메뉴 건너뛰기




Volumn 13, Issue 8, 2012, Pages 1029-1047

Discovery and mechanism of natural products as modulators of histone acetylation

Author keywords

Histone acetylation; Histone acetyltransferases; Histone deacetylases; Natural products; Secondary metabolites

Indexed keywords

ANACARDIC ACID; APICIDIN; AZACITIDINE; AZUMAMIDE A; AZUMAMIDE B; AZUMAMIDE C; AZUMAMIDE E; CURCUMIN; DEPUDECIN; FR 235222; FR 901375; GARCINOL; GUTTIFERONE A; GUTTIFERONES E; HISTONE DEACETYLASE INHIBITOR; ISOGARCINOL; LARGAZOLE; NATURAL PRODUCT; NEMOROSONE; POLYPHENOL; PSAMMAPLIN A; ROMIDEPSIN; SPIRUCHOSTATIN A; SPIRUCHOSTATIN B; TETRAPEPTIDE; TRAPOXIN A; TRAPOXIN B; TRICHOSTATIN A; UNCLASSIFIED DRUG; UNINDEXED DRUG; VORINOSTAT;

EID: 84863548736     PISSN: 13894501     EISSN: 18735592     Source Type: Journal    
DOI: 10.2174/138945012802008973     Document Type: Article
Times cited : (24)

References (172)
  • 1
    • 2942565810 scopus 로고    scopus 로고
    • Natural products and combinatorial chemistry: Back to the future
    • Ortholand J, Ganesan A. Natural products and combinatorial chemistry: back to the future. Curr Opin Chem Biol 2004; 8: 271-80.
    • (2004) Curr Opin Chem Biol , vol.8 , pp. 271-280
    • Ortholand, J.1    Ganesan, A.2
  • 2
    • 14944383798 scopus 로고    scopus 로고
    • The evolving role of natural products in drug discovery
    • Koehn FE, Carter GT. The evolving role of natural products in drug discovery. Nat Rev Drug Discov 2005; 4: 206-20.
    • (2005) Nat Rev Drug Discov , vol.4 , pp. 206-220
    • Koehn, F.E.1    Carter, G.T.2
  • 3
    • 84858308226 scopus 로고    scopus 로고
    • Natural products as sources of new drugs over the 30 years from 1981 to 2010
    • Newman DJ, Cragg GM. Natural products as sources of new drugs over the 30 years from 1981 to 2010. J Nat Prod 2012; 75: 311-35.
    • (2012) J Nat Prod , vol.75 , pp. 311-335
    • Newman, D.J.1    Cragg, G.M.2
  • 4
    • 0034614637 scopus 로고    scopus 로고
    • The hallmarks of cancer
    • Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000; 100: 57-70.
    • (2000) Cell , vol.100 , pp. 57-70
    • Hanahan, D.1    Weinberg, R.A.2
  • 5
    • 79952284127 scopus 로고    scopus 로고
    • Hallmarks of cancer: The next generation
    • Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011; 144: 646-74.
    • (2011) Cell , vol.144 , pp. 646-674
    • Hanahan, D.1    Weinberg, R.A.2
  • 6
    • 33847065486 scopus 로고    scopus 로고
    • The epigenomics of cancer
    • Jones PA, Baylin SB. The epigenomics of cancer. Cell 2007; 128: 683-92.
    • (2007) Cell , vol.128 , pp. 683-692
    • Jones, P.A.1    Baylin, S.B.2
  • 8
    • 33847070442 scopus 로고    scopus 로고
    • The role of chromatin during transcription
    • Li B, Carey M, Workman JL. The role of chromatin during transcription. Cell 2007; 128: 707-19.
    • (2007) Cell , vol.128 , pp. 707-719
    • Li, B.1    Carey, M.2    Workman, J.L.3
  • 9
    • 1842411320 scopus 로고    scopus 로고
    • Crystal structure of the nucleosome core particle at 2.8Å resolution
    • Luger K, Mader AW, Richmond RK, Sargent DF, Richmond TJ. Crystal structure of the nucleosome core particle at 2.8Å resolution. Nature 1997; 389: 251-60.
    • (1997) Nature , vol.389 , pp. 251-260
    • Luger, K.1    Mader, A.W.2    Richmond, R.K.3    Sargent, D.F.4    Richmond, T.J.5
  • 10
    • 0033529565 scopus 로고    scopus 로고
    • Twenty-five years of the nucleosome, fundamental particle of the eukaryote chromosome
    • Kornberg RD, Lorch Y. Twenty-five years of the nucleosome, fundamental particle of the eukaryote chromosome. Cell 1999; 98: 285-94.
    • (1999) Cell , vol.98 , pp. 285-294
    • Kornberg, R.D.1    Lorch, Y.2
  • 11
    • 33847076849 scopus 로고    scopus 로고
    • Chromatin modifications and their function
    • Kouzarides T. Chromatin modifications and their function. Cell 2007; 128: 693-705.
    • (2007) Cell , vol.128 , pp. 693-705
    • Kouzarides, T.1
  • 12
    • 77956153243 scopus 로고    scopus 로고
    • The language of histone crosstalk
    • Lee J, Smith E, Shilatifard A. The language of histone crosstalk. Cell 2010; 142: 682-5.
    • (2010) Cell , vol.142 , pp. 682-685
    • Lee, J.1    Smith, E.2    Shilatifard, A.3
  • 13
    • 78751491475 scopus 로고    scopus 로고
    • Dynamic interplay between histone H3 modifications and protein interpreters: Emerging evidence for a histone language
    • Oliver SS, Denu JM. Dynamic interplay between histone H3 modifications and protein interpreters: Emerging evidence for a "histone language". Chem Bio Chem 2011; 12: 299-307.
    • (2011) Chem Bio Chem , vol.12 , pp. 299-307
    • Oliver, S.S.1    Denu, J.M.2
  • 14
    • 68949212379 scopus 로고    scopus 로고
    • Lysine acetylation targets protein complexes and co-regulates major cellular functions
    • Choudhary C, Kumar C, Gnad F, et al. Lysine acetylation targets protein complexes and co-regulates major cellular functions. Science 2009; 325: 834-40.
    • (2009) Science , vol.325 , pp. 834-840
    • Choudhary, C.1    Kumar, C.2    Gnad, F.3
  • 15
    • 77149148756 scopus 로고    scopus 로고
    • Regulation of cellular metabolism by protein lysine acetylation
    • Zhao S, Xu W, Wenqing J, et al. Regulation of cellular metabolism by protein lysine acetylation. Science 2010; 327: 1000-4.
    • (2010) Science , vol.327 , pp. 1000-1004
    • Zhao, S.1    Xu, W.2    Wenqing, J.3
  • 16
    • 1842578986 scopus 로고    scopus 로고
    • Molecular evolution of the histone deacetylase family: Functional implications of phylogenetic analysis
    • Gregoretti IV, Lee Y, Goodson HV. Molecular evolution of the histone deacetylase family: Functional implications of phylogenetic analysis. J Mol Biol 2004; 338: 17-31.
    • (2004) J Mol Biol , vol.338 , pp. 17-31
    • Gregoretti, I.V.1    Lee, Y.2    Goodson, H.V.3
  • 18
    • 65449166411 scopus 로고    scopus 로고
    • Novel structural insights into class I and II histone deacetylases
    • Ficner R. Novel structural insights into class I and II histone deacetylases. Curr Top Med Chem 2009; 9: 235-240.
    • (2009) Curr Top Med Chem , vol.9 , pp. 235-240
    • Ficner, R.1
  • 19
    • 36849004821 scopus 로고    scopus 로고
    • Unraveling the hidden catalytic activity of vertebrate class IIa histone deacetylases
    • Lahm A, Paolini C, Pallaoro M, et al. Unraveling the hidden catalytic activity of vertebrate class IIa histone deacetylases. Proc Natl Acad Sci USA 2007; 104: 17335-40.
    • (2007) Proc Natl Acad Sci USA , vol.104 , pp. 17335-17340
    • Lahm, A.1    Paolini, C.2    Pallaoro, M.3
  • 20
    • 3142562372 scopus 로고    scopus 로고
    • Structural snapshots of human HDAC8 provide insights into the class I histone deacetylases
    • Somoza JR, Skene RJ, Katz BA, et al. Structural snapshots of human HDAC8 provide insights into the class I histone deacetylases. Structure 2004; 12: 1325-34.
    • (2004) Structure , vol.12 , pp. 1325-1334
    • Somoza, J.R.1    Skene, R.J.2    Katz, B.A.3
  • 21
    • 6344222799 scopus 로고    scopus 로고
    • Crystal structure of a eukaryotic zinc-dependent histone deacetylase, human HDAC8, complexed with a hydroxamic acid inhibitor
    • Vannini A, Volpari C, Filocamo G, et al. Crystal structure of a eukaryotic zinc-dependent histone deacetylase, human HDAC8, complexed with a hydroxamic acid inhibitor. Proc Natl Acad Sci USA 2004; 101: 15064-9.
    • (2004) Proc Natl Acad Sci USA , vol.101 , pp. 15064-15069
    • Vannini, A.1    Volpari, C.2    Filocamo, G.3
  • 22
    • 45549095066 scopus 로고    scopus 로고
    • Human HDAC7 harbors a class IIa histone deacetylase-specific zinc binding motif and cryptic deacetylase activity
    • Schuetz A, Min J, Allali-Hassani A, et al. Human HDAC7 harbors a class IIa histone deacetylase-specific zinc binding motif and cryptic deacetylase activity. J Biol Chem 2008; 283: 11355-63.
    • (2008) J Biol Chem , vol.283 , pp. 11355-11363
    • Schuetz, A.1    Min, J.2    Allali-Hassani, A.3
  • 23
    • 80051599485 scopus 로고    scopus 로고
    • Structural basis of the antiproliferative activity of largazole, a depsipeptide inhibitor of the histone deacetylases
    • Cole KE, Dowling DP, Boone MA, Phillips AJ, Christianson DW. Structural basis of the antiproliferative activity of largazole, a depsipeptide inhibitor of the histone deacetylases. J Am Chem Soc 2011; 133: 12474-7.
    • (2011) J Am Chem Soc , vol.133 , pp. 12474-12477
    • Cole, K.E.1    Dowling, D.P.2    Boone, M.A.3    Phillips, A.J.4    Christianson, D.W.5
  • 24
    • 58149144730 scopus 로고    scopus 로고
    • Structural studies of human histone deacetylase 8 and its sitespecific variants complexed with substrate and inhibitors
    • Dowling DP, Gantt SL, Gattis SG, Fierke CA, Christianson DW. Structural studies of human histone deacetylase 8 and its sitespecific variants complexed with substrate and inhibitors. Biochemistry 2008; 47: 13554-63.
