174
Views
99
CrossRef citations to date
0
Altmetric
Review

Histone deacetylase inhibitors: insights into mechanisms of lethality

&
Pages 809-824 | Published online: 05 Aug 2005

Bibliography

  • O'DWYER ME, MAURO MJ, DRUKER BJ: STI571 as a targeted therapy for CML. Cancer Invest. (2003) 21:429–438.
  • KHAN AU, KRISHNAMURTHY S: Histone modifications as key regulators of transcription. Front Biosci. (2005) 10:866–872.
  • GREGORY PD, WAGNER K, HORZ W:Histone acetylation and chromatin remodeling. Exp. Cell Res. (2001) 265:195–202.
  • PEART MJ, SMYTH GK, VAN LAAR RK et al.: Identification and functional significance of genes regulated by structurally different histone deacetylase inhibitors. Proc. Natl. Acad. Sci. USA (2005) 102:3697–3702.
  • •Extensive analysis of HDAC inhibitor-mediated modulation of gene expression in relation to apoptosis and cell cycle regulation.
  • MILLER TA, WITTER DJ, BELVEDERE S: Histone deacetylase inhibitors./ Med. Chem. (2003) 46:5097–5116.
  • NEWMARK HL, YOUNG CW: Butyrate and phenylacetate as differentiating agents: practical problems and opportunities. J. Cell Biochem. SuppL (1995) 22:247–253.
  • ROSATO RR, WANG Z, GOPALKRISHNAN RV et aL: Evidence of a functional role for the cyclin-dependent kinase-inhibitor p21WAF1/CIP1/MDA6 in promoting differentiation and preventing mitochondrial dysfunction and apoptosis induced by sodium butyrate in human myelomonocytic leukemia cells (U937). Int. J. OncoL (2001) 19:181–191.
  • WEISBERG E, CATLEY L, KUJAWA J et al.: Histone deacetylase inhibitor NVP-LAQ824 has significant activity against myeloid leukemia cells in vitro and in vivo. Leukemia (2004) 18: 1951-1963.
  • ROMANSKI A, BACIC B, BUG G et aL: Use of a novel histone deacetylase inhibitor to induce apoptosis in cell lines of acute lymphoblastic leukemia. Haematologica (2004) 89:419–426.
  • GUO F, SIGUA C, TAO J et ell.: Cotreatment with histone deacetylase inhibitor LAQ824 enhances Apo-2L/tumor necrosis factor-related apoptosis inducing ligand-induced death inducing signaling complex activity and apoptosis of human acute leukemia cells. Cancer Res. (2004) 64:2580–2589.
  • •This work represents one of series of recent publications demonstrating the regulation of the expression of TRAIL receptors by HDACIs.
  • 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) 265:17174–17179.
  • FINNIN MS, DONIGIAN JR, COHEN A et al.: Structures of a histone deacetylase homologue bound to the TSA and SAHA inhibitors. Nature (1999) 401:188–193.
  • MARKS PA, MILLER T, RICHON VM: Histone deacetylases. Curr. Opin. Pharmacol. (2003) 3:344–351.
  • ROSATO RR, GRANTS: Histone deacetylase inhibitors in clinical development. Expert. Opin. Investig. Drugs (2004) 13:21–38.
  • DE RUIJTER AJ, KEMP S, KRAMER Get al.: The novel histone deacetylase inhibitor BL1521 inhibits proliferation and induces apoptosis in neuroblastoma cells. Biochem. PharmacoL (2004) 68:1279–1288.
  • VILLAR-GAREA A, ESTELLER M: Histone deacetylase inhibitors: understanding a new wave of anticancer agents. Int. J. Cancer (2004) 112:171–178.
  • DRUMMOND DC, NOBLE CO, KIRPOTIN DB et al.: Clinical Development of Histone Deacetylase Inhibitors As Anticancer Agents. Ann. Rev. PharmacoL lbxicoL (2004) 45:495–528.
  • GEORGE P, BALI P, ANNAVARAPU S et aL: Combination of the histone deacetylase inhibitor LBH589 and the hsp90 inhibitor 17-AAG is highly active against human CML-BC cells and AML cells with activating mutation of FLT-3.Blood (2005) 105: 1768-1776.
  • CANDIDO EP, REEVES R, DAVIE JR: Sodium butyrate inhibits histone deacetylation in cultured cells. Cell (1978) 14:105–113.
  • COUSENS LS, GALLWITZ D, ALBERTS BM: Different accessibilities in chromatin to histone acetylase. J. Biol. Chem. (1979) 254:1716–1723.
