메뉴 건너뛰기




Volumn 61, Issue 6, 2015, Pages 69-84

Targeting post-translational modifications of histones for cancer therapy

Author keywords

Acetylation; Cancer; Histone; Methylation; Phosphorylation; Post translational modification

Indexed keywords

HISTONE; HISTONE DEACETYLASE INHIBITOR; PHOSPHOTRANSFERASE INHIBITOR; HISTONE DEACETYLASE;

EID: 84962157960     PISSN: 01455680     EISSN: 1165158X     Source Type: Journal    
DOI: 10.14715/cmb/2015.61.6.10     Document Type: Article
Times cited : (20)

References (239)
  • 1
    • 33847047461 scopus 로고    scopus 로고
    • Epigenetics: a landscape takes shape
    • Goldberg, A. D., Allis, C. D., Bernstein, E. Epigenetics: a landscape takes shape. Cell, 2007, 128: 635-638, doi:10.1016/j.cell.2007.02.006
    • (2007) Cell , vol.128 , pp. 635-638
    • Goldberg, A.D.1    Allis, C.D.2    Bernstein, E.3
  • 2
    • 84891071460 scopus 로고    scopus 로고
    • Histone deacetylases: a saga of perturbed acetylation homeostasis in cancer
    • Parbin, S., Kar, S., Shilpi, A., Sengupta, D., Deb, M., Rath, S. K., et al. Histone deacetylases: a saga of perturbed acetylation homeostasis in cancer. J Histochem Cytochem, 2014, 62: 11-33, doi:10.1369/0022155413506582
    • (2014) J Histochem Cytochem , vol.62 , pp. 11-33
    • Parbin, S.1    Kar, S.2    Shilpi, A.3    Sengupta, D.4    Deb, M.5    Rath, S.K.6
  • 4
    • 79953799195 scopus 로고    scopus 로고
    • Sirtuin-3 (SIRT3), a novel potential therapeutic target for oral cancer
    • Alhazzazi, T. Y., Kamarajan, P., Joo, N., Huang, J. Y., Verdin, E., D'Silva, N. J., et al. Sirtuin-3 (SIRT3), a novel potential therapeutic target for oral cancer. Cancer, 2011, 117: 1670-1678, doi:10.1002/cncr.25676
    • (2011) Cancer , vol.117 , pp. 1670-1678
    • Alhazzazi, T.Y.1    Kamarajan, P.2    Joo, N.3    Huang, J.Y.4    Verdin, E.5    D'Silva, N.J.6
  • 5
    • 47849083984 scopus 로고    scopus 로고
    • Epigenetic interplay between histone modifications and DNA methylation in gene silencing
    • Vaissiere, T., Sawan, C., Herceg, Z. Epigenetic interplay between histone modifications and DNA methylation in gene silencing. Mutat Res, 2008, 659: 40-48, doi:10.1016/j.mrrev.2008.02.004
    • (2008) Mutat Res , vol.659 , pp. 40-48
    • Vaissiere, T.1    Sawan, C.2    Herceg, Z.3
  • 6
    • 0030798245 scopus 로고    scopus 로고
    • Histone acetylation in chromatin structure and transcription
    • Grunstein, M. Histone acetylation in chromatin structure and transcription. Nature, 1997, 389: 349-352, doi:10.1038/38664
    • (1997) Nature , vol.389 , pp. 349-352
    • Grunstein, M.1
  • 7
    • 32444434989 scopus 로고    scopus 로고
    • Histone H4-K16 acetylation controls chromatin structure and protein interactions
    • 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, 311: 844-847, doi:10.1126/science.1124000
    • (2006) Science , vol.311 , pp. 844-847
    • Shogren-Knaak, M.1    Ishii, H.2    Sun, J.M.3    Pazin, M.J.4    Davie, J.R.5    Peterson, C.L.6
  • 8
    • 68949212379 scopus 로고    scopus 로고
    • Lysine acetylation targets protein complexes and co-regulates major cellular functions
    • Choudhary, C., Kumar, C., Gnad, F., Nielsen, M. L., Rehman, M., Walther, T. C., et al. Lysine acetylation targets protein complexes and co-regulates major cellular functions. Science, 2009, 325: 834-840, doi:10.1126/science.1175371
    • (2009) Science , vol.325 , pp. 834-840
    • Choudhary, C.1    Kumar, C.2    Gnad, F.3    Nielsen, M.L.4    Rehman, M.5    Walther, T.C.6
  • 9
    • 34547697009 scopus 로고    scopus 로고
    • Metabolism, cytoskeleton and cellular signalling in the grip of protein Nepsilon - and O-acetylation
    • Yang, X. J., Gregoire, S. Metabolism, cytoskeleton and cellular signalling in the grip of protein Nepsilon - and O-acetylation. EMBO Rep, 2007, 8: 556-562, doi:10.1038/sj.embor.7400977
    • (2007) EMBO Rep , vol.8 , pp. 556-562
    • Yang, X.J.1    Gregoire, S.2
  • 10
    • 77649101042 scopus 로고    scopus 로고
    • Histone deacetylase inhibition and the regulation of cell growth with particular reference to liver pathobiology
    • Joanna, F., van Grunsven, L. A., Mathieu, V., Sarah, S., Sarah, D., Karin, V., et al. Histone deacetylase inhibition and the regulation of cell growth with particular reference to liver pathobiology. J Cell Mol Med, 2009, 13: 2990-3005, doi:10.1111/j.1582-4934.2009.00831.x
    • (2009) J Cell Mol Med , vol.13 , pp. 2990-3005
    • Joanna, F.1    van Grunsven, L.A.2    Mathieu, V.3    Sarah, S.4    Sarah, D.5    Karin, V.6
  • 11
    • 0842285672 scopus 로고    scopus 로고
    • ETO protein of t(8;21) AML is a corepressor for Bcl-6 B-cell lymphoma oncoprotein
    • Chevallier, N., Corcoran, C. M., Lennon, C., Hyjek, E., Chadburn, A., Bardwell, V. J., et al. ETO protein of t(8;21) AML is a corepressor for Bcl-6 B-cell lymphoma oncoprotein. Blood, 2004, 103: 1454-1463, doi:10.1182/blood-2003-06-2081
    • (2004) Blood , vol.103 , pp. 1454-1463
    • Chevallier, N.1    Corcoran, C.M.2    Lennon, C.3    Hyjek, E.4    Chadburn, A.5    Bardwell, V.J.6
  • 12
    • 17144378591 scopus 로고    scopus 로고
    • Inhibition of histone deacetylase 2 increases apoptosis and p21Cip1/WAF1 expression, independent of histone deacetylase 1
    • Huang, B. H., Laban, M., Leung, C. H., Lee, L., Lee, C. K., Salto-Tellez, M., et al. Inhibition of histone deacetylase 2 increases apoptosis and p21Cip1/WAF1 expression, independent of histone deacetylase 1. Cell Death Differ, 2005, 12: 395-404, doi:10.1038/sj.cdd.4401567
    • (2005) Cell Death Differ , vol.12 , pp. 395-404
    • Huang, B.H.1    Laban, M.2    Leung, C.H.3    Lee, L.4    Lee, C.K.5    Salto-Tellez, M.6
  • 13
    • 21044447215 scopus 로고    scopus 로고
    • Increased expression of histone deacetylase 2 is found in human gastric cancer
    • Song, J., Noh, J. H., Lee, J. H., Eun, J. W., Ahn, Y. M., Kim, S. Y., et al. Increased expression of histone deacetylase 2 is found in human gastric cancer. APMIS, 2005, 113: 264-268, doi:10.1111/j.1600-0463.2005.apm_04.x
    • (2005) APMIS , vol.113 , pp. 264-268
    • Song, J.1    Noh, J.H.2    Lee, J.H.3    Eun, J.W.4    Ahn, Y.M.5    Kim, S.Y.6
  • 14
    • 0037383786 scopus 로고    scopus 로고
    • The tumor suppressor p53 and histone deacetylase 1 are antagonistic regulators of the cyclin-dependent kinase inhibitor p21/WAF1/CIP1 gene
    • Lagger, G., Doetzlhofer, A., Schuettengruber, B., Haidweger, E., Simboeck, E., Tischler, J., et al. The tumor suppressor p53 and histone deacetylase 1 are antagonistic regulators of the cyclin-dependent kinase inhibitor p21/WAF1/CIP1 gene. Mol Cell Biol, 2003, 23: 2669-2679.
    • (2003) Mol Cell Biol , vol.23 , pp. 2669-2679
    • Lagger, G.1    Doetzlhofer, A.2    Schuettengruber, B.3    Haidweger, E.4    Simboeck, E.5    Tischler, J.6
  • 15
    • 76749159433 scopus 로고    scopus 로고
    • The cyclin-dependent kinase inhibitor p21 is a crucial target for histone deacetylase 1 as a regulator of cellular proliferation
    • Zupkovitz, G., Grausenburger, R., Brunmeir, R., Senese, S., Tischler, J., Jurkin, J., et al. The cyclin-dependent kinase inhibitor p21 is a crucial target for histone deacetylase 1 as a regulator of cellular proliferation. Mol Cell Biol, 2010, 30: 1171-1181, doi:10.1128/MCB.01500-09
    • (2010) Mol Cell Biol , vol.30 , pp. 1171-1181
    • Zupkovitz, G.1    Grausenburger, R.2    Brunmeir, R.3    Senese, S.4    Tischler, J.5    Jurkin, J.6
  • 16
    • 54349100901 scopus 로고    scopus 로고
    • HDAC4 promotes growth of colon cancer cells via repression of p21
    • Wilson, A. J., Byun, D. S., Nasser, S., Murray, L. B., Ayyanar, K., Arango, D., et al. HDAC4 promotes growth of colon cancer cells via repression of p21. Mol Biol Cell, 2008, 19: 4062-4075, doi:10.1091/mbc.E08-02-0139
    • (2008) Mol Biol Cell , vol.19 , pp. 4062-4075
    • Wilson, A.J.1    Byun, D.S.2    Nasser, S.3    Murray, L.B.4    Ayyanar, K.5    Arango, D.6
  • 17
    • 58249096131 scopus 로고    scopus 로고
    • HDAC4 represses p21(WAF1/Cip1) expression in human cancer cells through a Sp1-dependent, p53-independent mechanism
    • Mottet, D., Pirotte, S., Lamour, V., Hagedorn, M., Javerzat, S., Bikfalvi, A., et al. HDAC4 represses p21(WAF1/Cip1) expression in human cancer cells through a Sp1-dependent, p53-independent mechanism. Oncogene, 2009, 28: 243-256, doi:10.1038/onc.2008.371
    • (2009) Oncogene , vol.28 , pp. 243-256
    • Mottet, D.1    Pirotte, S.2    Lamour, V.3    Hagedorn, M.4    Javerzat, S.5    Bikfalvi, A.6
  • 18
    • 0035964743 scopus 로고    scopus 로고
    • Histone deacetylase and DNA methyltransferase in human prostate cancer
    • Patra, S. K., Patra, A., Dahiya, R. Histone deacetylase and DNA methyltransferase in human prostate cancer. Biochem Biophys Res Commun, 2001, 287: 705-713, doi:10.1006/bbrc.2001.5639
    • (2001) Biochem Biophys Res Commun , vol.287 , pp. 705-713
    • Patra, S.K.1    Patra, A.2    Dahiya, R.3
  • 19
    • 80051614029 scopus 로고    scopus 로고
    • Molecular marks for epigenetic identification of developmental and cancer stem cells
    • Patra, S. K., Deb, M., Patra, A. Molecular marks for epigenetic identification of developmental and cancer stem cells. Clin Epigenetics, 2011, 2: 27-53, doi:10.1007/s13148-010-0016-0
    • (2011) Clin Epigenetics , vol.2 , pp. 27-53
    • Patra, S.K.1    Deb, M.2    Patra, A.3
  • 20
    • 1842631408 scopus 로고    scopus 로고
    • Upregulation and nuclear recruitment of HDAC1 in hormone refractory prostate cancer
    • Halkidou, K., Gaughan, L., Cook, S., Leung, H. Y., Neal, D. E., Robson, C. N. Upregulation and nuclear recruitment of HDAC1 in hormone refractory prostate cancer. Prostate, 2004, 59: 177-189, doi:10.1002/pros.20022
    • (2004) Prostate , vol.59 , pp. 177-189
    • Halkidou, K.1    Gaughan, L.2    Cook, S.3    Leung, H.Y.4    Neal, D.E.5    Robson, C.N.6
  • 21
    • 38949086502 scopus 로고    scopus 로고
    • Histone deacetylases 1, 2 and 3 are highly expressed in prostate cancer and HDAC2 expression is associated with shorter PSA relapse time after radical prostatectomy
    • Weichert, W., Roske, A., Gekeler, V., Beckers, T., Stephan, C., Jung, K., et al. Histone deacetylases 1, 2 and 3 are highly expressed in prostate cancer and HDAC2 expression is associated with shorter PSA relapse time after radical prostatectomy. Br J Cancer, 2008, 98: 604-610, doi:10.1038/sj.bjc.6604199
    • (2008) Br J Cancer , vol.98 , pp. 604-610
    • Weichert, W.1    Roske, A.2    Gekeler, V.3    Beckers, T.4    Stephan, C.5    Jung, K.6
  • 22
    • 0036500996 scopus 로고    scopus 로고
    • Colorectal cancer in mice genetically deficient in the mucin Muc2
    • Velcich, A., Yang, W., Heyer, J., Fragale, A., Nicholas, C., Viani, S., et al. Colorectal cancer in mice genetically deficient in the mucin Muc2. Science, 2002, 295: 1726-1729, doi:10.1126/science.1069094
    • (2002) Science , vol.295 , pp. 1726-1729
    • Velcich, A.1    Yang, W.2    Heyer, J.3    Fragale, A.4    Nicholas, C.5    Viani, S.6
  • 23
    • 33748482616 scopus 로고    scopus 로고
    • MUC2 expression is regulated by histone H3 modification and DNA methylation in pancreatic cancer
    • Yamada, N., Hamada, T., Goto, M., Tsutsumida, H., Higashi, M., Nomoto, M., et al. MUC2 expression is regulated by histone H3 modification and DNA methylation in pancreatic cancer. Int J Cancer, 2006, 119: 1850-1857, doi:10.1002/ijc.22047
    • (2006) Int J Cancer , vol.119 , pp. 1850-1857
    • Yamada, N.1    Hamada, T.2    Goto, M.3    Tsutsumida, H.4    Higashi, M.5    Nomoto, M.6
  • 24
    • 33947658915 scopus 로고    scopus 로고
    • The short chain fatty acid, butyrate, stimulates MUC2 mucin production in the human colon cancer cell line, LS174T
    • Hatayama, H., Iwashita, J., Kuwajima, A., Abe, T. The short chain fatty acid, butyrate, stimulates MUC2 mucin production in the human colon cancer cell line, LS174T. Biochem Biophys Res Commun, 2007, 356: 599-603, doi:10.1016/j.bbrc.2007.03.025
    • (2007) Biochem Biophys Res Commun , vol.356 , pp. 599-603
    • Hatayama, H.1    Iwashita, J.2    Kuwajima, A.3    Abe, T.4
  • 25
    • 66549120804 scopus 로고    scopus 로고
    • The regulation of intestinal mucin MUC2 expression by short-chain fatty acids: implications for epithelial protection
    • Burger-van Paassen, N., Vincent, A., Puiman, P. J., van der Sluis, M., Bouma, J., Boehm, G., et al. The regulation of intestinal mucin MUC2 expression by short-chain fatty acids: implications for epithelial protection. Biochem J, 2009, 420: 211-219, doi:10.1042/BJ20082222
    • (2009) Biochem J , vol.420 , pp. 211-219
    • Burger-van Paassen, N.1    Vincent, A.2    Puiman, P.J.3    van der Sluis, M.4    Bouma, J.5    Boehm, G.6
  • 26
    • 77950814683 scopus 로고    scopus 로고
    • HDAC2 attenuates TRAIL-induced apoptosis of pancreatic cancer cells
    • Schuler, S., Fritsche, P., Diersch, S., Arlt, A., Schmid, R. M., Saur, D., et al. HDAC2 attenuates TRAIL-induced apoptosis of pancreatic cancer cells. Mol Cancer, 2010, 9: 80, doi:10.1186/1476-4598-9-80
    • (2010) Mol Cancer , vol.9 , pp. 80
    • Schuler, S.1    Fritsche, P.2    Diersch, S.3    Arlt, A.4    Schmid, R.M.5    Saur, D.6
  • 27
    • 70349309923 scopus 로고    scopus 로고
    • HDAC2 mediates therapeutic resistance of pancreatic cancer cells via the BH3-only protein NOXA
    • Fritsche, P., Seidler, B., Schuler, S., Schnieke, A., Gottlicher, M., Schmid, R. M., et al. HDAC2 mediates therapeutic resistance of pancreatic cancer cells via the BH3-only protein NOXA. Gut, 2009, 58: 1399-1409, doi:10.1136/gut.2009.180711
    • (2009) Gut , vol.58 , pp. 1399-1409
    • Fritsche, P.1    Seidler, B.2    Schuler, S.3    Schnieke, A.4    Gottlicher, M.5    Schmid, R.M.6
  • 28
    • 34347339526 scopus 로고    scopus 로고
    • Role for histone deacetylase 1 in human tumor cell proliferation
    • Senese, S., Zaragoza, K., Minardi, S., Muradore, I., Ronzoni, S., Passafaro, A., et al. Role for histone deacetylase 1 in human tumor cell proliferation. Mol Cell Biol, 2007, 27: 4784-4795, doi:10.1128/MCB.00494-07
    • (2007) Mol Cell Biol , vol.27 , pp. 4784-4795
    • Senese, S.1    Zaragoza, K.2    Minardi, S.3    Muradore, I.4    Ronzoni, S.5    Passafaro, A.6
  • 29
    • 80053161379 scopus 로고    scopus 로고
    • Combined inhibition of DNA methyltransferase and histone deacetylase restores caspase-8 expression and sensitizes SCLC cells to TRAIL
    • Kaminskyy, V. O., Surova, O. V., Vaculova, A., Zhivotovsky, B. Combined inhibition of DNA methyltransferase and histone deacetylase restores caspase-8 expression and sensitizes SCLC cells to TRAIL. Carcinogenesis, 2011, 32: 1450-1458, doi:10.1093/carcin/bgr135
    • (2011) Carcinogenesis , vol.32 , pp. 1450-1458
    • Kaminskyy, V.O.1    Surova, O.V.2    Vaculova, A.3    Zhivotovsky, B.4
  • 30
    • 67349128584 scopus 로고    scopus 로고
    • Reactivation of death receptor 4 (DR4) expression sensitizes medulloblastoma cell lines to TRAIL
    • Aguilera, D. G., Das, C. M., Sinnappah-Kang, N. D., Joyce, C., Taylor, P. H., Wen, S., et al. Reactivation of death receptor 4 (DR4) expression sensitizes medulloblastoma cell lines to TRAIL. J Neurooncol, 2009, 93: 303-318, doi:10.1007/s11060-008-9788-x
    • (2009) J Neurooncol , vol.93 , pp. 303-318
    • Aguilera, D.G.1    Das, C.M.2    Sinnappah-Kang, N.D.3    Joyce, C.4    Taylor, P.H.5    Wen, S.6
  • 31
    • 84859565614 scopus 로고    scopus 로고
    • HDAC2 overexpression confers oncogenic potential to human lung cancer cells by deregulating expression of apoptosis and cell cycle proteins
    • Jung, K. H., Noh, J. H., Kim, J. K., Eun, J. W., Bae, H. J., Xie, H. J., et al. HDAC2 overexpression confers oncogenic potential to human lung cancer cells by deregulating expression of apoptosis and cell cycle proteins. J Cell Biochem, 2012, 113: 2167-2177, doi:10.1002/jcb.24090
    • (2012) J Cell Biochem , vol.113 , pp. 2167-2177
    • Jung, K.H.1    Noh, J.H.2    Kim, J.K.3    Eun, J.W.4    Bae, H.J.5    Xie, H.J.6
  • 32
    • 33745895281 scopus 로고    scopus 로고
    • The histone deacetylase inhibitor, suberoylanilide hydroxamic acid, overcomes resistance of human breast cancer cells to Apo2L/TRAIL