    • (2008) Biochemistry , vol.47 , pp. 13554-13563
    • Dowling, D.P.1    Gantt, S.L.2    Gattis, S.G.3    Fierke, C.A.4    Christianson, D.W.5
  • 25
    • 33748451151 scopus 로고    scopus 로고
    • Anticancer activities of histone deacetylase inhibitors
    • Bolden J, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Dis 2006; 5: 769-84.
    • (2006) Nat Rev Drug Dis , vol.5 , pp. 769-784
    • Bolden, J.1    Peart, M.J.2    Johnstone, R.W.3
  • 26
    • 80054078476 scopus 로고    scopus 로고
    • Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega
    • Sievers F, Wilm A, Dineen DG, et al. Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega. Mol Syst Biol 2011; 7: 539.
    • (2011) Mol Syst Biol , vol.7 , pp. 539
    • Sievers, F.1    Wilm, A.2    Dineen, D.G.3
  • 27
    • 0042594474 scopus 로고    scopus 로고
    • SATCHMO: Sequence alignment and tree construction using Hidden Markov Models
    • Edgar RC, Sjolander K. SATCHMO: Sequence alignment and tree construction using Hidden Markov Models. Bioinformatics 2003; 19: 1404-11.
    • (2003) Bioinformatics , vol.19 , pp. 1404-1411
    • Edgar, R.C.1    Sjolander, K.2
  • 28
    • 59349115177 scopus 로고    scopus 로고
    • Chemical mechanisms of histone lysine and arginine modifications
    • Smith BC, Denu JM. Chemical mechanisms of histone lysine and arginine modifications. Biochim Biophys Acta 2009; 1789: 45-57.
    • (2009) Biochim Biophys Acta , vol.1789 , pp. 45-57
    • Smith, B.C.1    Denu, J.M.2
  • 29
    • 34547911052 scopus 로고    scopus 로고
    • Chemistry of acetyl transfer by histone modifying enzymes: Structure, mechanism and implications for effector design
    • Hodawadekar SC, Marmorstein R. Chemistry of acetyl transfer by histone modifying enzymes: structure, mechanism and implications for effector design. Oncogene 2007; 26: 5528-40.
    • (2007) Oncogene , vol.26 , pp. 5528-5540
    • Hodawadekar, S.C.1    Marmorstein, R.2
  • 30
    • 33846374117 scopus 로고    scopus 로고
    • Catalytic mechanism of a MYST family histone acetyltransferase
    • Berndsen CE, Albaugh BN, Tan S, Denu JM. Catalytic mechanism of a MYST family histone acetyltransferase. Biochemistry 2007; 46: 623-9.
    • (2007) Biochemistry , vol.46 , pp. 623-629
    • Berndsen, C.E.1    Albaugh, B.N.2    Tan, S.3    Denu, J.M.4
  • 31
    • 0033603555 scopus 로고    scopus 로고
    • Catalytic mechanism and function of invariant glutamic acid 173 from the histone acetyltransferase GCN5 transcriptional coactivator
    • Tanner KG, Trievel RC, Kuoi M, et al. Catalytic mechanism and function of invariant glutamic acid 173 from the histone acetyltransferase GCN5 transcriptional coactivator. J Biol Chem 1999; 274: 18157-60.
    • (1999) J Biol Chem , vol.274 , pp. 18157-18160
    • Tanner, K.G.1    Trievel, R.C.2    Kuoi, M.3
  • 32
    • 0033168497 scopus 로고    scopus 로고
    • Solution structure of the catalytic domain of GCN5 histone acetyltransferase bound to coenzyme A
    • Lin Y, Fletcher CM, Zhou J, Allis CD, Wagner G. Solution structure of the catalytic domain of GCN5 histone acetyltransferase bound to coenzyme A. Nature 1999; 400: 86-9.
    • (1999) Nature , vol.400 , pp. 86-89
    • Lin, Y.1    Fletcher, C.M.2    Zhou, J.3    Allis, C.D.4    Wagner, G.5
  • 33
    • 39149109887 scopus 로고    scopus 로고
    • The structural basis of protein acetylation by the p300/CBP transcriptional coactivator
    • Liu X, Wang L, Zhao K, et al. The structural basis of protein acetylation by the p300/CBP transcriptional coactivator. Nature 2008; 451: 846-50.
    • (2008) Nature , vol.451 , pp. 846-850
    • Liu, X.1    Wang, L.2    Zhao, K.3
  • 34
    • 2442454683 scopus 로고    scopus 로고
    • Distinct localization of histone H3 acetylation and H3-K4 methylation to the transcription start sites in the human genome
    • Liang G, Lin JCY, Wei V, et al. Distinct localization of histone H3 acetylation and H3-K4 methylation to the transcription start sites in the human genome. Proc Natl Acad Sci USA 2004; 101: 7357-62.
    • (2004) Proc Natl Acad Sci USA , vol.101 , pp. 7357-7362
    • Liang, G.1    Lin, J.C.Y.2    Wei, V.3
  • 35
    • 20144388146 scopus 로고    scopus 로고
    • Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer
    • Fraga MF, Ballestar E, Villar-Garea A, et al. Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer. Nature Genet 2005; 37: 391-400.
    • (2005) Nature Genet , vol.37 , pp. 391-400
    • Fraga, M.F.1    Ballestar, E.2    Villar-Garea, A.3
  • 36
    • 21744457108 scopus 로고    scopus 로고
    • Global histone modification patterns predict risk of prostate cancer recurrence
    • Seligson DB, Horvath S, Shi T, et al. Global histone modification patterns predict risk of prostate cancer recurrence. Nature 2005; 435: 1262-6.
    • (2005) Nature , vol.435 , pp. 1262-1266
    • Seligson, D.B.1    Horvath, S.2    Shi, T.3
  • 37
    • 38449100788 scopus 로고    scopus 로고
    • Expression profile of class I histone deacetylases in human cancer tissues
    • Nakagawa M, Oda Y, Eguchi T, et al. Expression profile of class I histone deacetylases in human cancer tissues. Oncol Rep 2007; 18: 769-74.
    • (2007) Oncol Rep , vol.18 , pp. 769-774
    • Nakagawa, M.1    Oda, Y.2    Eguchi, T.3
  • 38
    • 1842631408 scopus 로고    scopus 로고
    • Upregulation and nuclear recruitment of HDAC1 in hormone refractory prostate cancer
    • Halkidou K, Gaughan L, Cook S, Leung HY, Neal DE, Robson CN. Upregulation and nuclear recruitment of HDAC1 in hormone refractory prostate cancer. Prostate 2004; 59: 177-89.
    • (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
  • 39
    • 21044447215 scopus 로고    scopus 로고
    • Increased expression of histone deacetylase 2 is found in human gastric cancer
    • Song J, Noh JH, Lee JH, et al. Increased expression of histone deacetylase 2 is found in human gastric cancer. Acta Path Micro Im 2005; 113: 264-8.
    • (2005) Acta Path Micro Im , vol.113 , pp. 264-268
    • Song, J.1    Noh, J.H.2    Lee, J.H.3
  • 40
    • 0023927992 scopus 로고
    • Transformation of diploid human fibroblasts by transfection with the v-sis, PDGF2/csis, or T24 H-ras genes
    • Fry DG, Hurlin PJ, Maher VM, McCormick JJ. Transformation of diploid human fibroblasts by transfection with the v-sis, PDGF2/csis, or T24 H-ras genes. Mutat Res 1988; 199: 341-51.
    • (1988) Mutat Res , vol.199 , pp. 341-351
    • Fry, D.G.1    Hurlin, P.J.2    Maher, V.M.3    McCormick, J.J.4
  • 41
    • 0037253808 scopus 로고    scopus 로고
    • A fluorogenic histone deacetylase assay well suited for high-throughput activity screening
    • Wegener D, Wirsching F, Riester D, Schwienhorst A. A fluorogenic histone deacetylase assay well suited for high-throughput activity screening. Chem Biol 2003; 10: 61-68.
    • (2003) Chem Biol , vol.10 , pp. 61-68
    • Wegener, D.1    Wirsching, F.2    Riester, D.3    Schwienhorst, A.4
  • 42
    • 0024996768 scopus 로고
    • Potent and specific inhibition of mammalian histone deacetylase both in vivo and in vitro by trichostatin A
    • Yoshida M, Kijima M, Akita M, Beppu T. Potent and specific inhibition of mammalian histone deacetylase both in vivo and in vitro by trichostatin A. J Biol Chem 1990; 265: 17174-9.
    • (1990) J Biol Chem , vol.265 , pp. 17174-17179
    • Yoshida, M.1    Kijima, M.2    Akita, M.3    Beppu, T.4
  • 43
    • 0027378351 scopus 로고
    • Trapoxin, an antitumor cyclic tetrapeptide, is an irreversible inhibitor of mammalian histone deacetylase
    • Kijima M, Yoshida M, Sugita K, Horinouchi S, Beppu T. Trapoxin, an antitumor cyclic tetrapeptide, is an irreversible inhibitor of mammalian histone deacetylase. J Biol Chem 1993; 268: 22429-35.
    • (1993) J Biol Chem , vol.268 , pp. 22429-22435
    • Kijima, M.1    Yoshida, M.2    Sugita, K.3    Horinouchi, S.4    Beppu, T.5
  • 44
    • 0033539092 scopus 로고    scopus 로고
    • Structures of a histone deacetylase homologue bound to the TSA and SAHA inhibitors
    • Finnin MS, Donigian JR, Cohen A, et al. Structures of a histone deacetylase homologue bound to the TSA and SAHA inhibitors. Nature 1999; 401: 188-93.
    • (1999) Nature , vol.401 , pp. 188-193
    • Finnin, M.S.1    Donigian, J.R.2    Cohen, A.3
  • 45
    • 0029795991 scopus 로고    scopus 로고
    • Synthesis of natural and modified trapoxins, useful reagents for exploring histone deacetylase function
    • Taunton J, Collins JL, Schreiber SL. Synthesis of natural and modified trapoxins, useful reagents for exploring histone deacetylase function. J Am Chem Soc 1996; 118: 10412-22.
    • (1996) J Am Chem Soc , vol.118 , pp. 10412-10422
    • Taunton, J.1    Collins, J.L.2    Schreiber, S.L.3
  • 46
    • 0344431240 scopus 로고    scopus 로고
    • FR901228, a potent antitumor antibiotic, is a novel histone deacetylase inhibitor
    • Nakajima H, Kim YB, Terano H, Yoshida M, Horinouchi S. FR901228, a potent antitumor antibiotic, is a novel histone deacetylase inhibitor. Expt Cell Res 1998; 241: 126-33.
