnant women.
    • Yang Q, Lu J, Wang S, Li H, Ge Q, Lu Z. Application of next-generation sequencing technology to profile the circulating microRNAs in the serum of preeclampsia versus normal pregnant women. Clin Chim Acta 2011, 412:2167-2173.
    • (2011) Clin Chim Acta , vol.412 , pp. 2167-2173
    • Yang, Q.1    Lu, J.2    Wang, S.3    Li, H.4    Ge, Q.5    Lu, Z.6
  • 117
    • 0033815579 scopus 로고    scopus 로고
    • Co-repressors 2000.
    • Burke LJ, Baniahmad A. Co-repressors 2000. FASEB J 2000, 14:1876-1888.
    • (2000) FASEB J , vol.14 , pp. 1876-1888
    • Burke, L.J.1    Baniahmad, A.2
  • 118
    • 0032055491 scopus 로고    scopus 로고
    • Transcriptional control and the role of silencers in transcriptional regulation in eukaryotes.
    • Ogbourne S, Antalis TM. Transcriptional control and the role of silencers in transcriptional regulation in eukaryotes. Biochem J 1998, 331:1-14.
    • (1998) Biochem J , vol.331 , pp. 1-14
    • Ogbourne, S.1    Antalis, T.M.2
  • 119
    • 53149107845 scopus 로고    scopus 로고
    • Chromatin insulators: regulatory mechanisms and epigenetic inheritance.
    • Bushey AM, Dorman ER, Corces VG. Chromatin insulators: regulatory mechanisms and epigenetic inheritance. Mol Cell 2008, 32:1-9.
    • (2008) Mol Cell , vol.32 , pp. 1-9
    • Bushey, A.M.1    Dorman, E.R.2    Corces, V.G.3
  • 120
    • 33747453473 scopus 로고    scopus 로고
    • Insulators: exploiting transcriptional and epigenetic mechanisms.
    • Gaszner M, Felsenfeld G. Insulators: exploiting transcriptional and epigenetic mechanisms. Nat Rev Genet 2006, 7:703-713.
    • (2006) Nat Rev Genet , vol.7 , pp. 703-713
    • Gaszner, M.1    Felsenfeld, G.2
  • 122
    • 34250369627 scopus 로고    scopus 로고
    • FAIRE (formaldehyde-assisted isolation of regulatory elements) isolates active regulatory elements from human chromatin.
    • Giresi PG, Kim J, McDaniell RM, Iyer VR, Lieb JD. FAIRE (formaldehyde-assisted isolation of regulatory elements) isolates active regulatory elements from human chromatin. Genome Res 2007, 17:877-885.
    • (2007) Genome Res , vol.17 , pp. 877-885
    • Giresi, P.G.1    Kim, J.2    McDaniell, R.M.3    Iyer, V.R.4    Lieb, J.D.5
  • 124
    • 70249088327 scopus 로고    scopus 로고
    • Genomic views of distant-acting enhancers.
    • Visel A, Rubin EM, Pennacchio LA. Genomic views of distant-acting enhancers. Nature 2009, 461:199-205.
    • (2009) Nature , vol.461 , pp. 199-205
    • Visel, A.1    Rubin, E.M.2    Pennacchio, L.A.3
  • 126
    • 73249137492 scopus 로고    scopus 로고
    • Determining spatial chromatin organization of large genomic regions using 5C technology.
    • van Berkum NL, Dekker J. Determining spatial chromatin organization of large genomic regions using 5C technology. Methods Mol Biol 2009, 567:189-213.
    • (2009) Methods Mol Biol , vol.567 , pp. 189-213
    • van Berkum, N.L.1    Dekker, J.2
  • 130
    • 77954199175 scopus 로고    scopus 로고
    • A dynamic Bayesian network for identifying protein-binding footprints from single molecule-based sequencing data.
    • Chen X, Hoffman MM, Bilmes JA, Hesselberth JR, Noble WS. A dynamic Bayesian network for identifying protein-binding footprints from single molecule-based sequencing data. Bioinformatics 2010, 26:i334-342.
    • (2010) Bioinformatics , vol.26
    • Chen, X.1    Hoffman, M.M.2    Bilmes, J.A.3    Hesselberth, J.R.4    Noble, W.S.5
  • 131
    • 70449711243 scopus 로고    scopus 로고
    • Computation for ChIP-seq and RNA-seq studies.
    • Pepke S, Wold B, Mortazavi A. Computation for ChIP-seq and RNA-seq studies. Nat Methods 2009, 6:S22-32.
    • (2009) Nat Methods , vol.6
    • Pepke, S.1    Wold, B.2    Mortazavi, A.3
  • 132
    • 79952095113 scopus 로고    scopus 로고
    • Statistical issues in the analysis of ChIP-Seq and RNA-Seq data.
    • Ghosh D, Qin ZS. Statistical issues in the analysis of ChIP-Seq and RNA-Seq data. Genes 2010, 1:317-334.
    • (2010) Genes , vol.1 , pp. 317-334
    • Ghosh, D.1    Qin, Z.S.2
  • 133
    • 83255164884 scopus 로고    scopus 로고
    • Comprehensive genome-wide protein-DNA interactions detected at single-nucleotide resolution.
    • Rhee PJ, Pugh BF. Comprehensive genome-wide protein-DNA interactions detected at single-nucleotide resolution. Cell 2011, 147:1408-1419.
    • (2011) Cell , vol.147 , pp. 1408-1419
    • Rhee, P.J.1    Pugh, B.F.2
  • 134
    • 71849118573 scopus 로고    scopus 로고
    • High-resolution analysis of DNA regulatory elements by synthetic saturation mutagenesis.
    • Patwardhan RP, Lee C, Litvin O, Young DL, Pe'er D, Shendure J. High-resolution analysis of DNA regulatory elements by synthetic saturation mutagenesis. Nat Biotechnol 2009, 27:1173-1175.
    • (2009) Nat Biotechnol , vol.27 , pp. 1173-1175
    • Patwardhan, R.P.1    Lee, C.2    Litvin, O.3    Young, D.L.4    Pe'er, D.5    Shendure, J.6
  • 140
    • 34248663609 scopus 로고    scopus 로고
    • U.S. Food and Drug Administration
    • U.S. Food and Drug Administration. , http://www.fda.gov/Drugs/ScienceResearch/ResearchAreas/Pharmacogenetics
  • 141
  • 142
    • 79951809825 scopus 로고    scopus 로고
    • CPIC: Clinical Pharmacogenetics Implementation Consortium of the Pharmacogenomics Research Network.
    • Relling MV, Klein TE. CPIC: Clinical Pharmacogenetics Implementation Consortium of the Pharmacogenomics Research Network. Clin Pharmacol Ther 2011, 89:464-467.
    • (2011) Clin Pharmacol Ther , vol.89 , pp. 464-467
    • Relling, M.V.1    Klein, T.E.2


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.