    • Butler, L. M., Liapis, V., Bouralexis, S., Welldon, K., Hay, S., Thaile, M., et al. The histone deacetylase inhibitor, suberoylanilide hydroxamic acid, overcomes resistance of human breast cancer cells to Apo2L/TRAIL. Int J Cancer, 2006, 119: 944-954, doi:10.1002/ijc.21939
    • (2006) Int J Cancer , vol.119 , pp. 944-954
    • Butler, L.M.1    Liapis, V.2    Bouralexis, S.3    Welldon, K.4    Hay, S.5    Thaile, M.6
  • 33
    • 67649354926 scopus 로고    scopus 로고
    • Suberoylanilide hydroxamic acid (Zolinza/vorinostat) sensitizes TRAIL-resistant breast cancer cells orthotopically implanted in BALB/c nude mice
    • Shankar, S., Davis, R., Singh, K. P., Kurzrock, R., Ross, D. D., Srivastava, R. K. Suberoylanilide hydroxamic acid (Zolinza/vorinostat) sensitizes TRAIL-resistant breast cancer cells orthotopically implanted in BALB/c nude mice. Mol Cancer Ther, 2009, 8: 1596-1605, doi:10.1158/1535-7163.MCT-08-1004
    • (2009) Mol Cancer Ther , vol.8 , pp. 1596-1605
    • Shankar, S.1    Davis, R.2    Singh, K.P.3    Kurzrock, R.4    Ross, D.D.5    Srivastava, R.K.6
  • 34
    • 84862125485 scopus 로고    scopus 로고
    • HDAC5 is required for maintenance of pericentric heterochromatin, and controls cell-cycle progression and survival of human cancer cells
    • Peixoto, P., Castronovo, V., Matheus, N., Polese, C., Peulen, O., Gonzalez, A., et al. HDAC5 is required for maintenance of pericentric heterochromatin, and controls cell-cycle progression and survival of human cancer cells. Cell Death Differ, 2012, 19: 1239-1252, doi:10.1038/cdd.2012.3
    • (2012) Cell Death Differ , vol.19 , pp. 1239-1252
    • Peixoto, P.1    Castronovo, V.2    Matheus, N.3    Polese, C.4    Peulen, O.5    Gonzalez, A.6
  • 36
    • 80055077899 scopus 로고    scopus 로고
    • HDAC4 protein regulates HIF1alpha protein lysine acetylation and cancer cell response to hypoxia
    • Geng, H., Harvey, C. T., Pittsenbarger, J., Liu, Q., Beer, T. M., Xue, C., et al. HDAC4 protein regulates HIF1alpha protein lysine acetylation and cancer cell response to hypoxia. J Biol Chem, 2011, 286: 38095-38102, doi:10.1074/jbc.M111.257055
    • (2011) J Biol Chem , vol.286 , pp. 38095-38102
    • Geng, H.1    Harvey, C.T.2    Pittsenbarger, J.3    Liu, Q.4    Beer, T.M.5    Xue, C.6
  • 37
    • 19944399717 scopus 로고    scopus 로고
    • 6-Phosphofructo-2-kinase (pfkfb3) gene promoter contains hypoxia-inducible factor-1 binding sites necessary for transactivation in response to hypoxia
    • Obach, M., Navarro-Sabate, A., Caro, J., Kong, X., Duran, J., Gomez, M., et al. 6-Phosphofructo-2-kinase (pfkfb3) gene promoter contains hypoxia-inducible factor-1 binding sites necessary for transactivation in response to hypoxia. J Biol Chem, 2004, 279: 53562-53570, doi:10.1074/jbc.M406096200
    • (2004) J Biol Chem , vol.279 , pp. 53562-53570
    • Obach, M.1    Navarro-Sabate, A.2    Caro, J.3    Kong, X.4    Duran, J.5    Gomez, M.6
  • 39
    • 71749091061 scopus 로고    scopus 로고
    • The expression and significance of HIF-1alpha and GLUT-3 in glioma
    • Liu, Y., Li, Y. M., Tian, R. F., Liu, W. P., Fei, Z., Long, Q. F., et al. The expression and significance of HIF-1alpha and GLUT-3 in glioma. Brain Res, 2009, 1304: 149-154, doi:10.1016/j.brainres.2009.09.083
    • (2009) Brain Res , vol.1304 , pp. 149-154
    • Liu, Y.1    Li, Y.M.2    Tian, R.F.3    Liu, W.P.4    Fei, Z.5    Long, Q.F.6
  • 40
    • 58149310725 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors and genomic instability
    • Eot-Houllier, G., Fulcrand, G., Magnaghi-Jaulin, L., Jaulin, C. Histone deacetylase inhibitors and genomic instability. Cancer Lett, 2009, 274: 169-176, doi:10.1016/j.canlet.2008.06.005
    • (2009) Cancer Lett , vol.274 , pp. 169-176
    • Eot-Houllier, G.1    Fulcrand, G.2    Magnaghi-Jaulin, L.3    Jaulin, C.4
  • 41
    • 32944468240 scopus 로고    scopus 로고
    • A histone deacetylase inhibitor, trichostatin A, enhances radiosensitivity by abrogating G2/M arrest in human carcinoma cells
    • Kim, I. A., Kim, J. H., Shin, J. H., Kim, I. H., Kim, J. S., Wu, H. G., et al. A histone deacetylase inhibitor, trichostatin A, enhances radiosensitivity by abrogating G2/M arrest in human carcinoma cells. Cancer Res Treat, 2005, 37: 122-128, doi:10.4143/crt.2005.37.2.122
    • (2005) Cancer Res Treat , vol.37 , pp. 122-128
    • Kim, I.A.1    Kim, J.H.2    Shin, J.H.3    Kim, I.H.4    Kim, J.S.5    Wu, H.G.6
  • 42
    • 0242522928 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors
    • Miller, T. A., Witter, D. J., Belvedere, S. Histone deacetylase inhibitors. J Med Chem, 2003, 46: 5097-5116, doi:10.1021/jm0303094
    • (2003) J Med Chem , vol.46 , pp. 5097-5116
    • Miller, T.A.1    Witter, D.J.2    Belvedere, S.3
  • 43
    • 67349232749 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors that target tubulin
    • Schemies, J., Sippl, W., Jung, M. Histone deacetylase inhibitors that target tubulin. Cancer Lett, 2009, 280: 222-232, doi:10.1016/j.canlet.2009.01.040
    • (2009) Cancer Lett , vol.280 , pp. 222-232
    • Schemies, J.1    Sippl, W.2    Jung, M.3
  • 44
    • 2942564591 scopus 로고    scopus 로고
    • Sirtuins: Sir2-related NAD-dependent protein deacetylases
    • North, B. J., Verdin, E. Sirtuins: Sir2-related NAD-dependent protein deacetylases. Genome Biol, 2004, 5: 224, doi:10.1186/gb-2004-5-5-224
    • (2004) Genome Biol , vol.5 , pp. 224
    • North, B.J.1    Verdin, E.2
  • 45
    • 36249016246 scopus 로고    scopus 로고
    • A therapeutic role for sirtuins in diseases of aging?
    • Westphal, C. H., Dipp, M. A., Guarente, L. A therapeutic role for sirtuins in diseases of aging? Trends Biochem Sci, 2007, 32: 555-560, doi:10.1016/j.tibs.2007.09.008
    • (2007) Trends Biochem Sci , vol.32 , pp. 555-560
    • Westphal, C.H.1    Dipp, M.A.2    Guarente, L.3
  • 46
    • 39349113616 scopus 로고    scopus 로고
    • Substituting N(epsilon)-thioacetyl-lysine for N(epsilon)-acetyl-lysine in peptide substrates as a general approach to inhibiting human NAD(+)-dependent protein deacetylases
    • Fatkins, D. G., Zheng, W. Substituting N(epsilon)-thioacetyl-lysine for N(epsilon)-acetyl-lysine in peptide substrates as a general approach to inhibiting human NAD(+)-dependent protein deacetylases. Int J Mol Sci, 2008, 9: 1-11
    • (2008) Int J Mol Sci , vol.9 , pp. 1-11
    • Fatkins, D.G.1    Zheng, W.2
  • 47
    • 4544318283 scopus 로고    scopus 로고
    • Design, synthesis, and biological evaluation of a small-molecule inhibitor of the histone acetyltransferase Gcn5
    • Biel, M., Kretsovali, A., Karatzali, E., Papamatheakis, J., Giannis, A. Design, synthesis, and biological evaluation of a small-molecule inhibitor of the histone acetyltransferase Gcn5. Angew Chem Int Ed Engl, 2004, 43: 3974-3976, doi:10.1002/anie.200453879
    • (2004) Angew Chem Int Ed Engl , vol.43 , pp. 3974-3976
    • Biel, M.1    Kretsovali, A.2    Karatzali, E.3    Papamatheakis, J.4    Giannis, A.5
  • 48
    • 33645221885 scopus 로고    scopus 로고
    • Inhibition of SIRT1 catalytic activity increases p53 acetylation but does not alter cell survival following DNA damage
    • Solomon, J. M., Pasupuleti, R., Xu, L., McDonagh, T., Curtis, R., DiStefano, P. S., et al. Inhibition of SIRT1 catalytic activity increases p53 acetylation but does not alter cell survival following DNA damage. Mol Cell Biol, 2006, 26: 28-38, doi:10.1128/MCB.26.1.28-38.2006
    • (2006) Mol Cell Biol , vol.26 , pp. 28-38
    • Solomon, J.M.1    Pasupuleti, R.2    Xu, L.3    McDonagh, T.4    Curtis, R.5    DiStefano, P.S.6
  • 49
    • 0842277812 scopus 로고    scopus 로고
    • Histone deacetylase (HDAC) inhibitor activation of p21WAF1 involves changes in promoter-associated proteins, including HDAC1
    • Gui, C. Y., Ngo, L., Xu, W. S., Richon, V. M., Marks, P. A. Histone deacetylase (HDAC) inhibitor activation of p21WAF1 involves changes in promoter-associated proteins, including HDAC1. Proc Natl Acad Sci U S A, 2004, 101: 1241-1246, doi:10.1073/pnas.0307708100
    • (2004) Proc Natl Acad Sci U S A , vol.101 , pp. 1241-1246
    • Gui, C.Y.1    Ngo, L.2    Xu, W.S.3    Richon, V.M.4    Marks, P.A.5
  • 50
    • 65649111534 scopus 로고    scopus 로고
    • Novel cambinol analogs as sirtuin inhibitors: synthesis, biological evaluation, and rationalization of activity
    • Medda, F., Russell, R. J., Higgins, M., McCarthy, A. R., Campbell, J., Slawin, A. M., et al. Novel cambinol analogs as sirtuin inhibitors: synthesis, biological evaluation, and rationalization of activity. J Med Chem, 2009, 52: 2673-2682, doi:10.1021/jm8014298
    • (2009) J Med Chem , vol.52 , pp. 2673-2682
    • Medda, F.1    Russell, R.J.2    Higgins, M.3    McCarthy, A.R.4    Campbell, J.5    Slawin, A.M.6
  • 51
    • 0033822112 scopus 로고    scopus 로고
    • P21-dependent g(1)arrest with downregulation of cyclin D1 and upregulation of cyclin E by the histone deacetylase inhibitor FR901228
    • Sandor, V., Senderowicz, A., Mertins, S., Sackett, D., Sausville, E., Blagosklonny, M. V., et al. P21-dependent g(1)arrest with downregulation of cyclin D1 and upregulation of cyclin E by the histone deacetylase inhibitor FR901228. Br J Cancer, 2000, 83: 817-825, doi:10.1054/bjoc.2000.1327
    • (2000) Br J Cancer , vol.83 , pp. 817-825
    • Sandor, V.1    Senderowicz, A.2    Mertins, S.3    Sackett, D.4    Sausville, E.5    Blagosklonny, M.V.6
  • 52
    • 0034721941 scopus 로고    scopus 로고
    • Inhibition of mitogenesis in Balb/c-3T3 cells by Trichostatin A. Multiple alterations in the induction and activation of cyclin-cyclin-dependent kinase complexes
    • Wharton, W., Savell, J., Cress, W. D., Seto, E., Pledger, W. J. Inhibition of mitogenesis in Balb/c-3T3 cells by Trichostatin A. Multiple alterations in the induction and activation of cyclin-cyclin-dependent kinase complexes. J Biol Chem, 2000, 275: 33981-33987, doi:10.1074/jbc.M005600200
    • (2000) J Biol Chem , vol.275 , pp. 33981-33987
    • Wharton, W.1    Savell, J.2    Cress, W.D.3    Seto, E.4    Pledger, W.J.5
  • 53
    • 33748451151 scopus 로고    scopus 로고
    • Anticancer activities of histone deacetylase inhibitors
    • Bolden, J. E., Peart, M. J., Johnstone, R. W. Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov, 2006, 5: 769-784, doi:10.1038/nrd2133
    • (2006) Nat Rev Drug Discov , vol.5 , pp. 769-784
    • Bolden, J.E.1    Peart, M.J.2    Johnstone, R.W.3
  • 54
    • 26444446597 scopus 로고    scopus 로고
    • Histone modifications as a platform for cancer therapy
    • Espino, P. S., Drobic, B., Dunn, K. L., Davie, J. R. Histone modifications as a platform for cancer therapy. J Cell Biochem, 2005, 94: 1088-1102, doi:10.1002/jcb.20387
    • (2005) J Cell Biochem , vol.94 , pp. 1088-1102
    • Espino, P.S.1    Drobic, B.2    Dunn, K.L.3    Davie, J.R.4
  • 55
    • 84897838897 scopus 로고    scopus 로고
    • Trichostatin A suppresses EGFR expression through induction of microRNA-7 in an HDAC-independent manner in lapatinib-treated cells
    • Tu, C. Y., Chen, C. H., Hsia, T. C., Hsu, M. H., Wei, Y. L., Yu, M. C., et al. Trichostatin A suppresses EGFR expression through induction of microRNA-7 in an HDAC-independent manner in lapatinib-treated cells. Biomed Res Int, 2014, 2014: 168949, doi:10.1155/2014/168949
    • (2014) Biomed Res Int , vol.2014 , pp. 168949
    • Tu, C.Y.1    Chen, C.H.2    Hsia, T.C.3    Hsu, M.H.4    Wei, Y.L.5    Yu, M.C.6
  • 56
    • 12444321545 scopus 로고    scopus 로고
    • Phase I clinical trial of histone deacetylase inhibitor: suberoylanilide hydroxamic acid administered intravenously
    • Kelly, W. K., Richon, V. M., O'Connor, O., Curley, T., MacGregor-Curtelli, B., Tong, W., et al. Phase I clinical trial of histone deacetylase inhibitor: suberoylanilide hydroxamic acid administered intravenously. Clin Cancer Res, 2003, 9: 3578-3588
    • (2003) Clin Cancer Res , vol.9 , pp. 3578-3588
    • Kelly, W.K.1    Richon, V.M.2    O'Connor, O.3    Curley, T.4    MacGregor-Curtelli, B.5    Tong, W.6
  • 57
    • 21244467166 scopus 로고    scopus 로고
    • Epigenetic and chromatin modifiers as targeted therapy of hematologic malignancies
    • Bhalla, K. N. Epigenetic and chromatin modifiers as targeted therapy of hematologic malignancies. J Clin Oncol, 2005, 23: 3971-3993, doi:10.1200/JCO.2005.16.600
    • (2005) J Clin Oncol , vol.23 , pp. 3971-3993
    • Bhalla, K.N.1
  • 58
    • 26444439216 scopus 로고    scopus 로고
    • Prospects: histone deacetylase inhibitors
    • Dokmanovic, M., Marks, P. A. Prospects: histone deacetylase inhibitors. J Cell Biochem, 2005, 96: 293-304, doi:10.1002/jcb.20532
    • (2005) J Cell Biochem , vol.96 , pp. 293-304
    • Dokmanovic, M.1    Marks, P.A.2
  • 59
    • 16544379283 scopus 로고    scopus 로고
    • Sequence-specific potentiation of topoisomerase II inhibitors by the histone deacetylase inhibitor suberoylanilide hydroxamic acid
    • Marchion, D. C., Bicaku, E., Daud, A. I., Richon, V., Sullivan, D. M., Munster, P. N. Sequence-specific potentiation of topoisomerase II inhibitors by the histone deacetylase inhibitor suberoylanilide hydroxamic acid. J Cell Biochem, 2004, 92: 223-237, doi:10.1002/jcb.20045
    • (2004) J Cell Biochem , vol.92 , pp. 223-237
    • Marchion, D.C.1    Bicaku, E.2    Daud, A.I.3    Richon, V.4    Sullivan, D.M.5    Munster, P.N.6
  • 60
    • 33745714230 scopus 로고    scopus 로고
    • Combined DNA methyltransferase and histone deacetylase inhibition in the treatment of myeloid neoplasms
    • Gore, S. D., Baylin, S., Sugar, E., Carraway, H., Miller, C. B., Carducci, M., et al. Combined DNA methyltransferase and histone deacetylase inhibition in the treatment of myeloid neoplasms. Cancer Res, 2006, 66: 6361-6369, doi:10.1158/0008-5472.CAN-06-0080
    • (2006) Cancer Res , vol.66 , pp. 6361-6369
    • Gore, S.D.1    Baylin, S.2    Sugar, E.3    Carraway, H.4    Miller, C.B.5    Carducci, M.6
  • 61
    • 31444438952 scopus 로고    scopus 로고
    • Pilot study of combination transcriptional modulation therapy with sodium phenylbutyrate and 5-azacytidine in patients with acute myeloid leukemia or myelodysplastic syndrome
    • Maslak, P., Chanel, S., Camacho, L. H., Soignet, S., Pandolfi, P. P., Guernah, I., et al. Pilot study of combination transcriptional modulation therapy with sodium phenylbutyrate and 5-azacytidine in patients with acute myeloid leukemia or myelodysplastic syndrome. Leukemia, 2006, 20: 212-217, doi:10.1038/sj.leu.2404050
    • (2006) Leukemia , vol.20 , pp. 212-217
    • Maslak, P.1    Chanel, S.2    Camacho, L.H.3    Soignet, S.4    Pandolfi, P.P.5    Guernah, I.6
  • 62
    • 33750530675 scopus 로고    scopus 로고
    • Phase 1/2 study of the combination of 5-aza-2'-deoxycytidine with valproic acid in patients with leukemia
    • Garcia-Manero, G., Kantarjian, H. M., Sanchez-Gonzalez, B., Yang, H., Rosner, G., Verstovsek, S., et al. Phase 1/2 study of the combination of 5-aza-2'-deoxycytidine with valproic acid in patients with leukemia. Blood, 2006, 108: 3271-3279, doi:10.1182/blood-2006-03-009142
    • (2006) Blood , vol.108 , pp. 3271-3279
    • Garcia-Manero, G.1    Kantarjian, H.M.2    Sanchez-Gonzalez, B.3    Yang, H.4    Rosner, G.5    Verstovsek, S.6
  • 63
    • 0038284073 scopus 로고    scopus 로고
    • Phase I study of oral CI-994 in combination with gemcitabine in treatment of patients with advanced cancer
    • Nemunaitis, J. J., Orr, D., Eager, R., Cunningham, C. C., Williams, A., Mennel, R., et al. Phase I study of oral CI-994 in combination with gemcitabine in treatment of patients with advanced cancer. Cancer J, 2003, 9: 58-66
    • (2003) Cancer J , vol.9 , pp. 58-66
    • Nemunaitis, J.J.1    Orr, D.2    Eager, R.3    Cunningham, C.C.4    Williams, A.5    Mennel, R.6
  • 64
    • 9444287120 scopus 로고    scopus 로고
    • A phase I study of the oral combination of CI-994, a putative histone deacetylase inhibitor, and capecitabine
    • Undevia, S. D., Kindler, H. L., Janisch, L., Olson, S. C., Schilsky, R. L., Vogelzang, N. J., et al. A phase I study of the oral combination of CI-994, a putative histone deacetylase inhibitor, and capecitabine. Ann Oncol, 2004, 15: 1705-1711, doi:10.1093/annonc/mdh438
    • (2004) Ann Oncol , vol.15 , pp. 1705-1711
    • Undevia, S.D.1    Kindler, H.L.2    Janisch, L.3    Olson, S.C.4    Schilsky, R.L.5    Vogelzang, N.J.6
  • 65