    • (1998) Expt Cell Res , vol.241 , pp. 126-133
    • Nakajima, H.1    Kim, Y.B.2    Terano, H.3    Yoshida, M.4    Horinouchi, S.5
  • 47
    • 0035698217 scopus 로고    scopus 로고
    • New trichostatin derivative, trichostatin RK, from Streptomyces sp. RK98-A74
    • Uekia M, Teruya T, Nie L, Usamib R, Yoshida M, Osada H. New trichostatin derivative, trichostatin RK, from Streptomyces sp. RK98-A74. J Antibiot 2001; 54: 1093-5.
    • (2001) J Antibiot , vol.54 , pp. 1093-1095
    • Uekia, M.1    Teruya, T.2    Nie, L.3    Usamib, R.4    Yoshida, M.5    Osada, H.6
  • 48
    • 62149098372 scopus 로고    scopus 로고
    • Psammaplin A as a general activator of cell-based signaling assays via HDAC inhibition and studies on some bromotyrosine derivatives
    • McCulloch MWB, Coombs GS, Banerjee N, et al. Psammaplin A as a general activator of cell-based signaling assays via HDAC inhibition and studies on some bromotyrosine derivatives. Bioorg Med Chem 2009; 17: 2189-98.
    • (2009) Bioorg Med Chem , vol.17 , pp. 2189-2198
    • McCulloch, M.W.B.1    Coombs, G.S.2    Banerjee, N.3
  • 49
    • 33744956666 scopus 로고    scopus 로고
    • Histone deacetylase 3 (HDAC3) and other class I HDACs regulate colon cell maturation and p21 expression and are deregulated in human colon cancer
    • Wilson AJ, Byun D, Popova N, et al. Histone deacetylase 3 (HDAC3) and other class I HDACs regulate colon cell maturation and p21 expression and are deregulated in human colon cancer. J Biol Chem 2006; 281: 13548-58.
    • (2006) J Biol Chem , vol.281 , pp. 13548-13558
    • Wilson, A.J.1    Byun, D.2    Popova, N.3
  • 51
    • 23944525341 scopus 로고    scopus 로고
    • Assays for mechanistic investigations of protein/histone acetyltransferases
    • Berndsen CE, Denu JM. Assays for mechanistic investigations of protein/histone acetyltransferases. Methods 2005; 36: 321-31.
    • (2005) Methods , vol.36 , pp. 321-331
    • Berndsen, C.E.1    Denu, J.M.2
  • 52
    • 41149089267 scopus 로고    scopus 로고
    • Histone deacetylase inhibitors: From bench to clinic
    • Paris M, Porcelloni M, Binaschi M, Fattori D. Histone deacetylase inhibitors: From bench to clinic. J Med Chem 2008; 51: 1505-29.
    • (2008) J Med Chem , vol.51 , pp. 1505-1529
    • Paris, M.1    Porcelloni, M.2    Binaschi, M.3    Fattori, D.4
  • 54
    • 84861157767 scopus 로고    scopus 로고
    • From discovery to broad-spectrum therapy
    • Hong J, Luesch H. Largazole: From discovery to broad-spectrum therapy. Nat Prod Rep 2012; 29: 449-56.
    • (2012) Nat Prod Rep , vol.29 , pp. 449-456
    • Hong, J.1    Largazole, L.H.2
  • 55
    • 31544436888 scopus 로고    scopus 로고
    • Dietary HDAC inhibitors: Time to rethink weak ligands in cancer chemoprevention?
    • Dashwood RH, Myzak MC, Ho E. Dietary HDAC inhibitors: time to rethink weak ligands in cancer chemoprevention? Carcinogenesis 2006; 27: 344-9.
    • (2006) Carcinogenesis , vol.27 , pp. 344-349
    • Dashwood, R.H.1    Myzak, M.C.2    Ho, E.3
  • 56
    • 0018371969 scopus 로고
    • Different accessibilities in chromatin to histone acetylase
    • Cousens LS, Gallwitz D, Alberts BM. Different accessibilities in chromatin to histone acetylase. J Biol Chem 1979; 254: 1716-23.
    • (1979) J Biol Chem , vol.254 , pp. 1716-1723
    • Cousens, L.S.1    Gallwitz, D.2    Alberts, B.M.3
  • 57
    • 70349525349 scopus 로고    scopus 로고
    • Discovery, biological activity, synthesis and potential therapeutic utility of naturally occurring histone deacetylase inhibitors
    • Newkirk TL, Bowers AA, Williams RM. Discovery, biological activity, synthesis and potential therapeutic utility of naturally occurring histone deacetylase inhibitors. Nat Prod Rep 2009; 26: 1293-320.
    • (2009) Nat Prod Rep , vol.26 , pp. 1293-1320
    • Newkirk, T.L.1    Bowers, A.A.2    Williams, R.M.3
  • 59
    • 0023195737 scopus 로고
    • Effects of trichostatins on differentiation of murine erythroleukemia cells
    • Yoshida M, Nomura S, Beppu T. Effects of trichostatins on differentiation of murine erythroleukemia cells. Cancer Res 1987; 47: 3688-91.
    • (1987) Cancer Res , vol.47 , pp. 3688-3691
    • Yoshida, M.1    Nomura, S.2    Beppu, T.3
  • 60
    • 0034054071 scopus 로고    scopus 로고
    • Trichostatin D, a new inducer of phenotypic reversion in transformed cells
    • Hayakawa Y, Nakai M, Furihata K, Shin-ya K, Seto H. Trichostatin D, a new inducer of phenotypic reversion in transformed cells. J Antibiot 2000; 53: 179-83.
    • (2000) J Antibiot , vol.53 , pp. 179-183
    • Hayakawa, Y.1    Nakai, M.2    Furihata, K.3    Shin-ya, K.4    Seto, H.5
  • 61
    • 0021878370 scopus 로고
    • A new differentiation inducer of Friend leukemia cells, trichostatic acid
    • Morioka H, Ishihara M, Takezawa M, et al. A new differentiation inducer of Friend leukemia cells, trichostatic acid. Agric Biol Chem 1985; 49: 1365-70.
    • (1985) Agric Biol Chem , vol.49 , pp. 1365-1370
    • Morioka, H.1    Ishihara, M.2    Takezawa, M.3
  • 62
    • 0018114158 scopus 로고
    • Trichostatin C, a glucopyranosyl hydroxamate
    • Tsuji N, Kobayashi M. Trichostatin C, a glucopyranosyl hydroxamate. J Antibiot 1978; 31: 940-4.
    • (1978) J Antibiot , vol.31 , pp. 940-944
    • Tsuji, N.1    Kobayashi, M.2
  • 63
    • 67649976728 scopus 로고    scopus 로고
    • JBIR-17, a novel trichostatin analog from Streptomyces sp. 26634
    • Ueda J, Hwang J, Maeda S, et al. JBIR-17, a novel trichostatin analog from Streptomyces sp. 26634. J Antibiot 2009; 62: 283-5.
    • (2009) J Antibiot , vol.62 , pp. 283-285
    • Ueda, J.1    Hwang, J.2    Maeda, S.3
  • 64
    • 0026755646 scopus 로고
    • Depudecin: A novel compound inducing the flat phenotype of NIH3T3 cells doubly transformed by ras-and src-oncogene, produced by Alternaria brassicicola
    • Matsumoto M, Matsutani S, Sugita K, et al. Depudecin: A novel compound inducing the flat phenotype of NIH3T3 cells doubly transformed by ras-and src-oncogene, produced by Alternaria brassicicola. J Antibiot 1992; 45: 879-5.
    • (1992) J Antibiot , vol.45 , pp. 879-885
    • Matsumoto, M.1    Matsutani, S.2    Sugita, K.3
  • 65
    • 0034132612 scopus 로고    scopus 로고
    • Biosynthesis of depudecin, a metabolite of Nimbya scirpicola
    • Tanaka M, Fujimori T, Nabeta K. Biosynthesis of depudecin, a metabolite of Nimbya scirpicola. Biosci Biotechnol Biochem 2000; 64: 244-7.
    • (2000) Biosci Biotechnol Biochem , vol.64 , pp. 244-247
    • Tanaka, M.1    Fujimori, T.2    Nabeta, K.3
  • 66
    • 0000055673 scopus 로고
    • Phenolic constituents of Psammaplysilla
    • Quinoa E, Crews P. Phenolic constituents of Psammaplysilla. Tetrahedron Lett 1987; 28: 3229-32.
    • (1987) Tetrahedron Lett , vol.28 , pp. 3229-3232
    • Quinoa, E.1    Crews, P.2
  • 67
    • 0026099706 scopus 로고
    • Novel marine sponge derived amino acids 13. Additional psammaplin derivatives from Psammaplysilla purpurea
    • Jimenez C, Crews P. Novel marine sponge derived amino acids 13. Additional psammaplin derivatives from Psammaplysilla purpurea. Tetrahedron 1991; 47: 2097-102.
    • (1991) Tetrahedron , vol.47 , pp. 2097-2102
    • Jimenez, C.1    Crews, P.2
  • 68
    • 0000529638 scopus 로고    scopus 로고
    • Isolation of psammaplin A 11'-sulfate and bisaprasin 11'-sulfate from the marine sponge Aplysinella rhax
    • Pham NB, Butler MS, Quinn RJ. Isolation of psammaplin A 11'-sulfate and bisaprasin 11'-sulfate from the marine sponge Aplysinella rhax. J Nat Prod 2000; 63: 393-5.
    • (2000) J Nat Prod , vol.63 , pp. 393-395
    • Pham, N.B.1    Butler, M.S.2    Quinn, R.J.3
  • 69
    • 0034634603 scopus 로고    scopus 로고
    • New bromotyrosine metabolites from the sponge Aplysinella rhax
    • Shin J, Lee H, Seo Y, Rho J, Cho KW, Paul VJ. New bromotyrosine metabolites from the sponge Aplysinella rhax. Tetrahedron 2000; 56: 9071-9077.
    • (2000) Tetrahedron , vol.56 , pp. 9071-9077
    • Shin, J.1    Lee, H.2    Seo, Y.3    Rho, J.4    Cho, K.W.5    Paul, V.J.6
  • 70
    • 0038627550 scopus 로고    scopus 로고
    • Psammaplins from the sponge Pseudoceratina purpurea: Inhibition of both histone deacetylase and DNA methyltransferase
    • Pina IC, Gautschi JT, Wang G, et al. Psammaplins from the sponge Pseudoceratina purpurea: Inhibition of both histone deacetylase and DNA methyltransferase. J Org Chem 2003; 68: 3866-73.
    • (2003) J Org Chem , vol.68 , pp. 3866-3873
    • Pina, I.C.1    Gautschi, J.T.2    Wang, G.3
  • 71
    • 33847657556 scopus 로고    scopus 로고
    • Psammaplin A is a natural prodrug that inhibits class I histone deacetylase
    • Kim DH, Shin J, Kwon HJ. Psammaplin A is a natural prodrug that inhibits class I histone deacetylase. Exp Mol Med 2007; 39: 47-55.