  • KRUH J: Effects of sodium butyrate, a new pharmacological agent, on cells in culture. MoL Cell Biochem. (1982) 42:65–82.
  • NUDELMAN A, GNIZI E, KATZ Yet al.: Prodrugs of butyric acid. Novel derivatives possessing increased aqueous solubility and potential for treating cancer and blood diseases. European J. Med. Chem. (2001) 36:63–74.
  • NUDELMAN A, REPHAELI A: Novel mutual prodrug of retinoic and butyric acids with enhanced anticancer activity. J. Med. Chem. (2000) 43:2962–2966.
  • REID T, VALONE F, LIPERA W et aL: Phase II trial of the histone deacetylase inhibitor pivaloyloxymethyl butyrate (Pivanex, AN-9) in advanced non-small cell lung cancer. Lung Cancer (2004) 45:381–386.
  • WITT 0, SCHWEIGERER L, DRIEVER PH et aL: Valproic acid treatment of glioblastoma multiforme in a child. Pediatr. Blood Cancer (2004) 43:181.
  • KELLY WK, O'CONNOR OA, MARKS PA: Histone deacetylase inhibitors: from target to clinical trials. Expert. Opin. Investig. Drugs (2002) 11:1695–1713.
  • PRAKASH S, FOSTER BJ, MEYER M et aL: Chronic oral administration of CI-994: a Phase I study. Invest. New Drugs (2001) 19:1–11.
  • ROSATO RR, ALMENARA JA, GRANTS: The histone deacetylase inhibitor MS-275 promotes differentiation or apoptosis in human leukemia cells through a process regulated by generation of reactive oxygen species and induction of p21CIP 1/WAF 11.Cancer Res. (2003) 63:3637–3645.
  • •This study emphasises the role that generation of ROS plays in the induction of cell death rather than differentiation by MS-275.
  • LORUSSO PM, DEMCHIK L, FOSTER B et aL: Preclinical antitumor activity of CI-994.Invest. New Drugs (1996) 14:349–356.
  • GRAZIANO MJ, SPOON TA, COCKRELL EA et al.: Induction of Apoptosis in Rat Peripheral Blood Lymphocytes by the Anticancer Drug CI-994 (Acetyldinaline)(*). j Biomed. BiotechnoL (2001) 1:52–61.
  • UNDEVIA SD, KINDLER HL, JANISCH Let 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.
  • NEMUNAITIS JJ, ORR D, EAGER 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.
  • CAMPHAUSEN K, SCOTT T, SPROULL M, TOFILON PJ: Enhancement of xenograft tumor radiosensitivity by the histone deacetylase inhibitor MS-275 and correlation with histone hyperacetylation. Clin. Cancer Res. (2004) 10:6066–6071.
  • MARKS PA, RIFKIND RA, RICHON VM et al.: Histone deacetylases and cancer: causes and therapies. Nature Reviews Cancer (2001) 1:194–202.
  • WANG C, FU M, MANI S et aL: Histone acetylation and the cell-cycle in cancer. Front Biosci. (2001) 6:D610–D629.
  • ACHARYA MR, FIGG WD: Histone deacetylase inhibitor enhances the anti-leukemic activity of an established nucleoside analogue. Cancer Biol. Ther. (2004) 3:719–720.
  • JOSE B, ONIKI Y, KATO T et al.: Novel histone deacetylase inhibitors: cyclic tetrapeptide with trifluoromethyl and pentafluoroethyl ketones. Bioorg. Med. Chem. Lett. (2004) 14:5343–5346.
  • NISHINO N, YOSHIKAWA D, WATANABE LA et al.: Synthesis and histone deacetylase inhibitory activity of cyclic tetrapeptides containing a retrohydroxamate as zinc ligand. Bioorg. Med. Chem. Lett. (2004) 14:2427–2431.
  • NISHINO N, JOSE B, OKAMURA S et al.: Cyclic tetrapeptides bearing a sulfhydryl group potently inhibit histone deacetylases. Org. Lett. (2003) 5:5079–5082.
  • MELNICK A, LIGHT JD: Histone deacetylases as therapeutic targets in hematologic malignancies. Curr. Opin. HematoL (2002) 9:322–332.
  • MINUCCI S, NERVI C, LO CF, PELICCI PG: Histone deacetylases: a common molecular target for differentiation treatment of acute myeloid leukemias? Oncogene (2001) 20:3110–3115.
  • JOHNSTONE RW, LICHT JD: Histone deacetylase inhibitors in cancer therapy: is transcription the primary target? Cancer Cell (2003) 4:13–18.