    • 0242493856 scopus 로고    scopus 로고
    • The proteasome inhibitor bortezomib interacts synergistically with histone deacetylase inhibitors to induce apoptosis in Bcr/Abl+ cells sensitive and resistant to STI571
    • Yu, C., Rahmani, M., Conrad, D., Subler, M., Dent, P., Grant, S. The proteasome inhibitor bortezomib interacts synergistically with histone deacetylase inhibitors to induce apoptosis in Bcr/Abl+ cells sensitive and resistant to STI571. Blood, 2003, 102: 3765-3774, doi:10.1182/blood-2003-03-0737
    • (2003) Blood , vol.102 , pp. 3765-3774
    • Yu, C.1    Rahmani, M.2    Conrad, D.3    Subler, M.4    Dent, P.5    Grant, S.6
  • 66
    • 33745888156 scopus 로고    scopus 로고
    • Combined effects of novel tyrosine kinase inhibitor AMN107 and histone deacetylase inhibitor LBH589 against Bcr-Abl-expressing human leukemia cells
    • Fiskus, W., Pranpat, M., Bali, P., Balasis, M., Kumaraswamy, S., Boyapalle, S., et al. Combined effects of novel tyrosine kinase inhibitor AMN107 and histone deacetylase inhibitor LBH589 against Bcr-Abl-expressing human leukemia cells. Blood, 2006, 108: 645-652, doi:10.1182/blood-2005-11-4639
    • (2006) Blood , vol.108 , pp. 645-652
    • Fiskus, W.1    Pranpat, M.2    Bali, P.3    Balasis, M.4    Kumaraswamy, S.5    Boyapalle, S.6
  • 67
    • 26444561533 scopus 로고    scopus 로고
    • Physical and functional interactions between the human DNMT3L protein and members of the de novo methyltransferase family
    • Chen, Z. X., Mann, J. R., Hsieh, C. L., Riggs, A. D., Chedin, F. Physical and functional interactions between the human DNMT3L protein and members of the de novo methyltransferase family. J Cell Biochem, 2005, 95: 902-917, doi:10.1002/jcb.20447
    • (2005) J Cell Biochem , vol.95 , pp. 902-917
    • Chen, Z.X.1    Mann, J.R.2    Hsieh, C.L.3    Riggs, A.D.4    Chedin, F.5
  • 68
    • 78651162036 scopus 로고
    • Acetylation and Methylation of Histones and Their Possible Role in the Regulation of Rna Synthesis
    • Allfrey, V. G., Faulkner, R., Mirsky, A. E. Acetylation and Methylation of Histones and Their Possible Role in the Regulation of Rna Synthesis. Proc Natl Acad Sci U S A, 1964, 51: 786-794
    • (1964) Proc Natl Acad Sci U S A , vol.51 , pp. 786-794
    • Allfrey, V.G.1    Faulkner, R.2    Mirsky, A.E.3
  • 69
    • 0028885077 scopus 로고
    • Identification of a gene encoding a yeast histone H4 acetyltransferase
    • Kleff, S., Andrulis, E. D., Anderson, C. W., Sternglanz, R. Identification of a gene encoding a yeast histone H4 acetyltransferase. J Biol Chem, 1995, 270: 24674-24677
    • (1995) J Biol Chem , vol.270 , pp. 24674-24677
    • Kleff, S.1    Andrulis, E.D.2    Anderson, C.W.3    Sternglanz, R.4
  • 70
    • 0022365083 scopus 로고
    • Deposition-related histone acetylation in micronuclei of conjugating Tetrahymena
    • Allis, C. D., Chicoine, L. G., Richman, R., Schulman, I. G. Deposition-related histone acetylation in micronuclei of conjugating Tetrahymena. Proc Natl Acad Sci U S A, 1985, 82: 8048-8052
    • (1985) Proc Natl Acad Sci U S A , vol.82 , pp. 8048-8052
    • Allis, C.D.1    Chicoine, L.G.2    Richman, R.3    Schulman, I.G.4
  • 71
    • 0016723918 scopus 로고
    • Processing of newly synthesized histone molecules
    • Ruiz-Carrillo, A., Wangh, L. J., Allfrey, V. G. Processing of newly synthesized histone molecules. Science, 1975, 190: 117-128
    • (1975) Science , vol.190 , pp. 117-128
    • Ruiz-Carrillo, A.1    Wangh, L.J.2    Allfrey, V.G.3
  • 72
    • 0030271392 scopus 로고    scopus 로고
    • The major cytoplasmic histone acetyltransferase in yeast: links to chromatin replication and histone metabolism
    • Parthun, M. R., Widom, J., Gottschling, D. E. The major cytoplasmic histone acetyltransferase in yeast: links to chromatin replication and histone metabolism. Cell, 1996, 87: 85-94
    • (1996) Cell , vol.87 , pp. 85-94
    • Parthun, M.R.1    Widom, J.2    Gottschling, D.E.3
  • 73
    • 84869988557 scopus 로고    scopus 로고
    • Histone acetyltransferases: Rising ancient counterparts to protein kinases
    • Yuan, H., Marmorstein, R. Histone acetyltransferases: Rising ancient counterparts to protein kinases. Biopolymers, 2013, 99: 98-111, doi:10.1002/bip.22128
    • (2013) Biopolymers , vol.99 , pp. 98-111
    • Yuan, H.1    Marmorstein, R.2
  • 74
    • 0020403437 scopus 로고
    • Inhibition of histone acetylation by N-[2-(S-coenzyme A)acetyl] spermidine amide, a multisubstrate analog
    • Cullis, P. M., Wolfenden, R., Cousens, L. S., Alberts, B. M. Inhibition of histone acetylation by N-[2-(S-coenzyme A)acetyl] spermidine amide, a multisubstrate analog. J Biol Chem, 1982, 257: 12165-12169
    • (1982) J Biol Chem , vol.257 , pp. 12165-12169
    • Cullis, P.M.1    Wolfenden, R.2    Cousens, L.S.3    Alberts, B.M.4
  • 75
    • 4043146501 scopus 로고    scopus 로고
    • Polyisoprenylated benzophenone, garcinol, a natural histone acetyltransferase inhibitor, represses chromatin transcription and alters global gene expression
    • Balasubramanyam, K., Altaf, M., Varier, R. A., Swaminathan, V., Ravindran, A., Sadhale, P. P., et al. Polyisoprenylated benzophenone, garcinol, a natural histone acetyltransferase inhibitor, represses chromatin transcription and alters global gene expression. J Biol Chem, 2004, 279: 33716-33726, doi:10.1074/jbc.M402839200
    • (2004) J Biol Chem , vol.279 , pp. 33716-33726
    • Balasubramanyam, K.1    Altaf, M.2    Varier, R.A.3    Swaminathan, V.4    Ravindran, A.5    Sadhale, P.P.6
  • 76
    • 10944243759 scopus 로고    scopus 로고
    • Curcumin, a novel p300/CREB-binding protein-specific inhibitor of acetyltransferase, represses the acetylation of histone/nonhistone proteins and histone acetyltransferase-dependent chromatin transcription
    • Balasubramanyam, K., Varier, R. A., Altaf, M., Swaminathan, V., Siddappa, N. B., Ranga, U., et al. Curcumin, a novel p300/CREB-binding protein-specific inhibitor of acetyltransferase, represses the acetylation of histone/nonhistone proteins and histone acetyltransferase-dependent chromatin transcription. J Biol Chem, 2004, 279: 51163-51171, doi:10.1074/jbc.M409024200
    • (2004) J Biol Chem , vol.279 , pp. 51163-51171
    • Balasubramanyam, K.1    Varier, R.A.2    Altaf, M.3    Swaminathan, V.4    Siddappa, N.B.5    Ranga, U.6
  • 77
    • 27644473204 scopus 로고    scopus 로고
    • Isothiazolones as inhibitors of PCAF and p300 histone acetyltransferase activity
    • Stimson, L., Rowlands, M. G., Newbatt, Y. M., Smith, N. F., Raynaud, F. I., Rogers, P., et al. Isothiazolones as inhibitors of PCAF and p300 histone acetyltransferase activity. Mol Cancer Ther, 2005, 4: 1521-1532, doi:10.1158/1535-7163.MCT-05-0135
    • (2005) Mol Cancer Ther , vol.4 , pp. 1521-1532
    • Stimson, L.1    Rowlands, M.G.2    Newbatt, Y.M.3    Smith, N.F.4    Raynaud, F.I.5    Rogers, P.6
  • 78
    • 34547864553 scopus 로고    scopus 로고
    • Distinct GCN5/PCAF-containing complex es function as co-activators and are involved in transcription factor and global histone acetylation
    • Nagy, Z., Tora, L. Distinct GCN5/PCAF-containing complex es function as co-activators and are involved in transcription factor and global histone acetylation. Oncogene, 2007, 26: 5341-5357, doi:10.1038/sj.onc.1210604
    • (2007) Oncogene , vol.26 , pp. 5341-5357
    • Nagy, Z.1    Tora, L.2
  • 79
    • 24944516931 scopus 로고    scopus 로고
    • A role for the Tip60 histone acetyltransferase in the acetylation and activation of ATM
    • Sun, Y., Jiang, X., Chen, S., Fernandes, N., Price, B. D. A role for the Tip60 histone acetyltransferase in the acetylation and activation of ATM. Proc Natl Acad Sci U S A, 2005, 102: 13182-13187, doi:10.1073/pnas.0504211102
    • (2005) Proc Natl Acad Sci U S A , vol.102 , pp. 13182-13187
    • Sun, Y.1    Jiang, X.2    Chen, S.3    Fernandes, N.4    Price, B.D.5
  • 80
    • 84870375316 scopus 로고    scopus 로고
    • Histone lysine methylation dynamics: establishment, regulation, and biological impact
    • Black, J. C., Van Rechem, C., Whetstine, J. R. Histone lysine methylation dynamics: establishment, regulation, and biological impact. Mol Cell, 2012, 48: 491-507, doi:10.1016/j.molcel.2012.11.006S1097-2765(12)00937-9 [pii]
    • (2012) Mol Cell , vol.48 , pp. 491-507
    • Black, J.C.1    Van Rechem, C.2    Whetstine, J.R.3
  • 81
    • 0034632829 scopus 로고    scopus 로고
    • Regulation of chromatin structure by site-specific histone H3 methyltransferases
    • Rea, S., Eisenhaber, F., O'Carroll, D., Strahl, B. D., Sun, Z. W., Schmid, M., et al. Regulation of chromatin structure by site-specific histone H3 methyltransferases. Nature, 2000, 406: 593-599, doi:10.1038/35020506
    • (2000) Nature , vol.406 , pp. 593-599
    • Rea, S.1    Eisenhaber, F.2    O'Carroll, D.3    Strahl, B.D.4    Sun, Z.W.5    Schmid, M.6
  • 82
    • 33745632390 scopus 로고    scopus 로고
    • The epigenetic magic of histone lysine methylation
    • Jenuwein, T. The epigenetic magic of histone lysine methylation. FEBS J, 2006, 273: 3121-3135, doi:10.1111/j.1742-4658.2006.05343.x
    • (2006) FEBS J , vol.273 , pp. 3121-3135
    • Jenuwein, T.1
  • 83
    • 23944509075 scopus 로고    scopus 로고
    • The SET-domain protein superfamily: protein lysine methyltransferases
    • Dillon, S. C., Zhang, X., Trievel, R. C., Cheng, X. The SET-domain protein superfamily: protein lysine methyltransferases. Genome Biol, 2005, 6: 227, doi:10.1186/gb-2005-6-8-227
    • (2005) Genome Biol , vol.6 , pp. 227
    • Dillon, S.C.1    Zhang, X.2    Trievel, R.C.3    Cheng, X.4
  • 84
    • 17444375685 scopus 로고    scopus 로고
    • hDOT1L links histone methylation to leukemogenesis
    • Okada, Y., Feng, Q., Lin, Y., Jiang, Q., Li, Y., Coffield, V. M., et al. hDOT1L links histone methylation to leukemogenesis. Cell, 2005, 121: 167-178, doi:10.1016/j.cell.2005.02.020
    • (2005) Cell , vol.121 , pp. 167-178
    • Okada, Y.1    Feng, Q.2    Lin, Y.3    Jiang, Q.4    Li, Y.5    Coffield, V.M.6
  • 85
    • 0037077178 scopus 로고    scopus 로고
    • Dot1p modulates silencing in yeast by methylation of the nucleosome core
    • van Leeuwen, F., Gafken, P. R., Gottschling, D. E. Dot1p modulates silencing in yeast by methylation of the nucleosome core. Cell, 2002, 109: 745-756
    • (2002) Cell , vol.109 , pp. 745-756
    • van Leeuwen, F.1    Gafken, P.R.2    Gottschling, D.E.3
  • 86
    • 79959960773 scopus 로고    scopus 로고
    • The diverse functions of Dot1 and H3K79 methylation
    • Nguyen, A. T., Zhang, Y. The diverse functions of Dot1 and H3K79 methylation. Genes Dev, 2011, 25: 1345-1358, doi:10.1101/gad.2057811
    • (2011) Genes Dev , vol.25 , pp. 1345-1358
    • Nguyen, A.T.1    Zhang, Y.2
  • 87
    • 0242669199 scopus 로고    scopus 로고
    • Coordinated histone modifications mediated by a CtBP co-repressor complex
    • Shi, Y., Sawada, J., Sui, G., Affarel, B., Whetstine, J. R., Lan, F., et al. Coordinated histone modifications mediated by a CtBP co-repressor complex. Nature, 2003, 422: 735-738, doi:10.1038/nature01550
    • (2003) Nature , vol.422 , pp. 735-738
    • Shi, Y.1    Sawada, J.2    Sui, G.3    Affarel, B.4    Whetstine, J.R.5    Lan, F.6
  • 88
    • 11144332565 scopus 로고    scopus 로고
    • Histone demethylation mediated by the nuclear amine oxidase homolog LSD1
    • Shi, Y., Lan, F., Matson, C., Mulligan, P., Whetstine, J. R., Cole, P. A., et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell, 2004, 119: 941-953, doi:10.1016/j.cell.2004.12.012
    • (2004) Cell , vol.119 , pp. 941-953
    • Shi, Y.1    Lan, F.2    Matson, C.3    Mulligan, P.4    Whetstine, J.R.5    Cole, P.A.6
  • 89
    • 33646138230 scopus 로고    scopus 로고
    • JHDM2A, a JmjC-containing H3K9 demethylase, facilitates transcription activation by androgen receptor
    • Yamane, K., Toumazou, C., Tsukada, Y., Erdjument-Bromage, H., Tempst, P., Wong, J., et al. JHDM2A, a JmjC-containing H3K9 demethylase, facilitates transcription activation by androgen receptor. Cell, 2006, 125: 483-495, doi:10.1016/j.cell.2006.03.027
    • (2006) Cell , vol.125 , pp. 483-495
    • Yamane, K.1    Toumazou, C.2    Tsukada, Y.3    Erdjument-Bromage, H.4    Tempst, P.5    Wong, J.6
  • 90
    • 32844454603 scopus 로고    scopus 로고
    • Histone demethylation by a family of JmjC domain-containing proteins
    • Tsukada, Y., Fang, J., Erdjument-Bromage, H., Warren, M. E., Borchers, C. H., Tempst, P., et al. Histone demethylation by a family of JmjC domain-containing proteins. Nature, 2006, 439: 811-816, doi:10.1038/nature04433
    • (2006) Nature , vol.439 , pp. 811-816
    • Tsukada, Y.1    Fang, J.2    Erdjument-Bromage, H.3    Warren, M.E.4    Borchers, C.H.5    Tempst, P.6
  • 91
    • 33846025127 scopus 로고    scopus 로고
    • Dynamic regulation of histone lysine methylation by demethylases
    • Shi, Y., Whetstine, J. R. Dynamic regulation of histone lysine methylation by demethylases. Mol Cell, 2007, 25: 1-14, doi:10.1016/j.molcel.2006.12.010
    • (2007) Mol Cell , vol.25 , pp. 1-14
    • Shi, Y.1    Whetstine, J.R.2
  • 92
    • 33745847680 scopus 로고    scopus 로고
    • The transcriptional repressor JHDM3A demethylates trimethyl histone H3 lysine 9 and lysine 36
    • Klose, R. J., Yamane, K., Bae, Y., Zhang, D., Erdjument-Bromage, H., Tempst, P., et al. The transcriptional repressor JHDM3A demethylates trimethyl histone H3 lysine 9 and lysine 36. Nature, 2006, 442: 312-316, doi:10.1038/nature04853
    • (2006) Nature , vol.442 , pp. 312-316
    • Klose, R.J.1    Yamane, K.2    Bae, Y.3    Zhang, D.4    Erdjument-Bromage, H.5    Tempst, P.6
  • 93
    • 33745146571 scopus 로고    scopus 로고
    • Jmjd2b antagonizes H3K9 trimethylation at pericentric heterochromatin in mammalian cells
    • Fodor, B. D., Kubicek, S., Yonezawa, M., O'Sullivan, R. J., Sengupta, R., Perez-Burgos, L., et al. Jmjd2b antagonizes H3K9 trimethylation at pericentric heterochromatin in mammalian cells. Genes Dev, 2006, 20: 1557-1562, doi:10.1101/gad.388206
    • (2006) Genes Dev , vol.20 , pp. 1557-1562
    • Fodor, B.D.1    Kubicek, S.2    Yonezawa, M.3    O'Sullivan, R.J.4    Sengupta, R.5    Perez-Burgos, L.6
  • 94
    • 33746332412 scopus 로고    scopus 로고
    • The putative oncogene GASC1 demethylates tri- and dimethylated lysine 9 on histone H3
    • Cloos, P. A., Christensen, J., Agger, K., Maiolica, A., Rappsilber, J., Antal, T., et al. The putative oncogene GASC1 demethylates tri- and dimethylated lysine 9 on histone H3. Nature, 2006, 442: 307-311, doi:10.1038/nature04837
    • (2006) Nature , vol.442 , pp. 307-311
    • Cloos, P.A.1    Christensen, J.2    Agger, K.3    Maiolica, A.4    Rappsilber, J.5    Antal, T.6
  • 95
    • 33646124469 scopus 로고    scopus 로고
    • Reversal of histone lysine trimethylation by the JMJD2 family of histone demethylases
    • Whetstine, J. R., Nottke, A., Lan, F., Huarte, M., Smolikov, S., Chen, Z., et al. Reversal of histone lysine trimethylation by the JMJD2 family of histone demethylases. Cell, 2006, 125: 467-481, doi:10.1016/j.cell.2006.03.028
    • (2006) Cell , vol.125 , pp. 467-481
    • Whetstine, J.R.1    Nottke, A.2    Lan, F.3    Huarte, M.4    Smolikov, S.5    Chen, Z.6
  • 96
    • 67649800263 scopus 로고    scopus 로고
    • Dynamic Histone H1 Isotype 4 Methylation and Demethylation by Histone Lysine Methyltransferase G9a/KMT1C and the Jumonji Domain-containing JMJD2/KDM4 Proteins
    • Trojer, P., Zhang, J., Yonezawa, M., Schmidt, A., Zheng, H., Jenuwein, T., et al. Dynamic Histone H1 Isotype 4 Methylation and Demethylation by Histone Lysine Methyltransferase G9a/KMT1C and the Jumonji Domain-containing JMJD2/KDM4 Proteins. J Biol Chem, 2009, 284: 8395-8405, doi:10.1074/jbc.M807818200
    • (2009) J Biol Chem , vol.284 , pp. 8395-8405
    • Trojer, P.1    Zhang, J.2    Yonezawa, M.3    Schmidt, A.4    Zheng, H.5    Jenuwein, T.6
  • 97
    • 33847076849 scopus 로고    scopus 로고
    • Chromatin modifications and their function
    • Kouzarides, T. Chromatin modifications and their function. Cell, 2007, 128: 693-705, doi:10.1016/j.cell.2007.02.005
    • (2007) Cell , vol.128 , pp. 693-705
    • Kouzarides, T.1
  • 98
    • 34948839944 scopus 로고    scopus 로고
    • Facultative heterochromatin: is there a distinctive molecular signature?