    • (2007) Exp Mol Med , vol.39 , pp. 47-55
    • Kim, D.H.1    Shin, J.2    Kwon, H.J.3
  • 72
    • 51849181148 scopus 로고
    • Determination of D-amino acids. II. Use of a bifunctional reagent, 1,5-difluoro-2,4-dinitrobenzene
    • Marfey P. Determination of D-amino acids. II. Use of a bifunctional reagent, 1,5-difluoro-2,4-dinitrobenzene. Carlsberg Res Commun 1984; 49: 591-6.
    • (1984) Carlsberg Res Commun , vol.49 , pp. 591-596
    • Marfey, P.1
  • 73
    • 0001479941 scopus 로고    scopus 로고
    • A nonempirical method using LC/MS for determination of the absolute configuration of constituent amino acids in a peptide: Combination of Marfey's method with mass spectrometry and its practical application
    • Fujii K, Ikai Y, Oka H, Suzuki M, Harada K. A nonempirical method using LC/MS for determination of the absolute configuration of constituent amino acids in a peptide: combination of Marfey's method with mass spectrometry and its practical application. Anal Chem 1997; 69: 5146-51.
    • (1997) Anal Chem , vol.69 , pp. 5146-5151
    • Fujii, K.1    Ikai, Y.2    Oka, H.3    Suzuki, M.4    Harada, K.5
  • 74
    • 33947085552 scopus 로고
    • Nuclear magnetic resonance enantiomer reagents. Configurational correlations via nuclear magnetic resonance chemical shifts of diastereomeric mandelate, Omethylmandelate, and α-methoxy-α-trifluoromethylphenylacetate (MTPA) esters
    • Dale JA, Mosher HS. Nuclear magnetic resonance enantiomer reagents. Configurational correlations via nuclear magnetic resonance chemical shifts of diastereomeric mandelate, Omethylmandelate, and α-methoxy-α-trifluoromethylphenylacetate (MTPA) esters. J Am Chem Soc 1973; 95: 512-9.
    • (1973) J Am Chem Soc , vol.95 , pp. 512-519
    • Dale, J.A.1    Mosher, H.S.2
  • 75
    • 0025673805 scopus 로고
    • Isolation and structural elucidation of new cyclotetrapeptides, trapoxins A and B, having detransformation activities as antitumor agents
    • Itazaki H, Nagashima K, Sugita K, et al. Isolation and structural elucidation of new cyclotetrapeptides, trapoxins A and B, having detransformation activities as antitumor agents. J Antibiot 1990; 43: 1524-32.
    • (1990) J Antibiot , vol.43 , pp. 1524-1532
    • Itazaki, H.1    Nagashima, K.2    Sugita, K.3
  • 76
    • 0039397898 scopus 로고
    • Isolation and biological activities of four selective toxins from Helminthosporium carbonum
    • Rasmussen JB, Scheffer RP. Isolation and biological activities of four selective toxins from Helminthosporium carbonum. Plant Physiol 1988; 86: 187-91.
    • (1988) Plant Physiol , vol.86 , pp. 187-191
    • Rasmussen, J.B.1    Scheffer, R.P.2
  • 77
    • 0016366116 scopus 로고
    • Isolierung und Strukturaufklärung von Chlamydocin
    • Closse A, Huguenin R. Isolierung und Strukturaufklärung von Chlamydocin. Helv Chim Acta 1974; 57: 533-45.
    • (1974) Helv Chim Acta , vol.57 , pp. 533-545
    • Closse, A.1    Huguenin, R.2
  • 78
    • 0020519166 scopus 로고
    • Studies on WF-3161, a new antitumor antibiotic
    • Umehara K, Nakahara K, Kiyoto S, et al. Studies on WF-3161, a new antitumor antibiotic. J Antibiot 1983; 36: 478-83.
    • (1983) J Antibiot , vol.36 , pp. 478-483
    • Umehara, K.1    Nakahara, K.2    Kiyoto, S.3
  • 79
    • 0015792162 scopus 로고
    • Isolation and biological activity of Cyl-2, a metabolite of Cylindrocladium scoparium
    • Hirota A, Suzuki A, Suzuki H, Tamura S. Isolation and biological activity of Cyl-2, a metabolite of Cylindrocladium scoparium. Agric Biol Chem 1973; 37: 643-47.
    • (1973) Agric Biol Chem , vol.37 , pp. 643-647
    • Hirota, A.1    Suzuki, A.2    Suzuki, H.3    Tamura, S.4
  • 80
    • 34249290099 scopus 로고    scopus 로고
    • Microsporins A and B: New histone deacetylase inhibitors from the marine-derived fungus Microsporum cf. gypseum and the solidphase synthesis of microsporin A
    • Gu W, Cueto M, Jensen PR, Fenical W, Silverman RB. Microsporins A and B: new histone deacetylase inhibitors from the marine-derived fungus Microsporum cf. gypseum and the solidphase synthesis of microsporin A. Tetrahedron 2007; 63: 6535-41.
    • (2007) Tetrahedron , vol.63 , pp. 6535-6541
    • Gu, W.1    Cueto, M.2    Jensen, P.R.3    Fenical, W.4    Silverman, R.B.5
  • 81
    • 0030569401 scopus 로고    scopus 로고
    • Apicidins: Novel cyclic tetrapeptides as coccidiostats and antimalarial agents from Fusarium pallidoroseum
    • Singh SB, Zink DL, Polishook JD, et al. Apicidins: novel cyclic tetrapeptides as coccidiostats and antimalarial agents from Fusarium pallidoroseum. Tetrahedron Lett 1996; 37: 8077-80.
    • (1996) Tetrahedron Lett , vol.37 , pp. 8077-8080
    • Singh, S.B.1    Zink, D.L.2    Polishook, J.D.3
  • 82
    • 0035817996 scopus 로고    scopus 로고
    • Structure, histone deacetylase, and antiprotozoal activities of apicidins B and C, congeners of apicidin with proline and valine substitutions
    • Singh SB, Zink DL, Liesch JM, et al. Structure, histone deacetylase, and antiprotozoal activities of apicidins B and C, congeners of apicidin with proline and valine substitutions. Org Lett 2001; 3: 2815-8.
    • (2001) Org Lett , vol.3 , pp. 2815-2818
    • Singh, S.B.1    Zink, D.L.2    Liesch, J.M.3
  • 83
    • 0037039919 scopus 로고    scopus 로고
    • Structure and chemistry of apicidins, a class of novel cyclic tetrapeptides without a terminal α-keto epoxide as inhibitors of histone deacetylase with potent antiprotozoal activities
    • Singh SB, Zink DL, Liesch JM, et al. Structure and chemistry of apicidins, a class of novel cyclic tetrapeptides without a terminal α-keto epoxide as inhibitors of histone deacetylase with potent antiprotozoal activities. J Org Chem 2002; 67: 815-25.
    • (2002) J Org Chem , vol.7 , pp. 15-25
    • Singh, S.B.1    Zink, D.L.2    Liesch, J.M.3
  • 84
    • 0037300444 scopus 로고    scopus 로고
    • FR235222, a fungal metabolite, is a novel immunosuppressant that inhibits mammalian histone deacetylase (HDAC). I. Taxonomy, fermentation, isolation and biological activities
    • Mori H, Urano Y, Abe F, et al. FR235222, a fungal metabolite, is a novel immunosuppressant that inhibits mammalian histone deacetylase (HDAC). I. Taxonomy, fermentation, isolation and biological activities. J Antibiot 2003; 56: 72-9.
    • (2003) J Antibiot , vol.56 , pp. 72-79
    • Mori, H.1    Urano, Y.2    Abe, F.3
  • 85
    • 0037300480 scopus 로고    scopus 로고
    • FR235222, a fungal metabolite, is a novel immunosuppressant that inhibits mammalian histone deacetylase (HDAC). II. Biological activities in animal models
    • Mori H, Abe F, Furukawa S, et al. FR235222, a fungal metabolite, is a novel immunosuppressant that inhibits mammalian histone deacetylase (HDAC). II. Biological activities in animal models. J Antibiot 2003; 56: 80-6.
    • (2003) J Antibiot , vol.56 , pp. 80-86
    • Mori, H.1    Abe, F.2    Furukawa, S.3
  • 86
    • 0037300543 scopus 로고    scopus 로고
    • FR235222, a fungal metabolite, is a novel immunosuppressant that inhibits mammalian histone deacetylase. III. Structure determination
    • Mori H, Urano Y, Kinoshita T, Yoshimura S, Takase S, Hino M. FR235222, a fungal metabolite, is a novel immunosuppressant that inhibits mammalian histone deacetylase. III. Structure determination. J Antibiot 2003; 56: 181-5.
    • (2003) J Antibiot , vol.56 , pp. 181-185
    • Mori, H.1    Urano, Y.2    Kinoshita, T.3    Yoshimura, S.4    Takase, S.5    Hino, M.6
  • 87
    • 41849109752 scopus 로고    scopus 로고
    • Design and synthesis of cyclopeptide analogs of the potent histone deacetylase inhibitor FR235222
    • Gomez-Paloma L, Bruno I, Cini E, et al. Design and synthesis of cyclopeptide analogs of the potent histone deacetylase inhibitor FR235222. Chem Med Chem 2007; 2: 1511-9.
    • (2007) Chem Med Chem , vol.2 , pp. 1511-1519
    • Gomez-Paloma, L.1    Bruno, I.2    Cini, E.3
  • 88
    • 78649648097 scopus 로고    scopus 로고
    • AS1387392, a novel immunosuppressive cyclic tetrapeptide compound with inhibitory activity against mammalian histone deacetylase
    • Sasamura S, Sakamoto K, Takagaki S, et al. AS1387392, a novel immunosuppressive cyclic tetrapeptide compound with inhibitory activity against mammalian histone deacetylase. J Antibiot 2010; 63: 633-36.
    • (2010) J Antibiot , vol.63 , pp. 633-636
    • Sasamura, S.1    Sakamoto, K.2    Takagaki, S.3
  • 89
    • 33845199114 scopus 로고    scopus 로고
    • Azumamides A-E: Histone deacetylase inhibitory cyclic tetrapeptides from the marine sponge Mycale izuensis
    • Nakao Y, Yoshida S, Matsunaga S, et al. Azumamides A-E: Histone deacetylase inhibitory cyclic tetrapeptides from the marine sponge Mycale izuensis. Angew Chem Int Ed 2006; 45: 7553-7.