    • Trojer, P., Reinberg, D. Facultative heterochromatin: is there a distinctive molecular signature? Mol Cell, 2007, 28: 1-13, doi:10.1016/j.molcel.2007.09.011
    • (2007) Mol Cell , vol.28 , pp. 1-13
    • Trojer, P.1    Reinberg, D.2
  • 99
    • 34848911602 scopus 로고    scopus 로고
    • Selective anchoring of TFIID to nucleosomes by trimethylation of histone H3 lysine 4
    • Vermeulen, M., Mulder, K. W., Denissov, S., Pijnappel, W. W., van Schaik, F. M., Varier, R. A., et al. Selective anchoring of TFIID to nucleosomes by trimethylation of histone H3 lysine 4. Cell, 2007, 131: 58-69, doi:10.1016/j.cell.2007.08.016
    • (2007) Cell , vol.131 , pp. 58-69
    • Vermeulen, M.1    Mulder, K.W.2    Denissov, S.3    Pijnappel, W.W.4    van Schaik, F.M.5    Varier, R.A.6
  • 100
    • 0035282573 scopus 로고    scopus 로고
    • Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins
    • Lachner, M., O'Carroll, D., Rea, S., Mechtler, K., Jenuwein, T. Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins. Nature, 2001, 410: 116-120, doi:10.1038/35065132
    • (2001) Nature , vol.410 , pp. 116-120
    • Lachner, M.1    O'Carroll, D.2    Rea, S.3    Mechtler, K.4    Jenuwein, T.5
  • 101
    • 84895925247 scopus 로고    scopus 로고
    • Context-specific regulation of cancer epigenomes by histone and transcription factor methylation
    • Sarris, M., Nikolaou, K., Talianidis, I. Context-specific regulation of cancer epigenomes by histone and transcription factor methylation. Oncogene, 2014, 33: 1207-1217, doi:10.1038/onc.2013.87onc201387 [pii]
    • (2014) Oncogene , vol.33 , pp. 1207-1217
    • Sarris, M.1    Nikolaou, K.2    Talianidis, I.3
  • 102
    • 45549087777 scopus 로고    scopus 로고
    • Direct interaction between SET8 and proliferating cell nuclear antigen couples H4-K20 methylation with DNA replication
    • Huen, M. S., Sy, S. M., van Deursen, J. M., Chen, J. Direct interaction between SET8 and proliferating cell nuclear antigen couples H4-K20 methylation with DNA replication. J Biol Chem, 2008, 283: 11073-11077, doi:10.1074/jbc.C700242200
    • (2008) J Biol Chem , vol.283 , pp. 11073-11077
    • Huen, M.S.1    Sy, S.M.2    van Deursen, J.M.3    Chen, J.4
  • 103
    • 38049075810 scopus 로고    scopus 로고
    • The histone methyltransferase SET8 is required for S-phase progression
    • Jorgensen, S., Elvers, I., Trelle, M. B., Menzel, T., Eskildsen, M., Jensen, O. N., et al. The histone methyltransferase SET8 is required for S-phase progression. J Cell Biol, 2007, 179: 1337-1345, doi:10.1083/jcb.200706150
    • (2007) J Cell Biol , vol.179 , pp. 1337-1345
    • Jorgensen, S.1    Elvers, I.2    Trelle, M.B.3    Menzel, T.4    Eskildsen, M.5    Jensen, O.N.6
  • 104
    • 0036714189 scopus 로고    scopus 로고
    • Mitotic-specific methylation of histone H4 Lys 20 follows increased PR-Set7 expression and its localization to mitotic chromosomes
    • Rice, J. C., Nishioka, K., Sarma, K., Steward, R., Reinberg, D., Allis, C. D. Mitotic-specific methylation of histone H4 Lys 20 follows increased PR-Set7 expression and its localization to mitotic chromosomes. Genes Dev, 2002, 16: 2225-2230, doi:10.1101/gad.1014902
    • (2002) Genes Dev , vol.16 , pp. 2225-2230
    • Rice, J.C.1    Nishioka, K.2    Sarma, K.3    Steward, R.4    Reinberg, D.5    Allis, C.D.6
  • 105
    • 78651330886 scopus 로고    scopus 로고
    • SET8 is degraded via PCNA-coupled CRL4(CDT2) ubiquitylation in S phase and after UV irradiation
    • Jorgensen, S., Eskildsen, M., Fugger, K., Hansen, L., Larsen, M. S., Kousholt, A. N., et al. SET8 is degraded via PCNA-coupled CRL4(CDT2) ubiquitylation in S phase and after UV irradiation. J Cell Biol, 2011, 192: 43-54, doi:10.1083/jcb.201009076
    • (2011) J Cell Biol , vol.192 , pp. 43-54
    • Jorgensen, S.1    Eskildsen, M.2    Fugger, K.3    Hansen, L.4    Larsen, M.S.5    Kousholt, A.N.6
  • 106
    • 78149281634 scopus 로고    scopus 로고
    • The histone H4 Lys 20 methyltransferase PR-Set7 regulates replication origins in mammalian cells
    • Tardat, M., Brustel, J., Kirsh, O., Lefevbre, C., Callanan, M., Sardet, C., et al. The histone H4 Lys 20 methyltransferase PR-Set7 regulates replication origins in mammalian cells. Nat Cell Biol, 2010, 12: 1086-1093, doi:10.1038/ncb2113
    • (2010) Nat Cell Biol , vol.12 , pp. 1086-1093
    • Tardat, M.1    Brustel, J.2    Kirsh, O.3    Lefevbre, C.4    Callanan, M.5    Sardet, C.6
  • 108
    • 33845666681 scopus 로고    scopus 로고
    • Structural basis for the methylation state-specific recognition of histone H4-K20 by 53BP1 and Crb2 in DNA repair
    • Botuyan, M. V., Lee, J., Ward, I. M., Kim, J. E., Thompson, J. R., Chen, J., et al. Structural basis for the methylation state-specific recognition of histone H4-K20 by 53BP1 and Crb2 in DNA repair. Cell, 2006, 127: 1361-1373, doi:10.1016/j.cell.2006.10.043
    • (2006) Cell , vol.127 , pp. 1361-1373
    • Botuyan, M.V.1    Lee, J.2    Ward, I.M.3    Kim, J.E.4    Thompson, J.R.5    Chen, J.6
  • 109
    • 84904884937 scopus 로고    scopus 로고
    • EGFR modulates DNA synthesis and repair through Tyr phosphorylation of histone H4
    • Chou, R. H., Wang, Y. N., Hsieh, Y. H., Li, L. Y., Xia, W., Chang, W. C., et al. EGFR modulates DNA synthesis and repair through Tyr phosphorylation of histone H4. Dev Cell, 2014, 30: 224-237, doi:10.1016/j.devcel.2014.06.008
    • (2014) Dev Cell , vol.30 , pp. 224-237
    • Chou, R.H.1    Wang, Y.N.2    Hsieh, Y.H.3    Li, L.Y.4    Xia, W.5    Chang, W.C.6
  • 110
    • 2642542643 scopus 로고    scopus 로고
    • A silencing pathway to induce H3-K9 and H4-K20 trimethylation at constitutive heterochromatin
    • Schotta, G., Lachner, M., Sarma, K., Ebert, A., Sengupta, R., Reuter, G., et al. A silencing pathway to induce H3-K9 and H4-K20 trimethylation at constitutive heterochromatin. Genes Dev, 2004, 18: 1251-1262, doi:10.1101/gad.300704
    • (2004) Genes Dev , vol.18 , pp. 1251-1262
    • Schotta, G.1    Lachner, M.2    Sarma, K.3    Ebert, A.4    Sengupta, R.5    Reuter, G.6
  • 111
    • 84855340668 scopus 로고    scopus 로고
    • SET8 promotes epithelial-mesenchymal transition and confers TWIST dual transcriptional activities
    • Yang, F., Sun, L., Li, Q., Han, X., Lei, L., Zhang, H., et al. SET8 promotes epithelial-mesenchymal transition and confers TWIST dual transcriptional activities. EMBO J, 2012, 31: 110-123, doi:10.1038/emboj.2011.364
    • (2012) EMBO J , vol.31 , pp. 110-123
    • Yang, F.1    Sun, L.2    Li, Q.3    Han, X.4    Lei, L.5    Zhang, H.6
  • 112
    • 0037172665 scopus 로고    scopus 로고
    • Methylation of H3-lysine 79 is mediated by a new family of HMTases without a SET domain
    • Feng, Q., Wang, H., Ng, H. H., Erdjument-Bromage, H., doi:Tempst, P., Struhl, K., et al. Methylation of H3-lysine 79 is mediated by a new family of HMTases without a SET domain. Curr Biol, 2002, 12: 1052-1058
    • (2002) Curr Biol , vol.12 , pp. 1052-1058
    • Feng, Q.1    Wang, H.2    Ng, H.H.3    Erdjument-Bromage, H.4    Tempst, P.5    Struhl, K.6
  • 113
    • 0344837759 scopus 로고    scopus 로고
    • Structure of the catalytic domain of human DOT1L, a non-SET domain nucleosomal histone methyltransferase
    • Min, J., Feng, Q., Li, Z., Zhang, Y., Xu, R. M. Structure of the catalytic domain of human DOT1L, a non-SET domain nucleosomal histone methyltransferase. Cell, 2003, 112: 711-723
    • (2003) Cell , vol.112 , pp. 711-723
    • Min, J.1    Feng, Q.2    Li, Z.3    Zhang, Y.4    Xu, R.M.5
  • 114
    • 0028210328 scopus 로고
    • Crystal structure of catechol O-methyltransferase
    • Vidgren, J., Svensson, L. A., Liljas, A. Crystal structure of catechol O-methyltransferase. Nature, 1994, 368: 354-358, doi:10.1038/368354a0
    • (1994) Nature , vol.368 , pp. 354-358
    • Vidgren, J.1    Svensson, L.A.2    Liljas, A.3
  • 115
    • 77949563562 scopus 로고    scopus 로고
    • Targeting DOT1L action and interactions in leukemia: the role of DOT1L in transformation and development
    • Barry, E. R., Corry, G. N., Rasmussen, T. P. Targeting DOT1L action and interactions in leukemia: the role of DOT1L in transformation and development. Expert Opin Ther Targets, 2010, 14: 405-418, doi:10.1517/14728221003623241
    • (2010) Expert Opin Ther Targets , vol.14 , pp. 405-418
    • Barry, E.R.1    Corry, G.N.2    Rasmussen, T.P.3
  • 116
    • 82455186371 scopus 로고    scopus 로고
    • A role for DOT1L in MLL-rearranged leukemias
    • Bernt, K. M., Armstrong, S. A. A role for DOT1L in MLL-rearranged leukemias. Epigenomics, 2011, 3: 667-670, doi:10.2217/epi.11.98
    • (2011) Epigenomics , vol.3 , pp. 667-670
    • Bernt, K.M.1    Armstrong, S.A.2
  • 117
    • 84878383491 scopus 로고    scopus 로고
    • Leukemic transformation by the MLL-AF6 fusion oncogene requires the H3K79 methyltransferase Dot1l
    • Deshpande, A. J., Chen, L., Fazio, M., Sinha, A. U., Bernt, K. M., Banka, D., et al. Leukemic transformation by the MLL-AF6 fusion oncogene requires the H3K79 methyltransferase Dot1l. Blood, 2013, 121: 2533-2541, doi:10.1182/blood-2012-11-465120
    • (2013) Blood , vol.121 , pp. 2533-2541
    • Deshpande, A.J.1    Chen, L.2    Fazio, M.3    Sinha, A.U.4    Bernt, K.M.5    Banka, D.6
  • 118
    • 43249118972 scopus 로고    scopus 로고
    • Polycomb group genes: keeping stem cell activity in balance
    • Sauvageau, M., Sauvageau, G. Polycomb group genes: keeping stem cell activity in balance. PLoS Biol, 2008, 6: e113, doi:10.1371/journal.pbio.0060113
    • (2008) PLoS Biol , vol.6 , pp. e113
    • Sauvageau, M.1    Sauvageau, G.2
  • 119
    • 80053139713 scopus 로고    scopus 로고
    • (-)-Epigallocatechin-3-gallate and DZNep reduce polycomb protein level via a proteasome-dependent mechanism in skin cancer cells
    • Choudhury, S. R., Balasubramanian, S., Chew, Y. C., Han, B., Marquez, V. E., Eckert, R. L. (-)-Epigallocatechin-3-gallate and DZNep reduce polycomb protein level via a proteasome-dependent mechanism in skin cancer cells. Carcinogenesis, 2011, 32: 1525-1532, doi:10.1093/carcin/bgr171
    • (2011) Carcinogenesis , vol.32 , pp. 1525-1532
    • Choudhury, S.R.1    Balasubramanian, S.2    Chew, Y.C.3    Han, B.4    Marquez, V.E.5    Eckert, R.L.6
  • 120
    • 55949136562 scopus 로고    scopus 로고
    • Roles of the EZH2 histone methyltransferase in cancer epigenetics
    • Simon, J. A., Lange, C. A. Roles of the EZH2 histone methyltransferase in cancer epigenetics. Mutat Res, 2008, 647: 21-29, doi:10.1016/j.mrfmmm.2008.07.010
    • (2008) Mutat Res , vol.647 , pp. 21-29
    • Simon, J.A.1    Lange, C.A.2
  • 121
    • 82555173684 scopus 로고    scopus 로고
    • EZH2 methyltransferase and H3K27 methylation in breast cancer
    • Yoo, K. H., Hennighausen, L. EZH2 methyltransferase and H3K27 methylation in breast cancer. Int J Biol Sci, 2012, 8: 59-65
    • (2012) Int J Biol Sci , vol.8 , pp. 59-65
    • Yoo, K.H.1    Hennighausen, L.2
  • 122
    • 73649102464 scopus 로고    scopus 로고
    • EZH2 is essential for glioblastoma cancer stem cell maintenance
    • Suva, M. L., Riggi, N., Janiszewska, M., Radovanovic, I., Provero, P., Stehle, J. C., et al. EZH2 is essential for glioblastoma cancer stem cell maintenance. Cancer Res, 2009, 69: 9211-9218, doi:10.1158/0008-5472.CAN-09-1622
    • (2009) Cancer Res , vol.69 , pp. 9211-9218
    • Suva, M.L.1    Riggi, N.2    Janiszewska, M.3    Radovanovic, I.4    Provero, P.5    Stehle, J.C.6
  • 123
    • 77957193274 scopus 로고    scopus 로고
    • Clinical significance of Polycomb gene expression in brain tumors
    • Crea, F., Hurt, E. M., Farrar, W. L. Clinical significance of Polycomb gene expression in brain tumors. Mol Cancer, 2010, 9: 265, doi:10.1186/1476-4598-9-265
    • (2010) Mol Cancer , vol.9 , pp. 265
    • Crea, F.1    Hurt, E.M.2    Farrar, W.L.3
  • 124
    • 79959416877 scopus 로고    scopus 로고
    • Enhancer of Zeste 2 (EZH2) is up-regulated in malignant gliomas and in glioma stem-like cells
    • Orzan, F., Pellegatta, S., Poliani, P. L., Pisati, F., Caldera, V., Menghi, F., et al. Enhancer of Zeste 2 (EZH2) is up-regulated in malignant gliomas and in glioma stem-like cells. Neuropathol Appl Neurobiol, 2011, 37: 381-394, doi:10.1111/j.1365-2990.2010.01132.x
    • (2011) Neuropathol Appl Neurobiol , vol.37 , pp. 381-394
    • Orzan, F.1    Pellegatta, S.2    Poliani, P.L.3    Pisati, F.4    Caldera, V.5    Menghi, F.6
  • 125
    • 84894093372 scopus 로고    scopus 로고
    • Altered expression of polycomb group genes in glioblastoma multiforme
    • Li, G., Warden, C., Zou, Z., Neman, J., Krueger, J. S., Jain, A., et al. Altered expression of polycomb group genes in glioblastoma multiforme. PLoS One, 2013, 8: e80970, doi:10.1371/journal.pone.0080970
    • (2013) PLoS One , vol.8 , pp. e80970
    • Li, G.1    Warden, C.2    Zou, Z.3    Neman, J.4    Krueger, J.S.5    Jain, A.6
  • 126
    • 79957563769 scopus 로고    scopus 로고
    • The roles of EZH2 in cell lineage commitment
    • Chou, R. H., Yu, Y. L., Hung, M. C. The roles of EZH2 in cell lineage commitment. Am J Transl Res, 2011, 3: 243-250
    • (2011) Am J Transl Res , vol.3 , pp. 243-250
    • Chou, R.H.1    Yu, Y.L.2    Hung, M.C.3
  • 127
    • 79953211498 scopus 로고    scopus 로고
    • EZH2 regulates neuronal differentiation of mesenchymal stem cells through PIP5K1C-dependent calcium signaling
    • Yu, Y. L., Chou, R. H., Chen, L. T., Shyu, W. C., Hsieh, S. C., Wu, C. S., et al. EZH2 regulates neuronal differentiation of mesenchymal stem cells through PIP5K1C-dependent calcium signaling. J Biol Chem, 2011, 286: 9657-9667, doi:10.1074/jbc.M110.185124
    • (2011) J Biol Chem , vol.286 , pp. 9657-9667
    • Yu, Y.L.1    Chou, R.H.2    Chen, L.T.3    Shyu, W.C.4    Hsieh, S.C.5    Wu, C.S.6
  • 128
    • 84875811405 scopus 로고    scopus 로고
    • Smurf2-mediated degradation of EZH2 enhances neuron differentiation and improves functional recovery after ischaemic stroke
    • Yu, Y. L., Chou, R. H., Shyu, W. C., Hsieh, S. C., Wu, C. S., Chiang, S. Y., et al. Smurf2-mediated degradation of EZH2 enhances neuron differentiation and improves functional recovery after ischaemic stroke. EMBO Mol Med, 2013, 5: 531-547, doi:10.1002/emmm.201201783
    • (2013) EMBO Mol Med , vol.5 , pp. 531-547
    • Yu, Y.L.1    Chou, R.H.2    Shyu, W.C.3    Hsieh, S.C.4    Wu, C.S.5    Chiang, S.Y.6
  • 129
    • 84927168923 scopus 로고    scopus 로고
    • The potential roles of EZH2 in regenerative medicine
    • Chou, R. H., Chiu, L., Yu, Y. L., Shyu, W. C. The potential roles of EZH2 in regenerative medicine. Cell Transplant, 2015, 24: 313-317, doi:10.3727/096368915X686823
    • (2015) Cell Transplant , vol.24 , pp. 313-317
    • Chou, R.H.1    Chiu, L.2    Yu, Y.L.3    Shyu, W.C.4
  • 130
    • 77953444201 scopus 로고    scopus 로고
    • High expression of EZH2 is associated with tumor aggres siveness and poor prognosis in patients with esophageal squamous cell carcinoma treated with definitive chemoradiotherapy
    • He, L. R., Liu, M. Z., Li, B. K., Jia, W. H., Zhang, Y., Liao, Y. J., et al. High expression of EZH2 is associated with tumor aggres siveness and poor prognosis in patients with esophageal squamous cell carcinoma treated with definitive chemoradiotherapy. Int J Cancer, 2010, 127: 138-147, doi:10.1002/ijc.25031
    • (2010) Int J Cancer , vol.127 , pp. 138-147
    • He, L.R.1    Liu, M.Z.2    Li, B.K.3    Jia, W.H.4    Zhang, Y.5    Liao, Y.J.6
  • 131
    • 78049307003 scopus 로고    scopus 로고
    • Overexpression of EZH2 contributes to acquired cisplatin resistance in ovarian cancer cells in vitro and in vivo
    • Hu, S., Yu, L., Li, Z., Shen, Y., Wang, J., Cai, J., et al. Overexpression of EZH2 contributes to acquired cisplatin resistance in ovarian cancer cells in vitro and in vivo. Cancer Biol Ther, 2010, 10: 788-795
    • (2010) Cancer Biol Ther , vol.10 , pp. 788-795
    • Hu, S.1    Yu, L.2    Li, Z.3    Shen, Y.4    Wang, J.5    Cai, J.6
  • 132
    • 79951825513 scopus 로고    scopus 로고
    • Ovarian cancer stem cell-like side populations are enriched following chemotherapy and overexpress EZH2
    • Rizzo, S., Hersey, J. M., Mellor, P., Dai, W., Santos-Silva, A., Liber, D., et al. Ovarian cancer stem cell-like side populations are enriched following chemotherapy and overexpress EZH2. Mol Cancer Ther, 2011, 10: 325-335, doi:10.1158/1535-7163.MCT-10-0788
    • (2011) Mol Cancer Ther , vol.10 , pp. 325-335
    • Rizzo, S.1    Hersey, J.M.2    Mellor, P.3    Dai, W.4    Santos-Silva, A.5    Liber, D.6
  • 133
    • 78651454089 scopus 로고    scopus 로고
    • EZH2 promotes expansion of breast tumor initiating cells through activation of RAF1-beta-catenin signaling
    • Chang, C. J., Yang, J. Y., Xia, W., Chen, C. T., Xie, X., Chao, C. H., et al. EZH2 promotes expansion of breast tumor initiating cells through activation of RAF1-beta-catenin signaling. Cancer Cell, 2011, 19: 86-100, doi:S1535-6108(10)00437-X [pii]10.1016/j.ccr.2010.10.035
    • (2011) Cancer Cell , vol.19 , pp. 86-100
    • Chang, C.J.1    Yang, J.Y.2    Xia, W.3    Chen, C.T.4    Xie, X.5    Chao, C.H.6
  • 134
    • 84863343713 scopus 로고    scopus 로고
    • Loss of let-7 up-regulates EZH2 in prostate cancer consistent with the acquisition of cancer stem cell signatures that are attenuated by BR-DIM
    • Kong, D., Heath, E., Chen, W., Cher, M. L., Powell, I., Heilbrun, L., et al. Loss of let-7 up-regulates EZH2 in prostate cancer consistent with the acquisition of cancer stem cell signatures that are attenuated by BR-DIM. PLoS One, 2012, 7: e33729, doi:10.1371/journal.pone.0033729
    • (2012) PLoS One , vol.7 , pp. e33729
    • Kong, D.1    Heath, E.2    Chen, W.3    Cher, M.L.4    Powell, I.5    Heilbrun, L.6
  • 135
    • 78650987804 scopus 로고    scopus 로고
    • Decreased expression of EZH2 is associated with upregulation of ER and favorable outcome to tamoxifen in advanced breast cancer
    • Reijm, E. A., Jansen, M. P., Ruigrok-Ritstier, K., van Staveren, I. L., Look, M. P., van Gelder, M. E., et al. Decreased expression of EZH2 is associated with upregulation of ER and favorable outcome to tamoxifen in advanced breast cancer. Breast Cancer Res Treat, 2011, 125: 387-394, doi:10.1007/s10549-010-0836-9
    • (2011) Breast Cancer Res Treat , vol.125 , pp. 387-394
    • Reijm, E.A.1    Jansen, M.P.2    Ruigrok-Ritstier, K.3    van Staveren, I.L.4    Look, M.P.5    van Gelder, M.E.6
  • 136
    • 84863722942 scopus 로고    scopus 로고
    • High miR-26a and low CDC2 levels associate with decreased EZH2 expression and with favorable outcome on tamoxifen in metastatic breast cancer
    • Jansen, M. P., Reijm, E. A., Sieuwerts, A. M., Ruigrok-Ritstier, K., Look, M. P., Rodriguez-Gonzalez, F. G., et al. High miR-26a and low CDC2 levels associate with decreased EZH2 expression and with favorable outcome on tamoxifen in metastatic breast cancer. Breast Cancer Res Treat, 2012, 133: 937-947, doi:10.1007/s10549-011-1877-4
    • (2012) Breast Cancer Res Treat , vol.133 , pp. 937-947
    • Jansen, M.P.1    Reijm, E.A.2    Sieuwerts, A.M.3    Ruigrok-Ritstier, K.4    Look, M.P.5    Rodriguez-Gonzalez, F.G.6
  • 137
    • 84876789029 scopus 로고    scopus 로고
    • Silencing the EZH2 gene by RNA interference reverses the drug resistance of human hepatic multidrug-resistant cancer cells to 5-Fu
    • Zhang, Y., Liu, G., Lin, C., Liao, G., Tang, B. Silencing the EZH2 gene by RNA interference reverses the drug resistance of human hepatic multidrug-resistant cancer cells to 5-Fu. Life Sci, 2013, 92: 896-902, doi:10.1016/j.lfs.2013.03.010
    • (2013) Life Sci , vol.92 , pp. 896-902
    • Zhang, Y.1    Liu, G.2    Lin, C.3    Liao, G.4    Tang, B.5
  • 138
    • 84872782925 scopus 로고    scopus 로고
    • RNAi-mediated EZH2 depletion decreases MDR1 expression and sensitizes multidrug-resistant hepatocellular carcinoma cells to chemotherapy
    • Tang, B., Zhang, Y., Liang, R., Gao, Z., Sun, D., Wang, L. RNAi-mediated EZH2 depletion decreases MDR1 expression and sensitizes multidrug-resistant hepatocellular carcinoma cells to chemotherapy. Oncol Rep, 2013, 29: 1037-1042, doi:10.3892/or.2013.2222
    • (2013) Oncol Rep , vol.29 , pp. 1037-1042
    • Tang, B.1    Zhang, Y.2    Liang, R.3    Gao, Z.4    Sun, D.5    Wang, L.6
  • 139
    • 79960058024 scopus 로고    scopus 로고
    • Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor
    • Daigle, S. R., Olhava, E. J., Therkelsen, C. A., Majer, C. R., Sneeringer, C. J., Song, J., et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell, 2011, 20: 53-65, doi:10.1016/j.ccr.2011.06.009
    • (2011) Cancer Cell , vol.20 , pp. 53-65
    • Daigle, S.R.1    Olhava, E.J.2    Therkelsen, C.A.3    Majer, C.R.4    Sneeringer, C.J.5    Song, J.6
  • 140
    • 84876143288 scopus 로고    scopus 로고
    • Abrogation of MLL-AF10 and CALM-AF10-mediated transformation through genetic inactivation or pharmacological inhibition of the H3K79 methyltransferase Dot1l
    • Chen, L., Deshpande, A. J., Banka, D., Bernt, K. M., Dias, S., Buske, C., et al. Abrogation of MLL-AF10 and CALM-AF10-mediated transformation through genetic inactivation or pharmacological inhibition of the H3K79 methyltransferase Dot1l. Leukemia, 2013, 27: 813-822, doi:10.1038/leu.2012.327
    • (2013) Leukemia , vol.27 , pp. 813-822
    • Chen, L.1    Deshpande, A.J.2    Banka, D.3    Bernt, K.M.4    Dias, S.5    Buske, C.6
  • 141
    • 84921370808 scopus 로고    scopus 로고
    • Regulation of Wnt signaling target gene expression by the histone methyltransferase DOT1L
    • Gibbons, G. S., Owens, S. R., Fearon, E. R., Nikolovska-Coleska, Z. Regulation of Wnt signaling target gene expression by the histone methyltransferase DOT1L. ACS Chem Biol, 2015, 10: 109-114, doi:10.1021/cb500668u
    • (2015) ACS Chem Biol , vol.10 , pp. 109-114
    • Gibbons, G.S.1    Owens, S.R.2    Fearon, E.R.3    Nikolovska-Coleska, Z.4
  • 143
    • 84899459437 scopus 로고    scopus 로고
    • Nonclinical pharmacokinetics and metabolism of EPZ-5676, a novel DOT1L histone methyltransferase inhibitor
    • Basavapathruni, A., Olhava, E. J., Daigle, S. R., Therkelsen, C. A., Jin, L., Boriack-Sjodin, P. A., et al. Nonclinical pharmacokinetics and metabolism of EPZ-5676, a novel DOT1L histone methyltransferase inhibitor. Biopharm Drug Dispos, 2014, 35: 237-252, doi:10.1002/bdd.1889
    • (2014) Biopharm Drug Dispos , vol.35 , pp. 237-252
    • Basavapathruni, A.1    Olhava, E.J.2    Daigle, S.R.3    Therkelsen, C.A.4    Jin, L.5    Boriack-Sjodin, P.A.6
  • 144
    • 84929100337 scopus 로고    scopus 로고
    • MLL partial tandem duplication leukemia cells are sensitive to small molecule DOT1L inhibition
    • Kuhn, M. W., Hadler, M. J., Daigle, S. R., Koche, R. P., Krivtsov, A. V., Olhava, E. J., et al. MLL partial tandem duplication leukemia cells are sensitive to small molecule DOT1L inhibition. Haematologica, 2015, 100: e190-193, doi:10.3324/haematol.2014.115337
    • (2015) Haematologica , vol.100 , pp. e190-193
    • Kuhn, M.W.1    Hadler, M.J.2    Daigle, S.R.3    Koche, R.P.4    Krivtsov, A.V.5    Olhava, E.J.6
  • 145
    • 84907291965 scopus 로고    scopus 로고
    • DOT1L inhibitor EPZ-5676 displays synergistic antiproliferative activity in combination with standard of care drugs and hypomethylating agents in MLL-rearranged leukemia cells
    • Klaus, C. R., Iwanowicz, D., Johnston, D., Campbell, C. A., Smith, J. J., Moyer, M. P., et al. DOT1L inhibitor EPZ-5676 displays synergistic antiproliferative activity in combination with standard of care drugs and hypomethylating agents in MLL-rearranged leukemia cells. J Pharmacol Exp Ther, 2014, 350: 646-656, doi:10.1124/jpet.114.214577
    • (2014) J Pharmacol Exp Ther , vol.350 , pp. 646-656
    • Klaus, C.R.1    Iwanowicz, D.2    Johnston, D.3    Campbell, C.A.4    Smith, J.J.5    Moyer, M.P.6
  • 146
    • 84870573126 scopus 로고    scopus 로고
    • EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations
    • McCabe, M. T., Ott, H. M., Ganji, G., Korenchuk, S., Thompson, C., Van Aller, G. S., et al. EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature, 2012, 492: 108-112, doi:10.1038/nature11606
    • (2012) Nature , vol.492 , pp. 108-112
    • McCabe, M.T.1    Ott, H.M.2    Ganji, G.3    Korenchuk, S.4    Thompson, C.5    Van Aller, G.S.6
  • 147
    • 84871841675 scopus 로고    scopus 로고
    • Selective inhibition of Ezh2 by a small molecule inhibitor blocks tumor cells proliferation
    • Qi, W., Chan, H., Teng, L., Li, L., Chuai, S., Zhang, R., et al. Selective inhibition of Ezh2 by a small molecule inhibitor blocks tumor cells proliferation. Proc Natl Acad Sci U S A, 2012, 109: 21360-21365, doi:10.1073/pnas.1210371110
    • (2012) Proc Natl Acad Sci U S A , vol.109 , pp. 21360-21365
    • Qi, W.1    Chan, H.2    Teng, L.3    Li, L.4    Chuai, S.5    Zhang, R.6
  • 148
    • 84879750981 scopus 로고    scopus 로고
    • An orally bioavailable chemical probe of the Lysine Methyltransferases EZH2 and EZH1
    • Konze, K. D., Ma, A., Li, F., Barsyte-Lovejoy, D., Parton, T., Macnevin, C. J., et al. An orally bioavailable chemical probe of the Lysine Methyltransferases EZH2 and EZH1. ACS Chem Biol, 2013, 8: 1324-1334, doi:10.1021/cb400133j
    • (2013) ACS Chem Biol , vol.8 , pp. 1324-1334
    • Konze, K.D.1    Ma, A.2    Li, F.3    Barsyte-Lovejoy, D.4    Parton, T.5    Macnevin, C.J.6
  • 149
    • 84921318899 scopus 로고    scopus 로고
    • Selective inhibition of EZH2 by EPZ-6438 leads to potent antitumor activity in EZH2-mutant non-Hodgkin lymphoma
    • Knutson, S. K., Kawano, S., Minoshima, Y., Warholic, N. M., Huang, K. C., Xiao, Y., et al. Selective inhibition of EZH2 by EPZ-6438 leads to potent antitumor activity in EZH2-mutant non-Hodgkin lymphoma. Mol Cancer Ther, 2014, 13: 842-854, doi:10.1158/1535-7163.MCT-13-0773
    • (2014) Mol Cancer Ther , vol.13 , pp. 842-854
    • Knutson, S.K.1    Kawano, S.2    Minoshima, Y.3    Warholic, N.M.4    Huang, K.C.5    Xiao, Y.6
  • 150
    • 84911462255 scopus 로고    scopus 로고
    • EZH2 inhibitor efficacy in non-Hodgkin's lymphoma does not require suppression of H3K27 monomethylation
    • Bradley, W. D., Arora, S., Busby, J., Balasubramanian, S., Gehling, V. S., Nasveschuk, C. G., et al. EZH2 inhibitor efficacy in non-Hodgkin's lymphoma does not require suppression of H3K27 monomethylation. Chem Biol, 2014, 21: 1463-1475, doi:10.1016/j.chembiol.2014.09.017
    • (2014) Chem Biol , vol.21 , pp. 1463-1475
    • Bradley, W.D.1    Arora, S.2    Busby, J.3    Balasubramanian, S.4    Gehling, V.S.5    Nasveschuk, C.G.6
  • 151
    • 78650062951 scopus 로고    scopus 로고
    • EZH2-mediated epigenetic silencing in germinal center B cells contributes to proliferation and lymphomagenesis
    • Velichutina, I., Shaknovich, R., Geng, H., Johnson, N. A., Gascoyne, R. D., Melnick, A. M., et al. EZH2-mediated epigenetic silencing in germinal center B cells contributes to proliferation and lymphomagenesis. Blood, 2010, 116: 5247-5255, doi:10.1182/blood-2010-04-280149
    • (2010) Blood , vol.116 , pp. 5247-5255
    • Velichutina, I.1    Shaknovich, R.2    Geng, H.3    Johnson, N.A.4    Gascoyne, R.D.5    Melnick, A.M.6
  • 152
    • 84877815031 scopus 로고    scopus 로고
    • EZH2 is required for germinal center formation and somatic EZH2 mutations promote lymphoid transformation
    • Beguelin, W., Popovic, R., Teater, M., Jiang, Y., Bunting, K. L., Rosen, M., et al. EZH2 is required for germinal center formation and somatic EZH2 mutations promote lymphoid transformation. Cancer Cell, 2013, 23: 677-692, doi:10.1016/j.ccr.2013.04.011
    • (2013) Cancer Cell , vol.23 , pp. 677-692