    • (2006) Angew Chem Int Ed , vol.45 , pp. 7553-7557
    • Nakao, Y.1    Yoshida, S.2    Matsunaga, S.3
  • 90
    • 78649235245 scopus 로고    scopus 로고
    • Weapons in disguise-activating mechanisms and protecting group chemistry in Nature
    • Kwan JC, Luesch H. Weapons in disguise-activating mechanisms and protecting group chemistry in Nature. Chem Eur J 2010; 16: 13020-9.
    • (2010) Chem Eur J , vol.16 , pp. 13020-13029
    • Kwan, J.C.1    Luesch, H.2
  • 91
    • 0028258610 scopus 로고
    • FR901228, a novel antitumor bicyclic depsipeptide produced by Chromobacterium violaceum No. 968. I. Taxonomy, fermentation, isolation, physico-chemical and biological properties, and antitumor activity
    • Ueda H, Nakajima H, Hori Y, et al. FR901228, a novel antitumor bicyclic depsipeptide produced by Chromobacterium violaceum No. 968. I. Taxonomy, fermentation, isolation, physico-chemical and biological properties, and antitumor activity. J Antibiot 1994; 47: 301-10.
    • (1994) J Antibiot , vol.47 , pp. 301-310
    • Ueda, H.1    Nakajima, H.2    Hori, Y.3
  • 92
    • 0028299638 scopus 로고
    • FR901228, a novel antitumor bicyclic depsipeptide produced by Chromobacterium violaceum No. 968. II. Structure determination
    • Shigematsu N, Ueda H, Takase S, Tanaka H. FR901228, a novel antitumor bicyclic depsipeptide produced by Chromobacterium violaceum No. 968. II. Structure determination. J Antibiot 1994; 47: 311-4.
    • (1994) J Antibiot , vol.47 , pp. 311-314
    • Shigematsu, N.1    Ueda, H.2    Takase, S.3    Tanaka, H.4
  • 93
    • 0036735385 scopus 로고    scopus 로고
    • FK228 (Depsipeptide) as a natural prodrug that inhibits class I histone deacetylases
    • Furumai R, Matsuyama A, Kobashi N, et al. FK228 (Depsipeptide) as a natural prodrug that inhibits class I histone deacetylases. Cancer Res 2002; 62: 4916-21.
    • (2002) Cancer Res , vol.62 , pp. 4916-4921
    • Furumai, R.1    Matsuyama, A.2    Kobashi, N.3
  • 94
    • 0035180541 scopus 로고    scopus 로고
    • Spiruchostatins A and B, novel gene expression-enhancing substances produced by Pseudomonas sp
    • Masuoka Y, Nagai A, Shin-ya K, et al. Spiruchostatins A and B, novel gene expression-enhancing substances produced by Pseudomonas sp. Tetrahedron Lett 2001; 42: 41-4.
    • (2001) Tetrahedron Lett , vol.42 , pp. 41-44
    • Masuoka, Y.1    Nagai, A.2    Shin-ya, K.3
  • 96
    • 39049135494 scopus 로고    scopus 로고
    • Structure and activity of largazole, a potent antiproliferative agent from the Floridian marine cyanobacterium Symploca sp
    • Taori K, Paul VJ, Luesch H. Structure and activity of largazole, a potent antiproliferative agent from the Floridian marine cyanobacterium Symploca sp. J Am Chem Soc 2008; 130: 1806-7.
    • (2008) J Am Chem Soc , vol.130 , pp. 1806-1807
    • Taori, K.1    Paul, V.J.2    Luesch, H.3
  • 97
    • 46049100010 scopus 로고    scopus 로고
    • Total synthesis and molecular target of largazole, a histone deacetylase inhibitor
    • Ying Y, Taori K, Kim H, Hong J, Luesch H. Total synthesis and molecular target of largazole, a histone deacetylase inhibitor. J Am Chem Soc 2008; 130: 8455-9.
    • (2008) J Am Chem Soc , vol.130 , pp. 8455-8459
    • Ying, Y.1    Taori, K.2    Kim, H.3    Hong, J.4    Luesch, H.5
  • 98
    • 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 RA, Altaf M, 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: 51163-71.
    • (2004) J Biol Chem , vol.279 , pp. 51163-51171
    • Balasubramanyam, K.1    Varier, R.A.2    Altaf, M.3
  • 99
    • 58349094740 scopus 로고    scopus 로고
    • Epigallocatechin-3-gallate, a histone acetyltransferase inhibitor, inhibits EBV-induced B lymphocyte transformation via suppression of RelA acetylation
    • Choi K, Jung MG, Lee YH, et al. Epigallocatechin-3-gallate, a histone acetyltransferase inhibitor, inhibits EBV-induced B lymphocyte transformation via suppression of RelA acetylation. Cancer Research 2009; 69: 583-92.
    • (2009) Cancer Research , vol.69 , pp. 583-592
    • Choi, K.1    Jung, M.G.2    Lee, Y.H.3
  • 100
    • 77955473296 scopus 로고    scopus 로고
    • Improved inhibition of the histone acetyltransferase PCAF by an anacardic acid derivative
    • Ghizzoni M, Boltjes A, Graaf C, Haisma HJ, Dekker FJ. Improved inhibition of the histone acetyltransferase PCAF by an anacardic acid derivative. Bioorg Med Chem 2010; 18: 5826-34.
    • (2010) Bioorg Med Chem , vol.18 , pp. 5826-5834
    • Ghizzoni, M.1    Boltjes, A.2    Graaf, C.3    Haisma, H.J.4    Dekker, F.J.5
  • 101
    • 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 RA, et al. Polyisoprenylated benzophenone, garcinol, a natural histone acetyltransferase inhibitor, represses chromatin transcription and alters global gene expression. J Biol Chem 2004; 279: 33716-26.
    • (2004) J Biol Chem , vol.279 , pp. 33716-33726
    • Balasubramanyam, K.1    Altaf, M.2    Varier, R.A.3
  • 102
    • 77950663978 scopus 로고    scopus 로고
    • The identification of a novel natural activator of p300 histone acetyltranferase provides new insights into the modulation mechanism of this enzyme
    • Dal Piaz F, Tosco A, Eletto D, et al. The identification of a novel natural activator of p300 histone acetyltranferase provides new insights into the modulation mechanism of this enzyme. ChemBio-Chem 2010; 11: 818-27.
    • (2010) ChemBio-Chem , vol.11 , pp. 818-827
    • Dal, P.F.1    Tosco, A.2    Eletto, D.3
  • 103
    • 60549106446 scopus 로고    scopus 로고
    • Mechanism of p300 specific histone acetyltransferase inhibition by small molecules
    • Arif M, Pradhan SK, Thanuja GK, et al. Mechanism of p300 specific histone acetyltransferase inhibition by small molecules. J Med Chem 2009; 52: 267-77.
    • (2009) J Med Chem , vol.52 , pp. 267-277
    • Arif, M.1    Pradhan, S.K.2    Thanuja, G.K.3
  • 104
    • 27444435580 scopus 로고    scopus 로고
    • Toward selective histone deacetylase inhibitor design: Homology modeling, docking studies, and molecular dynamics simulations of human class I histone deacetylases
    • Wang D, Helquist P, Wiech NL, Wiest O. Toward selective histone deacetylase inhibitor design: Homology modeling, docking studies, and molecular dynamics simulations of human class I histone deacetylases. J Med Chem 2005; 48: 6936-47.
    • (2005) J Med Chem , vol.48 , pp. 6936-6947
    • Wang, D.1    Helquist, P.2    Wiech, N.L.3    Wiest, O.4
  • 105
    • 76149120388 scopus 로고    scopus 로고
    • AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading
    • Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem 2010; 31: 455-61.
    • (2010) J Comput Chem , vol.31 , pp. 455-461
    • Trott, O.1    Olson, A.J.2
  • 106
    • 33846122993 scopus 로고    scopus 로고
    • Dimethyl sulfoxide to vorinostat: Development of this histone deacetylase inhibitor as an anticancer drug
    • Marks PA, Breslow R. Dimethyl sulfoxide to vorinostat: development of this histone deacetylase inhibitor as an anticancer drug. Nat Biotechnol 2007; 25: 84-90.
    • (2007) Nat Biotechnol , vol.25 , pp. 84-90
    • Marks, P.A.1    Breslow, R.2
  • 107
    • 33745314784 scopus 로고    scopus 로고
    • Solution, solid phase and computational structures of apicidin and its backbone-reduced analogs
    • Kranz M, Murray PJ, Taylor S, Upton RJ, Clegg W, Elsegood MR. Solution, solid phase and computational structures of apicidin and its backbone-reduced analogs. J Pept Sci 2006; 12: 383-8.
    • (2006) J Pept Sci , vol.12 , pp. 383-388
    • Kranz, M.1    Murray, P.J.2    Taylor, S.3    Upton, R.J.4    Clegg, W.5    Elsegood, M.R.6
  • 108
    • 0034619126 scopus 로고    scopus 로고
    • Synthesis of side chain modified apicidin derivatives: Potent mechanism-based histone deacetylase inhibitors
    • Meinke PT, Colletti SL, Ayer MB, et al. Synthesis of side chain modified apicidin derivatives: potent mechanism-based histone deacetylase inhibitors. Tetrahedron Lett 2000; 41: 7831-5.
    • (2000) Tetrahedron Lett , vol.41 , pp. 7831-7835
    • Meinke, P.T.1    Colletti, S.L.2    Ayer, M.B.3
  • 109
    • 0034619150 scopus 로고    scopus 로고
    • Tryptophan-replacement and indole-modified apicidins: Synthesis of potent and selective antiprotozoal agents
    • Colletti SL, Li C, Fisher MH, Wyvratt MJ, Meinke PT. Tryptophan-replacement and indole-modified apicidins: synthesis of potent and selective antiprotozoal agents. Tetrahedron Lett 2000; 41: 7825-9.
    • (2000) Tetrahedron Lett , vol.41 , pp. 7825-7829
    • Colletti, S.L.1    Li, C.2    Fisher, M.H.3    Wyvratt, M.J.4    Meinke, P.T.5
  • 110
    • 0035931503 scopus 로고    scopus 로고
    • Broad spectrum antiprotozoal agents that inhibit histone deacetylase: Structureactivity relationships of apicidin. Part 1
    • Colletti SL, Myers RW, Darkin-Rattray SJ, et al. Broad spectrum antiprotozoal agents that inhibit histone deacetylase: structureactivity relationships of apicidin. Part 1. Bioorg Med Chem Lett 2001; 11: 107-11.
    • (2001) Bioorg Med Chem Lett , vol.11 , pp. 107-111
    • Colletti, S.L.1    Myers, R.W.2    Darkin-Rattray, S.J.3
  • 111
    • 0035931503 scopus 로고    scopus 로고
    • Broad spectrum antiprotozoal agents that inhibit histone deacetylase: Structure-activity relationships of apicidin. Part 2
    • Colletti SL, Myers RW, Darkin-Rattray SJ, et al. Broad spectrum antiprotozoal agents that inhibit histone deacetylase: structure-activity relationships of apicidin. Part 2. Bioorg Med Chem Lett 2001; 11: 113-7.