    • Beguelin, W.1    Popovic, R.2    Teater, M.3    Jiang, Y.4    Bunting, K.L.5    Rosen, M.6
  • 153
    • 84877324084 scopus 로고    scopus 로고
    • Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2
    • Knutson, S. K., Warholic, N. M., Wigle, T. J., Klaus, C. R., Allain, C. J., Raimondi, A., et al. Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2. Proc Natl Acad Sci U S A, 2013, 110: 7922-7927, doi:10.1073/pnas.1303800110
    • (2013) Proc Natl Acad Sci U S A , vol.110 , pp. 7922-7927
    • Knutson, S.K.1    Warholic, N.M.2    Wigle, T.J.3    Klaus, C.R.4    Allain, C.J.5    Raimondi, A.6
  • 154
    • 84940093455 scopus 로고    scopus 로고
    • The Histone Methyltransferase Inhibitor A-366 Uncovers a Role for G9a/GLP in the Epigenetics of Leukemia
    • Pappano, W. N., Guo, J., He, Y., Ferguson, D., Jagadeeswaran, S., Osterling, D. J., et al. The Histone Methyltransferase Inhibitor A-366 Uncovers a Role for G9a/GLP in the Epigenetics of Leukemia. PLoS One, 2015, 10: e0131716, doi:10.1371/journal.pone.0131716
    • (2015) PLoS One , vol.10 , pp. e0131716
    • Pappano, W.N.1    Guo, J.2    He, Y.3    Ferguson, D.4    Jagadeeswaran, S.5    Osterling, D.J.6
  • 155
    • 84890834128 scopus 로고    scopus 로고
    • BIX-01294 induces autophagy-associated cell death via EHMT2/G9a dysfunction and intracellular reactive oxygen species production
    • Kim, Y., Kim, Y. S., Kim, D. E., Lee, J. S., Song, J. H., Kim, H. G., et al. BIX-01294 induces autophagy-associated cell death via EHMT2/G9a dysfunction and intracellular reactive oxygen species production. Autophagy, 2013, 9: 2126-2139, doi:10.4161/auto.26308
    • (2013) Autophagy , vol.9 , pp. 2126-2139
    • Kim, Y.1    Kim, Y.S.2    Kim, D.E.3    Lee, J.S.4    Song, J.H.5    Kim, H.G.6
  • 156
    • 84902087529 scopus 로고    scopus 로고
    • Euchromatic histone methyltransferase 2 inhibitor, BIX-01294, sensitizes human promyelocytic leukemia HL-60 and NB4 cells to growth inhibition and differentiation
    • Savickiene, J., Treigyte, G., Stirblyte, I., Valiuliene, G., Navakauskiene, R. Euchromatic histone methyltransferase 2 inhibitor, BIX-01294, sensitizes human promyelocytic leukemia HL-60 and NB4 cells to growth inhibition and differentiation. Leuk Res, 2014, 38: 822-829, doi:10.1016/j.leukres.2014.04.003
    • (2014) Leuk Res , vol.38 , pp. 822-829
    • Savickiene, J.1    Treigyte, G.2    Stirblyte, I.3    Valiuliene, G.4    Navakauskiene, R.5
  • 157
    • 84924060693 scopus 로고    scopus 로고
    • EHMT2 inhibitor BIX-01294 induces apoptosis through PMAIP1-USP9X-MCL1 axis in human bladder cancer cells
    • Cui, J., Sun, W., Hao, X., Wei, M., Su, X., Zhang, Y., et al. EHMT2 inhibitor BIX-01294 induces apoptosis through PMAIP1-USP9X-MCL1 axis in human bladder cancer cells. Cancer Cell Int, 2015, 15: 4, doi:10.1186/s12935-014-0149-x
    • (2015) Cancer Cell Int , vol.15 , pp. 4
    • Cui, J.1    Sun, W.2    Hao, X.3    Wei, M.4    Su, X.5    Zhang, Y.6
  • 158
    • 80052473151 scopus 로고    scopus 로고
    • Structural basis of substrate methylation and inhibition of SMYD2
    • Ferguson, A. D., Larsen, N. A., Howard, T., Pollard, H., Green, I., Grande, C., et al. Structural basis of substrate methylation and inhibition of SMYD2. Structure, 2011, 19: 1262-1273, doi:10.1016/j.str.2011.06.011
    • (2011) Structure , vol.19 , pp. 1262-1273
    • Ferguson, A.D.1    Larsen, N.A.2    Howard, T.3    Pollard, H.4    Green, I.5    Grande, C.6
  • 159
    • 84930225536 scopus 로고    scopus 로고
    • LLY-507, a Cell-active, Potent, and Selective Inhibitor of Protein-lysine Methyltransferase SMYD2
    • Nguyen, H., Allali-Hassani, A., Antonysamy, S., Chang, S., Chen, L. H., Curtis, C., et al. LLY-507, a Cell-active, Potent, and Selective Inhibitor of Protein-lysine Methyltransferase SMYD2. J Biol Chem, 2015, 290: 13641-13653, doi:10.1074/jbc.M114.626861
    • (2015) J Biol Chem , vol.290 , pp. 13641-13653
    • Nguyen, H.1    Allali-Hassani, A.2    Antonysamy, S.3    Chang, S.4    Chen, L.H.5    Curtis, C.6
  • 160
    • 34147173308 scopus 로고    scopus 로고
    • trans-2-Phenylcyclopropylamine is a mechanism-based inactivator of the histone demethylase LSD1
    • Schmidt, D. M., McCafferty, D. G. trans-2-Phenylcyclopropylamine is a mechanism-based inactivator of the histone demethylase LSD1. Biochemistry, 2007, 46: 4408-4416, doi:10.1021/bi0618621
    • (2007) Biochemistry , vol.46 , pp. 4408-4416
    • Schmidt, D.M.1    McCafferty, D.G.2
  • 161
    • 72249117352 scopus 로고    scopus 로고
    • Identification of cell-active lysine specific demethylase 1-selective inhibitors
    • Ueda, R., Suzuki, T., Mino, K., Tsumoto, H., Nakagawa, H., Hasegawa, M., et al. Identification of cell-active lysine specific demethylase 1-selective inhibitors. J Am Chem Soc, 2009, 131: 17536-17537, doi:10.1021/ja907055q
    • (2009) J Am Chem Soc , vol.131 , pp. 17536-17537
    • Ueda, R.1    Suzuki, T.2    Mino, K.3    Tsumoto, H.4    Nakagawa, H.5    Hasegawa, M.6
  • 162
    • 77955025931 scopus 로고    scopus 로고
    • Structurally designed trans-2-phenylcyclopropylamine derivatives potently inhibit histone demethylase LSD1/KDM1
    • Mimasu, S., Umezawa, N., Sato, S., Higuchi, T., Umehara, T., Yokoyama, S. Structurally designed trans-2-phenylcyclopropylamine derivatives potently inhibit histone demethylase LSD1/KDM1. Biochemistry, 2010, 49: 6494-6503, doi:10.1021/bi100299r
    • (2010) Biochemistry , vol.49 , pp. 6494-6503
    • Mimasu, S.1    Umezawa, N.2    Sato, S.3    Higuchi, T.4    Umehara, T.5    Yokoyama, S.6
  • 163
    • 79958247128 scopus 로고    scopus 로고
    • Enantioselective synthesis of tranylcypromine analogues as lysine demethylase (LSD1) inhibitors
    • Benelkebir, H., Hodgkinson, C., Duriez, P. J., Hayden, A. L., Bulleid, R. A., Crabb, S. J., et al. Enantioselective synthesis of tranylcypromine analogues as lysine demethylase (LSD1) inhibitors. Bioorg Med Chem, 2011, 19: 3709-3716, doi:10.1016/j.bmc.2011.02.017
    • (2011) Bioorg Med Chem , vol.19 , pp. 3709-3716
    • Benelkebir, H.1    Hodgkinson, C.2    Duriez, P.J.3    Hayden, A.L.4    Bulleid, R.A.5    Crabb, S.J.6
  • 164
    • 84859837026 scopus 로고    scopus 로고
    • The histone demethylase KDM1A sustains the oncogenic potential of MLL-AF9 leukemia stem cells
    • Harris, W. J., Huang, X., Lynch, J. T., Spencer, G. J., Hitchin, J. R., Li, Y., et al. The histone demethylase KDM1A sustains the oncogenic potential of MLL-AF9 leukemia stem cells. Cancer Cell, 2012, 21: 473-487, doi:10.1016/j.ccr.2012.03.014
    • (2012) Cancer Cell , vol.21 , pp. 473-487
    • Harris, W.J.1    Huang, X.2    Lynch, J.T.3    Spencer, G.J.4    Hitchin, J.R.5    Li, Y.6
  • 165
    • 84933501651 scopus 로고    scopus 로고
    • KDM1 histone lysine demethylases as targets for treatments of oncological and neurodegenerative disease
    • Maes, T., Mascaro, C., Ortega, A., Lunardi, S., Ciceri, F., Somervaille, T. C., et al. KDM1 histone lysine demethylases as targets for treatments of oncological and neurodegenerative disease. Epigenomics, 2015, 7: 609-626, doi:10.2217/epi.15.9
    • (2015) Epigenomics , vol.7 , pp. 609-626
    • Maes, T.1    Mascaro, C.2    Ortega, A.3    Lunardi, S.4    Ciceri, F.5    Somervaille, T.C.6
  • 166
    • 84937429405 scopus 로고    scopus 로고
    • A DNA Hypomethylation Signature Predicts Antitumor Activity of LSD1 Inhibitors in SCLC
    • Mohammad, H. P., Smitheman, K. N., Kamat, C. D., Soong, D., Federowicz, K. E., Van Aller, G. S., et al. A DNA Hypomethylation Signature Predicts Antitumor Activity of LSD1 Inhibitors in SCLC. Cancer Cell, 2015, 28: 57-69, doi:10.1016/j.ccell.2015.06.002
    • (2015) Cancer Cell , vol.28 , pp. 57-69
    • Mohammad, H.P.1    Smitheman, K.N.2    Kamat, C.D.3    Soong, D.4    Federowicz, K.E.5    Van Aller, G.S.6
  • 167
    • 84890448100 scopus 로고    scopus 로고
    • High-throughput virtual screening identifies novel N'-(1-phenylethylidene)-benzohydrazides as potent, specific, and reversible LSD1 inhibitors
    • Sorna, V., Theisen, E. R., Stephens, B., Warner, S. L., Bearss, D. J., Vankayalapati, H., et al. High-throughput virtual screening identifies novel N'-(1-phenylethylidene)-benzohydrazides as potent, specific, and reversible LSD1 inhibitors. J Med Chem, 2013, 56: 9496-9508, doi:10.1021/jm400870h
    • (2013) J Med Chem , vol.56 , pp. 9496-9508
    • Sorna, V.1    Theisen, E.R.2    Stephens, B.3    Warner, S.L.4    Bearss, D.J.5    Vankayalapati, H.6
  • 168
    • 84912072883 scopus 로고    scopus 로고
    • Development and classes of epigenetic drugs for cancer
    • Dhanak, D., Jackson, P. Development and classes of epigenetic drugs for cancer. Biochem Biophys Res Commun, 2014, 455: 58-69, doi:10.1016/j.bbrc.2014.07.006
    • (2014) Biochem Biophys Res Commun , vol.455 , pp. 58-69
    • Dhanak, D.1    Jackson, P.2
  • 169
    • 84922288173 scopus 로고    scopus 로고
    • Highly effective combination of LSD1 (KDM1A) antagonist and pan-histone deacetylase inhibitor against human AML cells
    • Fiskus, W., Sharma, S., Shah, B., Portier, B. P., Devaraj, S. G., Liu, K., et al. Highly effective combination of LSD1 (KDM1A) antagonist and pan-histone deacetylase inhibitor against human AML cells. Leukemia, 2014, 28: 2155-2164, doi:10.1038/leu.2014.119
    • (2014) Leukemia , vol.28 , pp. 2155-2164
    • Fiskus, W.1    Sharma, S.2    Shah, B.3    Portier, B.P.4    Devaraj, S.G.5    Liu, K.6
  • 170
    • 84866327150 scopus 로고    scopus 로고
    • Low molecular weight amidoximes that act as potent inhibitors of lysine-specific demethylase 1
    • Hazeldine, S., Pachaiyappan, B., Steinbergs, N., Nowotarski, S., Hanson, A. S., Casero, R. A., Jr., et al. Low molecular weight amidoximes that act as potent inhibitors of lysine-specific demethylase 1. J Med Chem, 2012, 55: 7378-7391, doi:10.1021/jm3002845
    • (2012) J Med Chem , vol.55 , pp. 7378-7391
    • Hazeldine, S.1    Pachaiyappan, B.2    Steinbergs, N.3    Nowotarski, S.4    Hanson, A.S.5    Casero, R.A.6
  • 171
    • 56749151276 scopus 로고    scopus 로고
    • Inhibitor scaffolds for 2-oxoglutarate-dependent histone lysine demethylases
    • Rose, N. R., Ng, S. S., Mecinovic, J., Lienard, B. M., Bello, S. H., Sun, Z., et al. Inhibitor scaffolds for 2-oxoglutarate-dependent histone lysine demethylases. J Med Chem, 2008, 51: 7053-7056, doi:10.1021/jm800936s
    • (2008) J Med Chem , vol.51 , pp. 7053-7056
    • Rose, N.R.1    Ng, S.S.2    Mecinovic, J.3    Lienard, B.M.4    Bello, S.H.5    Sun, Z.6
  • 172
    • 84865120905 scopus 로고    scopus 로고
    • A selective jumonji H3K27 demethylase inhibitor modulates the proinflammatory macrophage response
    • Kruidenier, L., Chung, C. W., Cheng, Z., Liddle, J., Che, K., Joberty, G., et al. A selective jumonji H3K27 demethylase inhibitor modulates the proinflammatory macrophage response. Nature, 2012, 488: 404-408, doi:10.1038/nature11262
    • (2012) Nature , vol.488 , pp. 404-408
    • Kruidenier, L.1    Chung, C.W.2    Cheng, Z.3    Liddle, J.4    Che, K.5    Joberty, G.6
  • 173
    • 84907526739 scopus 로고    scopus 로고
    • Contrasting roles of histone 3 lysine 27 demethylases in acute lymphoblastic leukaemia
    • Ntziachristos, P., Tsirigos, A., Welstead, G. G., Trimarchi, T., Bakogianni, S., Xu, L., et al. Contrasting roles of histone 3 lysine 27 demethylases in acute lymphoblastic leukaemia. Nature, 2014, 514: 513-517, doi:10.1038/nature13605
    • (2014) Nature , vol.514 , pp. 513-517
    • Ntziachristos, P.1    Tsirigos, A.2    Welstead, G.G.3    Trimarchi, T.4    Bakogianni, S.5    Xu, L.6
  • 174
    • 21044450362 scopus 로고    scopus 로고
    • MAP kinase-mediated phosphorylation of distinct pools of histone H3 at S10 or S28 via mitogen- and stress-activated kinase 1/2
    • Dyson, M. H., Thomson, S., Inagaki, M., Goto, H., Arthur, S. J., Nightingale, K., et al. MAP kinase-mediated phosphorylation of distinct pools of histone H3 at S10 or S28 via mitogen- and stress-activated kinase 1/2. J Cell Sci, 2005, 118: 2247-2259, doi:10.1242/jcs.02373
    • (2005) J Cell Sci , vol.118 , pp. 2247-2259
    • Dyson, M.H.1    Thomson, S.2    Inagaki, M.3    Goto, H.4    Arthur, S.J.5    Nightingale, K.6
  • 175
    • 21344450516 scopus 로고    scopus 로고
    • Phosphorylation of histone H3 at serine 10 is indispensable for neoplastic cell transformation
    • Choi, H. S., Choi, B. Y., Cho, Y. Y., Mizuno, H., Kang, B. S., Bode, A. M., et al. Phosphorylation of histone H3 at serine 10 is indispensable for neoplastic cell transformation. Cancer Res, 2005, 65: 5818-5827, doi:10.1158/0008-5472.CAN-05-0197
    • (2005) Cancer Res , vol.65 , pp. 5818-5827
    • Choi, H.S.1    Choi, B.Y.2    Cho, Y.Y.3    Mizuno, H.4    Kang, B.S.5    Bode, A.M.6
  • 176
    • 33744969113 scopus 로고    scopus 로고
    • The role of histone H3 phosphorylation (Ser10 and Ser28) in cell growth and cell transformation
    • Dong, Z., Bode, A. M. The role of histone H3 phosphorylation (Ser10 and Ser28) in cell growth and cell transformation. Mol Carcinog, 2006, 45: 416-421, doi:10.1002/mc.20220
    • (2006) Mol Carcinog , vol.45 , pp. 416-421
    • Dong, Z.1    Bode, A.M.2
  • 177
    • 58149280831 scopus 로고    scopus 로고
    • Genomic instability and histone H3 phosphorylation induction by the Ras-mitogen activated protein kinase pathway in pancreatic cancer cells
    • Espino, P. S., Pritchard, S., Heng, H. H., Davie, J. R. Genomic instability and histone H3 phosphorylation induction by the Ras-mitogen activated protein kinase pathway in pancreatic cancer cells. Int J Cancer, 2009, 124: 562-567, doi:10.1002/ijc.23959
    • (2009) Int J Cancer , vol.124 , pp. 562-567
    • Espino, P.S.1    Pritchard, S.2    Heng, H.H.3    Davie, J.R.4
  • 179
    • 84888195489 scopus 로고    scopus 로고
    • Histone 3 s10 phosphorylation: «caught in the R loop!»
    • Skourti-Stathaki, K., Proudfoot, N. J. Histone 3 s10 phosphorylation: «caught in the R loop!». Mol Cell, 2013, 52: 470-472, doi:10.1016/j.molcel.2013.11.006
    • (2013) Mol Cell , vol.52 , pp. 470-472
    • Skourti-Stathaki, K.1    Proudfoot, N.J.2
  • 180
    • 2342506598 scopus 로고    scopus 로고
    • TAF1 activates transcription by phosphorylation of serine 33 in histone H2B
    • Maile, T., Kwoczynski, S., Katzenberger, R. J., Wassarman, D. A., Sauer, F. TAF1 activates transcription by phosphorylation of serine 33 in histone H2B. Science, 2004, 304: 1010-1014, doi:10.1126/science.1095001
    • (2004) Science , vol.304 , pp. 1010-1014
    • Maile, T.1    Kwoczynski, S.2    Katzenberger, R.J.3    Wassarman, D.A.4    Sauer, F.5
  • 181
    • 37749026136 scopus 로고    scopus 로고
    • Phosphorylation of histone H3 at threonine 11 establishes a novel chromatin mark for transcriptional regulation
    • Metzger, E., Yin, N., Wissmann, M., Kunowska, N., Fischer, K., Friedrichs, N., et al. Phosphorylation of histone H3 at threonine 11 establishes a novel chromatin mark for transcriptional regulation. Nat Cell Biol, 2008, 10: 53-60, doi:10.1038/ncb1668
    • (2008) Nat Cell Biol , vol.10 , pp. 53-60
    • Metzger, E.1    Yin, N.2    Wissmann, M.3    Kunowska, N.4    Fischer, K.5    Friedrichs, N.6
  • 182
    • 58149242430 scopus 로고    scopus 로고
    • WSTF regulates the H2A.X DNA damage response via a novel tyrosine kinase activity
    • Xiao, A., Li, H., Shechter, D., Ahn, S. H., Fabrizio, L. A., Erdjument-Bromage, H., et al. WSTF regulates the H2A.X DNA damage response via a novel tyrosine kinase activity. Nature, 2009, 457: 57-62, doi:nature07668 [pii] 10.1038/nature07668
    • (2009) Nature , vol.457 , pp. 57-62
    • Xiao, A.1    Li, H.2    Shechter, D.3    Ahn, S.H.4    Fabrizio, L.A.5    Erdjument-Bromage, H.6
  • 183
    • 68249094946 scopus 로고    scopus 로고
    • Histone levels are regulated by phosphorylation and ubiquitylation-dependent proteolysis
    • Singh, R. K., Kabbaj, M. H., Paik, J., Gunjan, A. Histone levels are regulated by phosphorylation and ubiquitylation-dependent proteolysis. Nat Cell Biol, 2009, 11: 925-933, doi:ncb1903 [pii] 10.1038/ncb1903
    • (2009) Nat Cell Biol , vol.11 , pp. 925-933
    • Singh, R.K.1    Kabbaj, M.H.2    Paik, J.3    Gunjan, A.4
  • 184
    • 70349975711 scopus 로고    scopus 로고
    • JAK2 phosphorylates histone H3Y41 and excludes HP1alpha from chromatin
    • Dawson, M. A., Bannister, A. J., Gottgens, B., Foster, S. D., Bartke, T., Green, A. R., et al. JAK2 phosphorylates histone H3Y41 and excludes HP1alpha from chromatin. Nature, 2009, 461: 819-822, doi:nature08448 [pii] 10.1038/nature08448
    • (2009) Nature , vol.461 , pp. 819-822
    • Dawson, M.A.1    Bannister, A.J.2    Gottgens, B.3    Foster, S.D.4    Bartke, T.5    Green, A.R.6
  • 185
    • 84866112974 scopus 로고    scopus 로고
    • H2B Tyr37 phosphorylation suppresses expression of replication-dependent core histone genes
    • Mahajan, K., Fang, B., Koomen, J. M., Mahajan, N. P. H2B Tyr37 phosphorylation suppresses expression of replication-dependent core histone genes. Nat Struct Mol Biol, 2012, 19: 930-937, doi:10.1038/nsmb.2356
    • (2012) Nat Struct Mol Biol , vol.19 , pp. 930-937
    • Mahajan, K.1    Fang, B.2    Koomen, J.M.3    Mahajan, N.P.4
  • 186
    • 33751508862 scopus 로고    scopus 로고
    • Tyrosine phosphorylation controls PCNA function through protein stability
    • Wang, S. C., Nakajima, Y., Yu, Y. L., Xia, W., Chen, C. T., Yang, C. C., et al. Tyrosine phosphorylation controls PCNA function through protein stability. Nat Cell Biol, 2006, 8: 1359-1368, doi:10.1038/ncb1501
    • (2006) Nat Cell Biol , vol.8 , pp. 1359-1368
    • Wang, S.C.1    Nakajima, Y.2    Yu, Y.L.3    Xia, W.4    Chen, C.T.5    Yang, C.C.6
  • 187
    • 77958022249 scopus 로고    scopus 로고
    • RNA helicase A is a DNA-binding partner for EGFR-mediated transcriptional activation in the nucleus
    • Huo, L., Wang, Y. N., Xia, W., Hsu, S. C., Lai, C. C., Li, L. Y., et al. RNA helicase A is a DNA-binding partner for EGFR-mediated transcriptional activation in the nucleus. Proc Natl Acad Sci U S A, 2010, 107: 16125-16130, doi:10.1073/pnas.1000743107
    • (2010) Proc Natl Acad Sci U S A , vol.107 , pp. 16125-16130
    • Huo, L.1    Wang, Y.N.2    Xia, W.3    Hsu, S.C.4    Lai, C.C.5    Li, L.Y.6
  • 188
    • 84866128097 scopus 로고    scopus 로고
    • Nuclear EGFR suppresses ribonuclease activity of polynucleotide phosphorylase through DNAPK-mediated phosphorylation at serine 776
    • Yu, Y. L., Chou, R. H., Wu, C. H., Wang, Y. N., Chang, W. J., Tseng, Y. J., et al. Nuclear EGFR suppresses ribonuclease activity of polynucleotide phosphorylase through DNAPK-mediated phosphorylation at serine 776. J Biol Chem, 2012, 287: 31015-31026, doi:10.1074/jbc.M112.358077
    • (2012) J Biol Chem , vol.287 , pp. 31015-31026
    • Yu, Y.L.1    Chou, R.H.2    Wu, C.H.3    Wang, Y.N.4    Chang, W.J.5    Tseng, Y.J.6
  • 189
    • 78751558423 scopus 로고    scopus 로고
    • Targeting tyrosine phosphorylation of PCNA inhibits prostate cancer growth
    • Zhao, H., Lo, Y. H., Ma, L., Waltz, S. E., Gray, J. K., Hung, M. C., et al. Targeting tyrosine phosphorylation of PCNA inhibits prostate cancer growth. Mol Cancer Ther, 2011, 10: 29-36, doi:10.1158/1535-7163.MCT-10-0778
    • (2011) Mol Cancer Ther , vol.10 , pp. 29-36
    • Zhao, H.1    Lo, Y.H.2    Ma, L.3    Waltz, S.E.4    Gray, J.K.5    Hung, M.C.6
  • 190
    • 84875973325 scopus 로고    scopus 로고
    • Targeting the EGFR/PCNA signaling suppresses tumor growth of triple-negative breast cancer cells with cell-penetrating PCNA peptides
    • Yu, Y. L., Chou, R. H., Liang, J. H., Chang, W. J., Su, K. J., Tseng, Y. J., et al. Targeting the EGFR/PCNA signaling suppresses tumor growth of triple-negative breast cancer cells with cell-penetrating PCNA peptides. PLoS One, 2013, 8: e61362, doi:10.1371/journal.pone.0061362
    • (2013) PLoS One , vol.8 , pp. e61362
    • Yu, Y.L.1    Chou, R.H.2    Liang, J.H.3    Chang, W.J.4    Su, K.J.5    Tseng, Y.J.6
  • 191
    • 84897067023 scopus 로고    scopus 로고
    • Activation of mitogen- and stress-activated kinase 1 is required for proliferation of breast cancer cells in response to estrogens or progestins
    • Reyes, D., Ballare, C., Castellano, G., Soronellas, D., Bago, J. R., Blanco, J., et al. Activation of mitogen- and stress-activated kinase 1 is required for proliferation of breast cancer cells in response to estrogens or progestins. Oncogene, 2014, 33: 1570-1580, doi:10.1038/onc.2013.95
    • (2014) Oncogene , vol.33 , pp. 1570-1580
    • Reyes, D.1    Ballare, C.2    Castellano, G.3    Soronellas, D.4    Bago, J.R.5    Blanco, J.6
  • 192
    • 85019203234 scopus 로고    scopus 로고
    • Mitogen- and stress-activated Kinase 1 mediates Epstein-Barr virus latent membrane protein 1-promoted cell transformation in nasopharyngeal carcinoma through its induction of Fra-1 and c-Jun genes
    • Li, B., Wan, Z., Huang, G., Huang, Z., Zhang, X., Liao, D., et al. Mitogen- and stress-activated Kinase 1 mediates Epstein-Barr virus latent membrane protein 1-promoted cell transformation in nasopharyngeal carcinoma through its induction of Fra-1 and c-Jun genes. BMC Cancer, 2015, 15: 390, doi:10.1186/s12885-015-1398-3
    • (2015) BMC Cancer , vol.15 , pp. 390
    • Li, B.1    Wan, Z.2    Huang, G.3    Huang, Z.4    Zhang, X.5    Liao, D.6
  • 193
    • 84055223557 scopus 로고    scopus 로고
    • Characterization of the cellular action of the MSK inhibitor SB-747651A
    • Naqvi, S., Macdonald, A., McCoy, C. E., Darragh, J., Reith, A. D., Arthur, J. S. Characterization of the cellular action of the MSK inhibitor SB-747651A. Biochem J, 2012, 441: 347-357, doi:10.1042/BJ20110970
    • (2012) Biochem J , vol.441 , pp. 347-357
    • Naqvi, S.1    Macdonald, A.2    McCoy, C.E.3    Darragh, J.4    Reith, A.D.5    Arthur, J.S.6
  • 194
    • 84899048636 scopus 로고    scopus 로고
    • Inhibitors of NF-kappaB reverse cellular invasion and target gene upregulation in an experimental model of aggressive oral squamous cell carcinoma
    • Johnson, J., Shi, Z., Liu, Y., Stack, M. S. Inhibitors of NF-kappaB reverse cellular invasion and target gene upregulation in an experimental model of aggressive oral squamous cell carcinoma. Oral Oncol, 2014, 50: 468-477, doi:10.1016/j.oraloncology.2014.02.004
    • (2014) Oral Oncol , vol.50 , pp. 468-477
    • Johnson, J.1    Shi, Z.2    Liu, Y.3    Stack, M.S.4
  • 195
    • 38349053791 scopus 로고    scopus 로고
    • JAK2 inhibitor therapy in myeloproliferative disorders: rationale, preclinical studies and ongoing clinical trials
    • Pardanani, A. JAK2 inhibitor therapy in myeloproliferative disorders: rationale, preclinical studies and ongoing clinical trials. Leukemia, 2008, 22: 23-30, doi:10.1038/sj.leu.2404948
    • (2008) Leukemia , vol.22 , pp. 23-30
    • Pardanani, A.1
  • 196
    • 84937395764 scopus 로고    scopus 로고
    • Activity of the Type II JAK2 Inhibitor CHZ868 in B Cell Acute Lymphoblastic Leukemia
    • Wu, S. C., Li, L. S., Kopp, N., Montero, J., Chapuy, B., Yoda, A., et al. Activity of the Type II JAK2 Inhibitor CHZ868 in B Cell Acute Lymphoblastic Leukemia. Cancer Cell, 2015, 28: 29-41, doi:10.1016/j.ccell.2015.06.005
    • (2015) Cancer Cell , vol.28 , pp. 29-41
    • Wu, S.C.1    Li, L.S.2    Kopp, N.3    Montero, J.4    Chapuy, B.5    Yoda, A.6
  • 197
    • 84945218784 scopus 로고    scopus 로고
    • WEE1 Kinase As a Target for Cancer Therapy
    • Mueller, S., Haas-Kogan, D. A. WEE1 Kinase As a Target for Cancer Therapy. J Clin Oncol, 2015, doi:10.1200/JCO.2015.62.2290
    • J Clin Oncol , pp. 2015
    • Mueller, S.1    Haas-Kogan, D.A.2
  • 198
    • 0036202004 scopus 로고    scopus 로고
    • Wild-type TP53 inhibits G(2)-phase checkpoint abrogation and radiosensitization induced by PD0166285, a WEE1 kinase inhibitor
    • Li, J., Wang, Y., Sun, Y., Lawrence, T. S. Wild-type TP53 inhibits G(2)-phase checkpoint abrogation and radiosensitization induced by PD0166285, a WEE1 kinase inhibitor. Radiat Res, 2002, 157: 322-330
    • (2002) Radiat Res , vol.157 , pp. 322-330
    • Li, J.1    Wang, Y.2    Sun, Y.3    Lawrence, T.S.4
  • 199
    • 33846608696 scopus 로고    scopus 로고
    • Cell cycle regulation by the Wee1 inhibitor PD0166285, pyrido [2,3-d] pyimidine, in the B16 mouse melanoma cell line
    • Hashimoto, O., Shinkawa, M., Torimura, T., Nakamura, T., Selvendiran, K., Sakamoto, M., et al. Cell cycle regulation by the Wee1 inhibitor PD0166285, pyrido [2,3-d] pyimidine, in the B16 mouse melanoma cell line. BMC Cancer, 2006, 6: 292, doi:10.1186/1471-2407-6-292
    • (2006) BMC Cancer , vol.6 , pp. 292
    • Hashimoto, O.1    Shinkawa, M.2    Torimura, T.3    Nakamura, T.4    Selvendiran, K.5    Sakamoto, M.6
  • 200
    • 77953711939 scopus 로고    scopus 로고
    • MK-1775, a small molecule Wee1 inhibitor, enhances anti-tumor efficacy of various DNA-damaging agents, including 5-fluorouracil
    • Hirai, H., Arai, T., Okada, M., Nishibata, T., Kobayashi, M., Sakai, N., et al. MK-1775, a small molecule Wee1 inhibitor, enhances anti-tumor efficacy of various DNA-damaging agents, including 5-fluorouracil. Cancer Biol Ther, 2010, 9: 514-522
    • (2010) Cancer Biol Ther , vol.9 , pp. 514-522
    • Hirai, H.1    Arai, T.2    Okada, M.3    Nishibata, T.4    Kobayashi, M.5    Sakai, N.6
  • 201
    • 80052491760 scopus 로고    scopus 로고
    • MK-1775, a novel Wee1 kinase inhibitor, radiosensitizes p53-defective human tumor cells
    • Bridges, K. A., Hirai, H., Buser, C. A., Brooks, C., Liu, H., Buchholz, T. A., et al. MK-1775, a novel Wee1 kinase inhibitor, radiosensitizes p53-defective human tumor cells. Clin Cancer Res, 2011, 17: 5638-5648, doi:10.1158/1078-0432.CCR-11-0650
    • (2011) Clin Cancer Res , vol.17 , pp. 5638-5648
    • Bridges, K.A.1    Hirai, H.2    Buser, C.A.3    Brooks, C.4    Liu, H.5    Buchholz, T.A.6
  • 202
    • 79955492036 scopus 로고    scopus 로고
    • MK-1775, a potent Wee1 inhibitor, synergizes with gemcitabine to achieve tumor regressions, selectively in p53-deficient pancreatic cancer xenografts
    • Rajeshkumar, N. V., De Oliveira, E., Ottenhof, N., Watters, J., Brooks, D., Demuth, T., et al. MK-1775, a potent Wee1 inhibitor, synergizes with gemcitabine to achieve tumor regressions, selectively in p53-deficient pancreatic cancer xenografts. Clin Cancer Res, 2011, 17: 2799-2806, doi:10.1158/1078-0432.CCR-10-2580
    • (2011) Clin Cancer Res , vol.17 , pp. 2799-2806
    • Rajeshkumar, N.V.1    De Oliveira, E.2    Ottenhof, N.3    Watters, J.4    Brooks, D.5    Demuth, T.6
  • 203
    • 84855645086 scopus 로고    scopus 로고
    • MK1775, a selective Wee1 inhibitor, shows single-agent antitumor activity against sarcoma cells
    • Kreahling, J. M., Gemmer, J. Y., Reed, D., Letson, D., Bui, M., Altiok, S. MK1775, a selective Wee1 inhibitor, shows single-agent antitumor activity against sarcoma cells. Mol Cancer Ther, 2012, 11: 174-182, doi:10.1158/1535-7163.MCT-11-0529
    • (2012) Mol Cancer Ther , vol.11 , pp. 174-182
    • Kreahling, J.M.1    Gemmer, J.Y.2    Reed, D.3    Letson, D.4    Bui, M.5    Altiok, S.6
  • 204
    • 84882253419 scopus 로고    scopus 로고
    • Preclinical evaluation of the WEE1 inhibitor MK-1775 as single-agent anticancer therapy
    • Guertin, A. D., Li, J., Liu, Y., Hurd, M. S., Schuller, A. G., Long, B., et al. Preclinical evaluation of the WEE1 inhibitor MK-1775 as single-agent anticancer therapy. Mol Cancer Ther, 2013, 12: 1442-1452, doi:10.1158/1535-7163.MCT-13-0025
    • (2013) Mol Cancer Ther , vol.12 , pp. 1442-1452
    • Guertin, A.D.1    Li, J.2    Liu, Y.3    Hurd, M.S.4    Schuller, A.G.5    Long, B.6
  • 205
    • 84929279071 scopus 로고    scopus 로고
    • CHK1 plays a critical role in the anti-leukemic activity of the wee1 inhibitor MK-1775 in acute myeloid leukemia cells
    • Qi, W., Xie, C., Li, C., Caldwell, J. T., Edwards, H., Taub, J. W., et al. CHK1 plays a critical role in the anti-leukemic activity of the wee1 inhibitor MK-1775 in acute myeloid leukemia cells. J Hematol Oncol, 2014, 7: 53, doi:10.1186/s13045-014-0053-9
    • (2014) J Hematol Oncol , vol.7 , pp. 53
    • Qi, W.1    Xie, C.2    Li, C.3    Caldwell, J.T.4    Edwards, H.5    Taub, J.W.6
  • 206
    • 84945181744 scopus 로고    scopus 로고
    • Phase I Study of Single-Agent AZD1775 (MK-1775), a Wee1 Kinase Inhibitor, in Patients With Refractory Solid Tumors
    • Do, K., Wilsker, D., Ji, J., Zlott, J., Freshwater, T., Kinders, R. J., et al. Phase I Study of Single-Agent AZD1775 (MK-1775), a Wee1 Kinase Inhibitor, in Patients With Refractory Solid Tumors. J Clin Oncol, 2015, doi:10.1200/JCO.2014.60.4009
    • J Clin Oncol , pp. 2015
    • Do, K.1    Wilsker, D.2    Ji, J.3    Zlott, J.4    Freshwater, T.5    Kinders, R.J.6
  • 207
    • 84927639824 scopus 로고    scopus 로고
    • The Efficacy of the Wee1 Inhibitor MK-1775 Combined with Temozolomide Is Limited by Heterogeneous Distribution across the Blood-Brain Barrier in Glioblastoma