    • (2001) Bioorg Med Chem Lett , vol.11 , pp. 113-117
    • Colletti, S.L.1    Myers, R.W.2    Darkin-Rattray, S.J.3
  • 112
    • 30444433547 scopus 로고    scopus 로고
    • Total synthesis, NMR solution structure, and binding model of the potent histone deacetylase inhibitor FR235222
    • Rodriquez M, Terracciano S, Cini E, et al. Total synthesis, NMR solution structure, and binding model of the potent histone deacetylase inhibitor FR235222. Angew Chem Int Ed 2006; 45: 423-7.
    • (2006) Angew Chem Int Ed , vol.45 , pp. 423-427
    • Rodriquez, M.1    Terracciano, S.2    Cini, E.3
  • 113
    • 41849134699 scopus 로고    scopus 로고
    • Synthesis and biological evaluation of histone deacetylase inhibitors that are based on FR235222: A cyclic tetrapeptide scaffold
    • Singh EK, Ravula S, Pan CM, et al. Synthesis and biological evaluation of histone deacetylase inhibitors that are based on FR235222: a cyclic tetrapeptide scaffold. Bioorg Med Chem Lett 2008; 18: 2549-54.
    • (2008) Bioorg Med Chem Lett , vol.18 , pp. 2549-2554
    • Singh, E.K.1    Ravula, S.2    Pan, C.M.3
  • 114
    • 51549109903 scopus 로고    scopus 로고
    • Molecular modeling studies toward the structural optimization of new cyclopeptide-based HDAC inhibitors modeled on the natural product FR235222
    • Di Micco S, Terracciano S, Bruno I, et al. Molecular modeling studies toward the structural optimization of new cyclopeptide-based HDAC inhibitors modeled on the natural product FR235222. Bioorg Med Chem 2008; 16: 8635-42.
    • (2008) Bioorg Med Chem , vol.16 , pp. 8635-8642
    • Di Micco, S.1    Terracciano, S.2    Bruno, I.3
  • 115
    • 33947239221 scopus 로고    scopus 로고
    • Molecular insights into azumamide E histone deacetylases inhibitory activity
    • Maulucci N, Chini MG, Di Micco SG, et al. Molecular insights into azumamide E histone deacetylases inhibitory activity. J Am Chem Soc 2007; 129: 3007-12.
    • (2007) J Am Chem Soc , vol.129 , pp. 3007-3012
    • Maulucci, N.1    Chini, M.G.2    Di Micco, S.G.3
  • 116
    • 50249144006 scopus 로고    scopus 로고
    • Total synthesis and biological mode of action of largazole: A potent class I histone deacetylase inhibitor
    • Bowers A, West N, Taunton J, Schreiber SL, Bradner JE, Williams RM. Total synthesis and biological mode of action of largazole: A potent class I histone deacetylase inhibitor. J Am Chem Soc 2008; 130: 11219-22.
    • (2008) J Am Chem Soc , vol.130 , pp. 11219-11222
    • Bowers, A.1    West, N.2    Taunton, J.3    Schreiber, S.L.4    Bradner, J.E.5    Williams, R.M.6
  • 117
    • 54049152858 scopus 로고    scopus 로고
    • A concise total synthesis of largazole, solution structure, and some preliminary structure activity relationships
    • Nasveschuk CG, Ungermannova D, Liu X, Phillips AJ. A concise total synthesis of largazole, solution structure, and some preliminary structure activity relationships. Org Lett 2008; 10: 3595-8.
    • (2008) Org Lett , vol.10 , pp. 3595-3598
    • Nasveschuk, C.G.1    Ungermannova, D.2    Liu, X.3    Phillips, A.J.4
  • 118
    • 52049119362 scopus 로고    scopus 로고
    • Synthesis and biological activity of largazole and derivatives
    • Seiser T, Kamena F, Cramer N. Synthesis and biological activity of largazole and derivatives. Angew Chem Int Ed 2008; 47: 6483-5.
    • (2008) Angew Chem Int Ed , vol.47 , pp. 6483-6485
    • Seiser, T.1    Kamena, F.2    Cramer, N.3
  • 119
    • 54149114729 scopus 로고    scopus 로고
    • Total synthesis of largazole and its biological evaluation
    • Numajiri Y, Takahashi T, Takagi M, Shin-ya K, Doi T. Total synthesis of largazole and its biological evaluation. Synlett 2008; 16: 2483-6.
    • (2008) Synlett , vol.16 , pp. 2483-2486
    • Numajiri, Y.1    Takahashi, T.2    Takagi, M.3    Shin-ya, K.4    Doi, T.5
  • 120
    • 55949099039 scopus 로고    scopus 로고
    • Enantioselective total synthesis of (+)-largazole, a potent inhibitor of histone deacetylase
    • Ghosh AK, Kulkarni S. Enantioselective total synthesis of (+)-largazole, a potent inhibitor of histone deacetylase. Org Lett 2008; 10: 3907-9.
    • (2008) Org Lett , vol.10 , pp. 3907-3909
    • Ghosh, A.K.1    Kulkarni, S.2
  • 121
    • 79951632653 scopus 로고    scopus 로고
    • Total syntheses of the histone deacetylase inhibitors largazole and 2-epi-largazole: Application of N-heterocyclic carbene mediated acylations in complex molecule synthesis
    • Wang B, Huang PH, Chen CS, Forsyth CJ. Total syntheses of the histone deacetylase inhibitors largazole and 2-epi-largazole: application of N-heterocyclic carbene mediated acylations in complex molecule synthesis. J Org Chem 2011; 76: 1140-50.
    • (2011) J Org Chem , vol.76 , pp. 1140-1150
    • Wang, B.1    Huang, P.H.2    Chen, C.S.3    Forsyth, C.J.4
  • 122
    • 55949094932 scopus 로고    scopus 로고
    • Synthesis and activity of largazole analogues with linker and macrocycle modification
    • Ying Y, Liu Y, Byeon SR, Kim H, Luesch H, Hong J. Synthesis and activity of largazole analogues with linker and macrocycle modification. Org Lett 2008; 10: 4021-4.
    • (2008) Org Lett , vol.10 , pp. 4021-4024
    • Ying, Y.1    Liu, Y.2    Byeon, S.R.3    Kim, H.4    Luesch, H.5    Hong, J.6
  • 123
    • 68149161259 scopus 로고    scopus 로고
    • Synthesis and biological evaluation of C7-demethyl largazole analogues
    • Chen F, Gao AH, Li J, Nan FJ. Synthesis and biological evaluation of C7-demethyl largazole analogues. Chem Med Chem 2009; 4: 1269-72.
    • (2009) Chem Med Chem , vol.4 , pp. 1269-1272
    • Chen, F.1    Gao, A.H.2    Li, J.3    Nan, F.J.4
  • 124
    • 64049106355 scopus 로고    scopus 로고
    • Synthesis and histone deacetylase inhibitory activity of largazole analogs: Alteration of the zinc-binding domain and macrocyclic scaffold
    • Bowers AA, West N, Newkirk TL, et al. Synthesis and histone deacetylase inhibitory activity of largazole analogs: alteration of the zinc-binding domain and macrocyclic scaffold. Org Lett 2009; 11: 1301-4.
    • (2009) Org Lett , vol.11 , pp. 1301-1304
    • Bowers, A.A.1    West, N.2    Newkirk, T.L.3
  • 125
    • 67749124307 scopus 로고    scopus 로고
    • Synthesis and conformation-activity relationships of the peptide isosteres of FK228 and largazole
    • Bowers AA, Greshock TJ, West N, et al. Synthesis and conformation-activity relationships of the peptide isosteres of FK228 and largazole. J Am Chem Soc 2009; 131: 2900-5.
    • (2009) J Am Chem Soc , vol.131 , pp. 2900-2905
    • Bowers, A.A.1    Greshock, T.J.2    West, N.3
  • 126
    • 77949831890 scopus 로고    scopus 로고
    • Total synthesis and biological evaluation of largazole and derivatives with promising selectivity for cancers cells
    • Zeng X, Yin B, Hu Z, et al. Total synthesis and biological evaluation of largazole and derivatives with promising selectivity for cancers cells. Org Lett 2010; 12: 1368-71.
    • (2010) Org Lett , vol.12 , pp. 1368-1371
    • Zeng, X.1    Yin, B.2    Hu, Z.3
  • 127
    • 78149267515 scopus 로고    scopus 로고
    • Anticolon cancer activity of largazole, a marine-derived tunable histone deacetylase inhibitor
    • Liu Y, Salvador LA, Byeon S, et al. Anticolon cancer activity of largazole, a marine-derived tunable histone deacetylase inhibitor. J Pharmacol Exp Ther 2010; 335: 351-61.
    • (2010) J Pharmacol Exp Ther , vol.335 , pp. 351-361
    • Liu, Y.1    Salvador, L.A.2    Byeon, S.3
  • 128
    • 0029347704 scopus 로고
    • Synthesis and cellular characterization of the detransformation agent, (-)-depudecin
    • Shimada J, Kwon HJ, Sawamura M, Schreiber S. Synthesis and cellular characterization of the detransformation agent, (-)-depudecin. Chem Biol 1995; 2: 517-25.
    • (1995) Chem Biol , vol.2 , pp. 517-525
    • Shimada, J.1    Kwon, H.J.2    Sawamura, M.3    Schreiber, S.4
  • 129
    • 0029932598 scopus 로고    scopus 로고
    • A mammalian histone deacetylase related to the yeast transcriptional regulator Rpd3p
    • Taunton J, Hassig CA, Schreiber SL. A mammalian histone deacetylase related to the yeast transcriptional regulator Rpd3p. Science 1996; 272: 408-11.
    • (1996) Science , vol.272 , pp. 408-411
    • Taunton, J.1    Hassig, C.A.2    Schreiber, S.L.3
  • 130
    • 0035694469 scopus 로고    scopus 로고
    • Acetylation of p53 activates transcription through recruitment of coactivators/histone acetyltransferases
    • Barlev NA, Liu L, Chehab NH, et al. Acetylation of p53 activates transcription through recruitment of coactivators/histone acetyltransferases. Mol Cell 2001; 8: 1243-54.
    • (2001) Mol Cell , vol.8 , pp. 1243-1254
    • Barlev, N.A.1    Liu, L.2    Chehab, N.H.3
  • 131
    • 0038491354 scopus 로고    scopus 로고
    • Identification and characterization of a novel p300-mediated p53 acetylation site, lysine 305
    • Wang YH, Tsay YG, Tan BC, Lo WY, Lee SC. Identification and characterization of a novel p300-mediated p53 acetylation site, lysine 305. J Biol Chem 2003; 278: 25568-76.