    • Pokorny, J. L., Calligaris, D., Gupta, S. K., Iyekegbe, D. O., Jr., Mueller, D., Bakken, K. K., et al. The Efficacy of the Wee1 Inhibitor MK-1775 Combined with Temozolomide Is Limited by Heterogeneous Distribution across the Blood-Brain Barrier in Glioblastoma. Clin Cancer Res, 2015, 21: 1916-1924, doi:10.1158/1078-0432.CCR-14-2588
    • (2015) Clin Cancer Res , vol.21 , pp. 1916-1924
    • Pokorny, J.L.1    Calligaris, D.2    Gupta, S.K.3    Iyekegbe, D.O.4    Mueller, D.5    Bakken, K.K.6
  • 208
    • 84927618199 scopus 로고    scopus 로고
    • A regimen combining the Wee1 inhibitor AZD1775 with HDAC inhibitors targets human acute myeloid leukemia cells harboring various genetic mutations
    • Zhou, L., Zhang, Y., Chen, S., Kmieciak, M., Leng, Y., Lin, H., et al. A regimen combining the Wee1 inhibitor AZD1775 with HDAC inhibitors targets human acute myeloid leukemia cells harboring various genetic mutations. Leukemia, 2015, 29: 807-818, doi:10.1038/leu.2014.296
    • (2015) Leukemia , vol.29 , pp. 807-818
    • Zhou, L.1    Zhang, Y.2    Chen, S.3    Kmieciak, M.4    Leng, Y.5    Lin, H.6
  • 209
    • 84880941947 scopus 로고    scopus 로고
    • miR-381, a novel intrinsic WEE1 inhibitor, sensitizes renal cancer cells to 5-FU by up-regulation of Cdc2 activities in 786-O
    • Chen, B., Duan, L., Yin, G., Tan, J., Jiang, X. miR-381, a novel intrinsic WEE1 inhibitor, sensitizes renal cancer cells to 5-FU by up-regulation of Cdc2 activities in 786-O. J Chemother, 2013, 25: 229-238, doi:10.1179/1973947813Y.0000000092
    • (2013) J Chemother , vol.25 , pp. 229-238
    • Chen, B.1    Duan, L.2    Yin, G.3    Tan, J.4    Jiang, X.5
  • 210
    • 10044232731 scopus 로고    scopus 로고
    • Prognostic significance of p53, Her-2, and EGFR overexpression in borderline and epithelial ovarian cancer
    • Nielsen, J. S., Jakobsen, E., Holund, B., Bertelsen, K., Jakobsen, A. Prognostic significance of p53, Her-2, and EGFR overexpression in borderline and epithelial ovarian cancer. Int J Gynecol Cancer, 2004, 14: 1086-1096, doi:10.1111/j.1048-891X.2004.14606.x
    • (2004) Int J Gynecol Cancer , vol.14 , pp. 1086-1096
    • Nielsen, J.S.1    Jakobsen, E.2    Holund, B.3    Bertelsen, K.4    Jakobsen, A.5
  • 211
    • 34249325876 scopus 로고    scopus 로고
    • Modeling breast cancer-associated c-Src and EGFR overexpression in human MECs: c-Src and EGFR cooperatively promote aberrant three-dimensional acinar structure and invasive behavior
    • Dimri, M., Naramura, M., Duan, L., Chen, J., Ortega-Cava, C., Chen, G., et al. Modeling breast cancer-associated c-Src and EGFR overexpression in human MECs: c-Src and EGFR cooperatively promote aberrant three-dimensional acinar structure and invasive behavior. Cancer Res, 2007, 67: 4164-4172, doi:10.1158/0008-5472.CAN-06-2580
    • (2007) Cancer Res , vol.67 , pp. 4164-4172
    • Dimri, M.1    Naramura, M.2    Duan, L.3    Chen, J.4    Ortega-Cava, C.5    Chen, G.6
  • 212
    • 35348848403 scopus 로고    scopus 로고
    • Prognostic significance of overexpression and phosphorylation of epidermal growth factor receptor (EGFR) and the presence of truncated EGFRvIII in locoregionally advanced breast cancer
    • Nieto, Y., Nawaz, F., Jones, R. B., Shpall, E. J., Nawaz, S. Prognostic significance of overexpression and phosphorylation of epidermal growth factor receptor (EGFR) and the presence of truncated EGFRvIII in locoregionally advanced breast cancer. J Clin Oncol, 2007, 25: 4405-4413, doi:10.1200/JCO.2006.09.8822
    • (2007) J Clin Oncol , vol.25 , pp. 4405-4413
    • Nieto, Y.1    Nawaz, F.2    Jones, R.B.3    Shpall, E.J.4    Nawaz, S.5
  • 213
    • 80053643861 scopus 로고    scopus 로고
    • Patterns of skin and soft tissue metastases from breast cancer according to subtypes: relationship between EGFR overexpression and skin manifestations
    • Kong, J. H., Park, Y. H., Kim, J. A., Kim, J. H., Yun, J., Sun, J. M., et al. Patterns of skin and soft tissue metastases from breast cancer according to subtypes: relationship between EGFR overexpression and skin manifestations. Oncology, 2011, 81: 55-62, doi:10.1159/000331417
    • (2011) Oncology , vol.81 , pp. 55-62
    • Kong, J.H.1    Park, Y.H.2    Kim, J.A.3    Kim, J.H.4    Yun, J.5    Sun, J.M.6
  • 214
    • 84860511478 scopus 로고    scopus 로고
    • EGFR overexpression relates to triple negative profile and poor prognosis in breast cancer patients in Tunisia
    • Kallel, I., Khabir, A., Boujelbene, N., Abdennadher, R., Daoud, J., Frikha, M., et al. EGFR overexpression relates to triple negative profile and poor prognosis in breast cancer patients in Tunisia. J Recept Signal Transduct Res, 2012, 32: 142-149, doi:10.3109/10799893.2012.664552
    • (2012) J Recept Signal Transduct Res , vol.32 , pp. 142-149
    • Kallel, I.1    Khabir, A.2    Boujelbene, N.3    Abdennadher, R.4    Daoud, J.5    Frikha, M.6
  • 215
    • 84877683942 scopus 로고    scopus 로고
    • Overexpression of epithelial growth factor receptor (EGFR) predicts better response to neo-adjuvant chemotherapy in patients with triple-negative breast cancer
    • Tang, Y., Zhu, L., Li, Y., Ji, J., Li, J., Yuan, F., et al. Overexpression of epithelial growth factor receptor (EGFR) predicts better response to neo-adjuvant chemotherapy in patients with triple-negative breast cancer. J Transl Med, 2012, 10 Suppl 1:S4, doi:10.1186/1479-5876-10-S1-S4
    • (2012) J Transl Med , pp. S4
    • Tang, Y.1    Zhu, L.2    Li, Y.3    Ji, J.4    Li, J.5    Yuan, F.6
  • 216
    • 38949101107 scopus 로고    scopus 로고
    • Expression signatures in lung cancer reveal a profile for EGFR-mutant tumours and identify selective PIK3CA overexpression by gene amplification
    • Angulo, B., Suarez-Gauthier, A., Lopez-Rios, F., Medina, P. P., Conde, E., Tang, M., et al. Expression signatures in lung cancer reveal a profile for EGFR-mutant tumours and identify selective PIK3CA overexpression by gene amplification. J Pathol, 2008, 214: 347-356, doi:10.1002/path.2267
    • (2008) J Pathol , vol.214 , pp. 347-356
    • Angulo, B.1    Suarez-Gauthier, A.2    Lopez-Rios, F.3    Medina, P.P.4    Conde, E.5    Tang, M.6
  • 217
    • 68949213744 scopus 로고    scopus 로고
    • Predictive value of EGFR and HER2 overexpression in advanced non-small-cell lung cancer
    • Hirsch, F. R., Varella-Garcia, M., Cappuzzo, F. Predictive value of EGFR and HER2 overexpression in advanced non-small-cell lung cancer. Oncogene, 2009, 28 Suppl 1:S32-37, doi:10.1038/onc.2009.199
    • (2009) Oncogene , vol.28 , pp. S32-S37
    • Hirsch, F.R.1    Varella-Garcia, M.2    Cappuzzo, F.3
  • 218
    • 77349083210 scopus 로고    scopus 로고
    • Overexpression of EGFR pathway-related genes in the circulation is highly correlated with EGFR mutations and overexpression in paired cancer tissue from patients with non-small cell lung cancer
    • Chen, C. C., Chiu, H. H., Yen, L. C., Chang, H. J., Chang, M. S., Tsai, J. R., et al. Overexpression of EGFR pathway-related genes in the circulation is highly correlated with EGFR mutations and overexpression in paired cancer tissue from patients with non-small cell lung cancer. Oncol Rep, 2010, 23: 639-645
    • (2010) Oncol Rep , vol.23 , pp. 639-645
    • Chen, C.C.1    Chiu, H.H.2    Yen, L.C.3    Chang, H.J.4    Chang, M.S.5    Tsai, J.R.6
  • 219
    • 84905378090 scopus 로고    scopus 로고
    • EGFR overexpression in canine primary lung cancer: pathogenetic implications and impact on survival
    • Sabattini, S., Mancini, F. R., Marconato, L., Bacci, B., Rossi, F., Vignoli, M., et al. EGFR overexpression in canine primary lung cancer: pathogenetic implications and impact on survival. Vet Comp Oncol, 2014, 12: 237-248, doi:10.1111/vco.12002
    • (2014) Vet Comp Oncol , vol.12 , pp. 237-248
    • Sabattini, S.1    Mancini, F.R.2    Marconato, L.3    Bacci, B.4    Rossi, F.5    Vignoli, M.6
  • 220
    • 84655170171 scopus 로고    scopus 로고
    • Overexpression of SGLT1 and EGFR in colorectal cancer showing a correlation with the prognosis
    • Guo, G. F., Cai, Y. C., Zhang, B., Xu, R. H., Qiu, H. J., Xia, L. P., et al. Overexpression of SGLT1 and EGFR in colorectal cancer showing a correlation with the prognosis. Med Oncol, 2011, 28 Suppl 1:S197-203, doi:10.1007/s12032-010-9696-8
    • (2011) Med Oncol , vol.28 , pp. S197-S203
    • Guo, G.F.1    Cai, Y.C.2    Zhang, B.3    Xu, R.H.4    Qiu, H.J.5    Xia, L.P.6
  • 221
    • 0038545803 scopus 로고    scopus 로고
    • Erlotinib (Tarceva): a promising drug targeting epidermal growth factor receptor tyrosine kinase
    • Bulgaru, A. M., Mani, S., Goel, S., Perez-Soler, R. Erlotinib (Tarceva): a promising drug targeting epidermal growth factor receptor tyrosine kinase. Expert Rev Anticancer Ther, 2003, 3: 269-279, doi:10.1586/14737140.3.3.269
    • (2003) Expert Rev Anticancer Ther , vol.3 , pp. 269-279
    • Bulgaru, A.M.1    Mani, S.2    Goel, S.3    Perez-Soler, R.4
  • 222
    • 0346991841 scopus 로고    scopus 로고
    • EGFR tyrosine kinase inhibitor «gefitinib (Iressa)» for cancer therapy
    • Yano, S., Yamaguchi, M., Dong, R. P. EGFR tyrosine kinase inhibitor «gefitinib (Iressa)» for cancer therapy. Nihon Yakurigaku Zasshi, 2003, 122: 491-497
    • (2003) Nihon Yakurigaku Zasshi , vol.122 , pp. 491-497
    • Yano, S.1    Yamaguchi, M.2    Dong, R.P.3
  • 223
    • 10444267243 scopus 로고    scopus 로고
    • Cetuximab (Erbitux)--an emerging targeted therapy for epidermal growth factor receptor-expressing tumours
    • Ng, M., Cunningham, D. Cetuximab (Erbitux)--an emerging targeted therapy for epidermal growth factor receptor-expressing tumours. Int J Clin Pract, 2004, 58: 970-976
    • (2004) Int J Clin Pract , vol.58 , pp. 970-976
    • Ng, M.1    Cunningham, D.2
  • 224
    • 33747146789 scopus 로고    scopus 로고
    • Lapatinib: a novel EGFR/HER2 tyrosine kinase inhibitor for cancer
    • Johnston, S. R., Leary, A. Lapatinib: a novel EGFR/HER2 tyrosine kinase inhibitor for cancer. Drugs Today (Barc), 2006, 42: 441-453, doi:10.1358/dot.2006.42.7.985637
    • (2006) Drugs Today (Barc) , vol.42 , pp. 441-453
    • Johnston, S.R.1    Leary, A.2
  • 225
    • 84887326965 scopus 로고    scopus 로고
    • Lapatinib-induced NF-kappaB activation sensitizes triple-negative breast cancer cells to proteasome inhibitors
    • Chen, Y. J., Yeh, M. H., Yu, M. C., Wei, Y. L., Chen, W. S., Chen, J. Y., et al. Lapatinib-induced NF-kappaB activation sensitizes triple-negative breast cancer cells to proteasome inhibitors. Breast Cancer Res, 2013, 15: R108, doi:10.1186/bcr3575
    • (2013) Breast Cancer Res , vol.15 , pp. R108
    • Chen, Y.J.1    Yeh, M.H.2    Yu, M.C.3    Wei, Y.L.4    Chen, W.S.5    Chen, J.Y.6
  • 226
    • 84875408831 scopus 로고    scopus 로고
    • Lapatinib-mediated cyclooxygenase-2 expression via epidermal growth factor receptor/HuR interaction enhances the aggressiveness of triple-negative breast cancer cells
    • Hsia, T. C., Tu, C. Y., Chen, Y. J., Wei, Y. L., Yu, M. C., Hsu, S. C., et al. Lapatinib-mediated cyclooxygenase-2 expression via epidermal growth factor receptor/HuR interaction enhances the aggressiveness of triple-negative breast cancer cells. Mol Pharmacol, 2013, 83: 857-869, doi:10.1124/mol.112.082743
    • (2013) Mol Pharmacol , vol.83 , pp. 857-869
    • Hsia, T.C.1    Tu, C.Y.2    Chen, Y.J.3    Wei, Y.L.4    Yu, M.C.5    Hsu, S.C.6
  • 227
    • 33845674883 scopus 로고    scopus 로고
    • Panitumumab the first fully human monoclonal antibody: from the bench to the clinic
    • Cohenuram, M., Saif, M. W. Panitumumab the first fully human monoclonal antibody: from the bench to the clinic. Anticancer Drugs, 2007, 18: 7-15, doi:10.1097/CAD.0b013e32800feecb
    • (2007) Anticancer Drugs , vol.18 , pp. 7-15
    • Cohenuram, M.1    Saif, M.W.2
  • 228
    • 84962205018 scopus 로고    scopus 로고
    • ASPP and iASPP: Implication in cancer development and progression
    • Li, Y., Ahmad, A., Sarkar and F. H., ASPP and iASPP: Implication in cancer development and progression. Cell. Mol. Biol. 2015, 61(6): 2-8.
    • (2015) Cell. Mol. Biol. , vol.61 , Issue.6 , pp. 2-8
    • Li, Y.1    Ahmad, A.2    Sarkar, F.H.3
  • 229
    • 84962175330 scopus 로고    scopus 로고
    • Association of NOTCH with different microRNAs in head and neck cancer
    • Masood, N., Qureshi, M. Z. and Yasmin, A., Association of NOTCH with different microRNAs in head and neck cancer. Cell. Mol. Biol. 2015, 61(6): 9-16.
    • (2015) Cell. Mol. Biol. , vol.61 , Issue.6 , pp. 9-16
    • Masood, N.1    Qureshi, M.Z.2    Yasmin, A.3
  • 231
    • 84962167853 scopus 로고    scopus 로고
    • The Notch signaling pathway in esophageal adenocarcinoma
    • Wang, Z., Chen, J. and Capobianco, A. J., The Notch signaling pathway in esophageal adenocarcinoma. Cell. Mol. Biol. 2015, 61(6): 24-32.
    • (2015) Cell. Mol. Biol. , vol.61 , Issue.6 , pp. 24-32
    • Wang, Z.1    Chen, J.2    Capobianco, A.J.3
  • 232
    • 84962191062 scopus 로고    scopus 로고
    • TRAIL and targeting cancer cells: between promises and obstacles
    • Limami, Y., Pinon, A., Riaz, A. and Simon, A., TRAIL and targeting cancer cells: between promises and obstacles. Cell. Mol. Biol. 2015, 61(6): 33-38.
    • (2015) Cell. Mol. Biol. , vol.61 , Issue.6 , pp. 33-38
    • Limami, Y.1    Pinon, A.2    Riaz, A.3    Simon, A.4
  • 236
  • 237
    • 84962141567 scopus 로고    scopus 로고
    • Hesperetin exerts apoptotic effect on A431 skin carcinoma cells by regulating mitogen activated protein kinases and cyclins
    • Smina, T. P., Mohan, A., Ayyappa, K. A., Sethuraman, S. and Krishnan, U. M., Hesperetin exerts apoptotic effect on A431 skin carcinoma cells by regulating mitogen activated protein kinases and cyclins. Cell. Mol. Biol. 2015, 61(6): 92-99.
    • (2015) Cell. Mol. Biol. , vol.61 , Issue.6 , pp. 92-99
    • Smina, T.P.1    Mohan, A.2    Ayyappa, K.A.3    Sethuraman, S.4    Krishnan, U.M.5
  • 238
    • 84962168850 scopus 로고    scopus 로고
    • Indirect role of microRNAs and transcription factors in the regulation of important cancer genes: A network biology approach
    • Ahmadi, M., Jafari, R., Marashi, S. A. and Farazmand, A., Indirect role of microRNAs and transcription factors in the regulation of important cancer genes: A network biology approach. Cell. Mol. Biol. 2015, 61(6): 100-107.
    • (2015) Cell. Mol. Biol. , vol.61 , Issue.6 , pp. 100-107
    • Ahmadi, M.1    Jafari, R.2    Marashi, S.A.3    Farazmand, A.4
  • 239
    • 84962165962 scopus 로고    scopus 로고
    • Genetic variants in the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and death receptor (DR4) genes contribute to susceptibility to colorectal cancer in pakistani population
    • Zahoor, A., Mansoor, Q., Farooqi, A. A., Fayyaz, S., Naz, G. and Ismail, M., Genetic variants in the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and death receptor (DR4) genes contribute to susceptibility to colorectal cancer in pakistani population. Cell. Mol. Biol. 2015, 61(6): 108-112.
    • (2015) Cell. Mol. Biol. , vol.61 , Issue.6 , pp. 108-112
    • Zahoor, A.1    Mansoor, Q.2    Farooqi, A.A.3    Fayyaz, S.4    Naz, G.5    Ismail, M.6


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.