    • (2003) J Biol Chem , vol.278 , pp. 25568-25576
    • Wang, Y.H.1    Tsay, Y.G.2    Tan, B.C.3    Lo, W.Y.4    Lee, S.C.5
  • 132
    • 43049163953 scopus 로고    scopus 로고
    • Acetylation is indispensable for p53 activation
    • Tang Y, Zhao W, Chen Y, Zhao Y, Gu W. Acetylation is indispensable for p53 activation. Cell 2008; 133: 612-26.
    • (2008) Cell , vol.133 , pp. 612-626
    • Tang, Y.1    Zhao, W.2    Chen, Y.3    Zhao, Y.4    Gu, W.5
  • 133
    • 66449099052 scopus 로고    scopus 로고
    • p53 acetylation is crucial for its transcription-independent proapoptotic functions
    • Yamaguchi H, Woods NT, Piluso LG, et al. p53 acetylation is crucial for its transcription-independent proapoptotic functions. J Biol Chem 2009; 284: 11171-83.
    • (2009) J Biol Chem , vol.284 , pp. 11171-11183
    • Yamaguchi, H.1    Woods, N.T.2    Piluso, L.G.3
  • 135
    • 33645230001 scopus 로고    scopus 로고
    • Acetylation of p53 at lysine 373/382 by the histone deacetylase inhibitor depsipeptide induces expression of p21Waf1/Cip1
    • Zhao Y, Lu S, Wu L, et al. Acetylation of p53 at lysine 373/382 by the histone deacetylase inhibitor depsipeptide induces expression of p21Waf1/Cip1. Mol Cell Biol 2006; 26: 2782-90.
    • (2006) Mol Cell Biol , vol.26 , pp. 2782-2790
    • Zhao, Y.1    Lu, S.2    Wu, L.3
  • 136
    • 4143083971 scopus 로고    scopus 로고
    • Regulation at multiple levels of NF-κBmediated transactivation by protein acetylation
    • Quivy V, Van Lint C. Regulation at multiple levels of NF-κBmediated transactivation by protein acetylation. Biochem Pharmacol 2004; 68: 1221-9.
    • (2004) Biochem Pharmacol , vol.68 , pp. 1221-1229
    • Quivy, V.1    van Lint, C.2
  • 137
    • 24344468250 scopus 로고    scopus 로고
    • NF-κB RelA phosphorylation regulates RelA acetylation
    • Chen LF, Williams SA, Mu Y, et al. NF-κB RelA phosphorylation regulates RelA acetylation. Mol Cell Biol 2005; 25: 7966-75.
    • (2005) Mol Cell Biol , vol.25 , pp. 7966-7975
    • Chen, L.F.1    Williams, S.A.2    Mu, Y.3
  • 138
    • 0037462725 scopus 로고    scopus 로고
    • Post-activation turn-off of NF-κB-dependent transcription is regulated by acetylation of p65
    • Kiernan R, Brès V, Ng RW, et al. Post-activation turn-off of NF-κB-dependent transcription is regulated by acetylation of p65. J Biol Chem 2003; 278: 2758-66.
    • (2003) J Biol Chem , vol.278 , pp. 2758-2766
    • Kiernan, R.1    Brès, V.2    Ng, R.W.3
  • 139
    • 0346996870 scopus 로고    scopus 로고
    • Regulation of inducible Nitric Oxide Synthase expression by p300 and p50 acetylation
    • Deng WG, Wu KK. Regulation of inducible Nitric Oxide Synthase expression by p300 and p50 acetylation. J Immunol 2003; 171: 6581-8.
    • (2003) J Immunol , vol.171 , pp. 6581-6588
    • Deng, W.G.1    Wu, K.K.2
  • 140
    • 0038813706 scopus 로고    scopus 로고
    • Up-regulation of p300 binding and p50 acetylation in Tumor Necrosis Factor-α-induced Cyclooxygenase-2 promoter activation
    • Deng WG, Zhu Y, Wu KK. Up-regulation of p300 binding and p50 acetylation in Tumor Necrosis Factor-α-induced Cyclooxygenase-2 promoter activation. J Biol Chem 2003; 278: 4770-7.
    • (2003) J Biol Chem , vol.278 , pp. 4770-4777
    • Deng, W.G.1    Zhu, Y.2    Wu, K.K.3
  • 141
    • 0037416151 scopus 로고    scopus 로고
    • HDAC-6 interacts with and deacetylates tubulin and microtubules in vivo
    • Zhang Y, Li N, Caron C, et al. HDAC-6 interacts with and deacetylates tubulin and microtubules in vivo. EMBO 2003; 22: 1168-79.
    • (2003) EMBO , vol.22 , pp. 1168-1179
    • Zhang, Y.1    Li, N.2    Caron, C.3
  • 142
    • 0023389780 scopus 로고
    • Identification of an acetylation site of Chlamydomonas α-tubulin
    • LeDizet M, Piperno G. Identification of an acetylation site of Chlamydomonas α-tubulin. Proc Natl Acad Sci USA 1987; 84: 5720-4.
    • (1987) Proc Natl Acad Sci USA , vol.84 , pp. 5720-5724
    • Ledizet, M.1    Piperno, G.2
  • 143
    • 0023293040 scopus 로고
    • Microtubules containing acetylated α-tubulin in mammalian cells in culture
    • Piperno G, LeDizet M, Chang XJ. Microtubules containing acetylated α-tubulin in mammalian cells in culture. J Cell Biol 1987; 104: 289-302.
    • (1987) J Cell Biol , vol.104 , pp. 289-302
    • Piperno, G.1    Ledizet, M.2    Chang, X.J.3
  • 144
    • 0022452231 scopus 로고
    • The acetylation of Alpha-Tubulin and its relationship to the assembly and disassembly of microtubules
    • Maruta H, Greet K, Rosenbaum JL. The acetylation of Alpha-Tubulin and its relationship to the assembly and disassembly of microtubules. J Cell Biol 1986; 103: 571-9.
    • (1986) J Cell Biol , vol.103 , pp. 571-579
    • Maruta, H.1    Greet, K.2    Rosenbaum, J.L.3
  • 145
    • 0346020435 scopus 로고    scopus 로고
    • The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress
    • Kawaguchi Y, Kovacs JJ, McLaurin A, Vance JM, Ito A, Yao TP. The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell 2003; 115: 727-38.
    • (2003) Cell , vol.115 , pp. 727-738
    • Kawaguchi, Y.1    Kovacs, J.J.2    McLaurin, A.3    Vance, J.M.4    Ito, A.5    Yao, T.P.6
  • 146
    • 0037161744 scopus 로고    scopus 로고
    • HDAC6 is a microtubuleassociated deacetylase
    • Hubbert C, Guardiola A, Shao R, et al. HDAC6 is a microtubuleassociated deacetylase. Nature 2002; 417: 455-8.
    • (2002) Nature , vol.417 , pp. 455-458
    • Hubbert, C.1    Guardiola, A.2    Shao, R.3
  • 148
    • 12244251445 scopus 로고    scopus 로고
    • Stat3 dimerization regulated by reversible acetylation of a single lysine residue
    • Yuan ZL, Guan YJ, Chatterjee D, Chin YE. Stat3 dimerization regulated by reversible acetylation of a single lysine residue. Science 2005; 307: 269-73.
    • (2005) Science , vol.307 , pp. 269-273
    • Yuan, Z.L.1    Guan, Y.J.2    Chatterjee, D.3    Chin, Y.E.4
  • 149
    • 48349105443 scopus 로고    scopus 로고
    • Requirement of histone deacetylase1 (HDAC1) in signal transducer and activator of transcription 3 (STAT3) nucleocytoplasmic distribution
    • Ray S, Lee C, Hou T, Boldogh I, Brasier AR. Requirement of histone deacetylase1 (HDAC1) in signal transducer and activator of transcription 3 (STAT3) nucleocytoplasmic distribution. Nucleic Acids Res 2008; 36: 4510-20.
    • (2008) Nucleic Acids Res , vol.36 , pp. 4510-4520
    • Ray, S.1    Lee, C.2    Hou, T.3    Boldogh, I.4    Brasier, A.R.5
  • 150
    • 27744479712 scopus 로고    scopus 로고
    • Modulating molecular chaperone Hsp90 functions through reversible acetylation
    • Aoyagi S, Archer TK. Modulating molecular chaperone Hsp90 functions through reversible acetylation. Trends Cell Biol 2005; 15: 565-7.
    • (2005) Trends Cell Biol , vol.15 , pp. 565-567
    • Aoyagi, S.1    Archer, T.K.2
  • 151
    • 33846014703 scopus 로고    scopus 로고
    • An acetylation site in the middle domain of Hsp90 regulates chaperone function
    • Scroggins BT, Robzyk K, Wang D, et al. An acetylation site in the middle domain of Hsp90 regulates chaperone function. Mol Cell 2007; 25: 151-9.
    • (2007) Mol Cell , vol.25 , pp. 151-159
    • Scroggins, B.T.1    Robzyk, K.2    Wang, D.3
  • 152
    • 49649108912 scopus 로고    scopus 로고
    • Role of acetylation and extracellular location of Heat Shock Protein 90A in tumor cell invasion
    • Yang Y, Rao R, Shen J, et al. Role of acetylation and extracellular location of Heat Shock Protein 90A in tumor cell invasion. Cancer Res 2008; 68: 4833-42.
    • (2008) Cancer Res , vol.68 , pp. 4833-4842
    • Yang, Y.1    Rao, R.2    Shen, J.3
  • 153
    • 0034901985 scopus 로고    scopus 로고
    • Trichostatin A is a histone deacetylase inhibitor with potent antitumor activity against breast cancer in vivo
    • Vigushin DM, Ali S, Pace PE, et al. Trichostatin A is a histone deacetylase inhibitor with potent antitumor activity against breast cancer in vivo. Clin Cancer Res 2001; 7: 971-6.
    • (2001) Clin Cancer Res , vol.7 , pp. 971-976
    • Vigushin, D.M.1    Ali, S.2    Pace, P.E.3
  • 154
    • 4644314055 scopus 로고    scopus 로고
    • Plasma pharmacokinetics and metabolism of the histone deacetylase inhibitor trichostatin A after intraperitoneal administration to mice
    • Sanderson L, Taylor GW, Aboagye EO, et al. Plasma pharmacokinetics and metabolism of the histone deacetylase inhibitor trichostatin A after intraperitoneal administration to mice. Drug Met Disp 2004; 32: 1132-8.
    • (2004) Drug Met Disp , vol.32 , pp. 1132-1138
    • Sanderson, L.1    Taylor, G.W.2    Aboagye, E.O.3
  • 155
    • 67651170387 scopus 로고    scopus 로고
    • Apicidin induces apoptosis via cytochrome c-mediated intrinsic pathway in human ovarian cancer cells
    • Ahn MY, Na YJ, Lee J, Lee BM, Kim HS. Apicidin induces apoptosis via cytochrome c-mediated intrinsic pathway in human ovarian cancer cells. Biomol Ther 2009; 17: 17-24.
    • (2009) Biomol Ther , vol.17 , pp. 17-24
    • Ahn, M.Y.1    Na, Y.J.2    Lee, J.3    Lee, B.M.4    Kim, H.S.5
  • 156
    • 62549116922 scopus 로고    scopus 로고
    • Mechanism of apicidin-induced cell cycle arrest and apoptosis in Ishikawa human endometrial cancer cells
    • Ahn MY, Lee J, Na YJ, et al. Mechanism of apicidin-induced cell cycle arrest and apoptosis in Ishikawa human endometrial cancer cells. Chem Biol Interact 2009; 179: 169-77.
    • (2009) Chem Biol Interact , vol.179 , pp. 169-177
    • Ahn, M.Y.1    Lee, J.2    Na, Y.J.3
  • 157
    • 74549147514 scopus 로고    scopus 로고
    • Antitumor effect of apicidin on Ishikawa human endometrial cancer cells both in vitro and in vivo by blocking histone deacetylase 3 and 4
    • Ahn MY, Chung HY, Choi WS, Lee BM, Yoon S, Kim HS. Antitumor effect of apicidin on Ishikawa human endometrial cancer cells both in vitro and in vivo by blocking histone deacetylase 3 and 4. Int J Oncol 2010; 36: 125-31.
    • (2010) Int J Oncol , vol.36 , pp. 125-131
    • Ahn, M.Y.1    Chung, H.Y.2    Choi, W.S.3    Lee, B.M.4    Yoon, S.5    Kim, H.S.6
  • 158
    • 33645299037 scopus 로고    scopus 로고
    • Pharmacokinetics of a novel histone deacetylase inhibitor, apicidin, in rats
    • Shin BS, Chang HS, Park EH, et al. Pharmacokinetics of a novel histone deacetylase inhibitor, apicidin, in rats. Biopharm Drug Dispos 2006; 27: 69-75.
    • (2006) Biopharm Drug Dispos , vol.27 , pp. 69-75
    • Shin, B.S.1    Chang, H.S.2    Park, E.H.3
  • 159
    • 70349309607 scopus 로고    scopus 로고
    • Systemic cell-cycle suppression by apicidin, a histone deacetylase inhibitor, in MDA-MB-435 cells
    • Noh JH, Song JH, Eun JW, et al. Systemic cell-cycle suppression by apicidin, a histone deacetylase inhibitor, in MDA-MB-435 cells. Int J Mol Med 2009; 24: 205-26.
    • (2009) Int J Mol Med , vol.24 , pp. 205-226
    • Noh, J.H.1    Song, J.H.2    Eun, J.W.3
  • 160
    • 40849151859 scopus 로고    scopus 로고
    • Effects of FR235222, a novel HDAC inhibitor, in proliferation and apoptosis of human leukaemia cell lines: Role of annexin A1
    • Petrella A, D'Acunto CW, Rodriquez M, et al. Effects of FR235222, a novel HDAC inhibitor, in proliferation and apoptosis of human leukaemia cell lines: role of annexin A1. Eur J Cancer 2008; 44: 740-9.
    • (2008) Eur J Cancer , vol.44 , pp. 740-749
    • Petrella, A.1    D'Acunto, C.W.2    Rodriquez, M.3
  • 161
    • 77953610301 scopus 로고    scopus 로고
    • Histone deacetylase inhibitor FR235222 sensitizes human prostate adenocarcinoma cells to apoptosis through upregulation of annexin A1
    • D'Acunto CW, Fontanella B, Rodriquez M, Taddei M, Parente L, Petrella A. Histone deacetylase inhibitor FR235222 sensitizes human prostate adenocarcinoma cells to apoptosis through upregulation of annexin A1. Cancer Lett 2010; 295: 85-91.
    • (2010) Cancer Lett , vol.295 , pp. 85-91
    • D'Acunto, C.W.1    Fontanella, B.2    Rodriquez, M.3    Taddei, M.4    Parente, L.5    Petrella, A.6
  • 162
    • 77951034685 scopus 로고    scopus 로고
    • LGP1, a histone deacetylase inhibitor analogue of FR235222, sensitizes promyelocytic leukemia U937 cells to TRAIL-mediated apoptosis
    • D'Acunto CW, Carratu A, Rodriguez M, Taddei M, Parente L, Petrella A. LGP1, a histone deacetylase inhibitor analogue of FR235222, sensitizes promyelocytic leukemia U937 cells to TRAIL-mediated apoptosis. Anticancer Res 2010; 30: 887-94.
    • (2010) Anticancer Res , vol.30 , pp. 887-894
    • D'Acunto, C.W.1    Carratu, A.2    Rodriguez, M.3    Taddei, M.4    Parente, L.5    Petrella, A.6
  • 163
    • 0028267278 scopus 로고
    • FR901228, a novel antitumor bicyclic depsipeptide produced by Chromobacterium violaceum No. 968 III. Antitumor activities on experimental tumors in mice
    • Ueda H, Manda T, Matsumoto S, et al. FR901228, a novel antitumor bicyclic depsipeptide produced by Chromobacterium violaceum No. 968 III. Antitumor activities on experimental tumors in mice. J Antibiot 1994; 47: 315-23.
    • (1994) J Antibiot , vol.47 , pp. 315-323
    • Ueda, H.1    Manda, T.2    Matsumoto, S.3
  • 164
    • 0038060250 scopus 로고    scopus 로고
    • Effects of FK228, a novel histone deacetylase inhibitor, on tumor growth and expression of p21 and c-myc genes in vivo
    • Sasakawa Y, Naoe Y, Inoue T, et al. Effects of FK228, a novel histone deacetylase inhibitor, on tumor growth and expression of p21 and c-myc genes in vivo. Cancer Lett 2003; 195: 161-8.
    • (2003) Cancer Lett , vol.195 , pp. 161-168
    • Sasakawa, Y.1    Naoe, Y.2    Inoue, T.3
  • 165
    • 19444376494 scopus 로고    scopus 로고
    • Significant growth suppression of synovial sarcomas by the histone deacetylase inhibitor FK228 in vitro and in vivo
    • Ito T, Ouchida M, Morimoto Y, et al. Significant growth suppression of synovial sarcomas by the histone deacetylase inhibitor FK228 in vitro and in vivo. Cancer Lett 2005; 224: 311-9.
    • (2005) Cancer Lett , vol.224 , pp. 311-319
    • Ito, T.1    Ouchida, M.2    Morimoto, Y.3
  • 166
    • 12744251611 scopus 로고    scopus 로고
    • Marker genes to predict sensitivity to FK228, a histone deacetylase inhibitor
    • Sasakawa Y, Naoe Y, Sogo N, et al. Marker genes to predict sensitivity to FK228, a histone deacetylase inhibitor. Biochem Pharmacol 2005; 69: 603-16.
    • (2005) Biochem Pharmacol , vol.69 , pp. 603-616
    • Sasakawa, Y.1    Naoe, Y.2    Sogo, N.3
  • 167
    • 38449110249 scopus 로고    scopus 로고
    • Gene expression profiling induced by histone deacetylase inhibitor, FK228, in human esophageal squamous cancer cells
    • Hoshino I, Hisahiro M, Akutsu Y, et al. Gene expression profiling induced by histone deacetylase inhibitor, FK228, in human esophageal squamous cancer cells. Oncol Rep 2007; 18: 585-92.
    • (2007) Oncol Rep , vol.18 , pp. 585-592
    • Hoshino, I.1    Hisahiro, M.2    Akutsu, Y.3
  • 168
    • 51649114803 scopus 로고    scopus 로고
    • Proteomic analysis identifies protein targets responsible for depsipeptide sensitivity in tumor cells
    • Chen G, Li A, Zhao M, et al. Proteomic analysis identifies protein targets responsible for depsipeptide sensitivity in tumor cells. J Proteome Res 2008; 7: 2733-42.
    • (2008) J Proteome Res , vol.7 , pp. 2733-2742
    • Chen, G.1    Li, A.2    Zhao, M.3
  • 169
    • 35048856900 scopus 로고    scopus 로고
    • Depsipeptide (FK228) as a novel histone deacetylase inhibitor: Mechanism of action and anticancer activity
    • Lech-Maranda E, Robak E, Korycka A, Robak T. Depsipeptide (FK228) as a novel histone deacetylase inhibitor: mechanism of action and anticancer activity. Mini-Rev Med Chem 2007; 7: 1062-69.
    • (2007) Mini-Rev Med Chem , vol.7 , pp. 1062-1069
    • Lech-Maranda, E.1    Robak, E.2    Korycka, A.3    Robak, T.4
  • 170
    • 70350507928 scopus 로고    scopus 로고
    • Total synthesis of the bicyclic depsipeptide HDAC inhibitors spiruchostatins A and B, 5″-epi-spiruchostatin B, FK228 (FR901228) and preliminary evaluation of their biological activity
    • Narita K, Kikuchi T, Watanabe K, et al. Total synthesis of the bicyclic depsipeptide HDAC inhibitors spiruchostatins A and B, 5″-epi-spiruchostatin B, FK228 (FR901228) and preliminary evaluation of their biological activity. Chem Eur J 2009; 15: 11174-86.
    • (2009) Chem Eur J , vol.15 , pp. 11174-11186
    • Narita, K.1    Kikuchi, T.2    Watanabe, K.3
  • 171
    • 48149089435 scopus 로고    scopus 로고
    • Characterisation of the in vitro activity of the depsipeptide histone deacetylase inhibitor spiruchostatin A
    • Crabb SJ, Howell M, Rogers H, et al. Characterisation of the in vitro activity of the depsipeptide histone deacetylase inhibitor spiruchostatin A. Biochem Pharmacol 2008; 76: 463-75.
    • (2008) Biochem Pharmacol , vol.76 , pp. 463-475
    • Crabb, S.J.1    Howell, M.2    Rogers, H.3
  • 172
    • 79953127793 scopus 로고    scopus 로고
    • Next generation histone deacetylase inhibitors: The answer to the search for optimized epigenetic therapies?
    • Thaler F, Minucci S. Next generation histone deacetylase inhibitors: The answer to the search for optimized epigenetic therapies? Expert Opin Drug Discov 2011; 6: 393-404
    • (2011) Expert Opin Drug Discov , vol.6 , pp. 393-404
    • Thaler, F.1    Minucci, S.2


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.