  • MARKS PA, JIANG X: Histone deacetylase inhibitors in programmed cell death and cancer therapy. Cell Cycle (2005) 4:e8–e10.
  • BHALLA K, LIST A: Histone deacetylase inhibitors in myelodysplastic syndrome. Best. Pract. Res. Clin. HaematoL (2004) 17:595–611.
  • ROSATO RR, GRANT S: Histone deacetylase inhibitors in cancer therapy. Cancer Biol. Ther. (2003) 2:30–37.
  • QIU L, KELSO MJ, HANSEN C et al.: Anti-tumour activity in vitro and in vivo of selective differentiating agents containing hydroxamate. Br. J. Cancer (1999) 80:1252–1258.
  • QIU L, BURGESS A, FAIRLIE DP et al.:Histone deacetylase inhibitors trigger a G2 checkpoint in normal cells that is defective in tumor cells. MoL Biol. Cell (2000) 11:2069–2083.
  • BUTLER LM, AGUS DB, SCHER HI et al.: Suberoylanilide hydroxamic acid, an inhibitor of histone deacetylase, suppresses the growth of prostate cancer cells in vitro and in vivo. Cancer Res. (2000) 60:5165–5170.
  • WARRENER R, BEAMISH H, BURGESS A et al.: Tumor cell-selective cytotoxicity by targeting cell cycle checkpoints. FASEB J. (2003) 17:1550–1552.
  • WADE PA: Transcriptional control at regulatory checkpoints by histone deacetylases: molecular connections between cancer and chromatin. Hum. Mol. Genet. (2001) 10:693–698.
  • JENUWEIN T, ALLIS CD: Translating the histone code. Science (2001) 293:1074–1080.
  • EGGER G, LIANG G, APARICIO A, JONES PA: Epigenetics in human disease and prospects for epigenetic therapy. Nature (2004) 429:457–463.
  • FORSBERG EC, BRESNICK EH: Histone acetylation beyond promoters: long-range acetylation patterns in the chromatin world. Bioessays (2001) 23:820–830.
  • STRAHL BD, ALLIS CD: The language of covalent histone modifications. Nature (2000) 403:41–45.
  • GRAY SG, TEH BT: Histone acetylation/deacetylation and cancer: an 'open' and 'shut' case? Curr. MoL Med. (2001) 1:401–429.
  • DE RUIJTER AJ, VAN GENNIP AH, CARON HN et al.: Histone deacetylases: characterisation of the classical HDAC family. Biochem. J. (2003) 370:737–749.
  • •This review provides an extensive description of the structure and function of HDACs.
  • GRAY SG, EKSTROM TJ: The human histone deacetylase family. Exp. Cell Res. (2001) 262:75–83.
  • ROTH SY, DENU JM, ALLIS CD: Histone acetyltransferases. Ann. Rev. Biochem. (2001) 70:81–120.
  • BLANDER G, GUARENTE L: The Sir2 family of protein deacetylases. Ann. Rev. Biochem. (2004) 73:417–435.
  • YANG XJ, GREGOIRE S: Class II histone deacetylases: from sequence to function, regulation, and clinical implication. MoL Cell. Biol. (2005) 25:2873–2884.
  • ANDERSON RM, LATORRE-ESTEVES M, NEVES AR et al.: Yeast life-span extension by calorie restriction is independent of NAD fluctuation. Science (2003) 302:2124–2126.
  • BORDONE L, GUARENTE L: Calorie restriction, SIRT1 and metabolism: understanding longevity. Nat. Rev. MoL Cell. Biol. (2005) 6:298–305.
  • GUARENTE L, PICARD F: Calorie restriction-the SIR2 connection. Cell (2005) 120:473–482.
  • KAO HY, VERDEL A, TSAI CC et aL: Mechanism for nucleocytoplasmic shuttling of histone deacetylase 7.j Biol. Chem. (2001) 276:47496–47507.
  • TIMMERMANN S, LEHRMANN H, POLESSKAYA A. HAREL-BELLAN k Histone acetylation and disease. Cell Mot Life Sci. (2001) 58:728–736.
  • KARPF AR, JONES DA: Reactivating the expression of methylation silenced genes in human cancer. Oncogene (2002) 21:5496–5503.
  • JONES PA, BAYLIN SB: The fundamental role of epigenetic events in cancer. Nat. Rev. Genet. (2002) 3:415–428.
  • ESTELLER M, CORDON-CARDO C, CORN PG et al.: p14ARF silencing by promoter hypermethylation mediates abnormal intracellular localization of MDM2.Cancer Res. (2001) 61:2816–2821.
  • CAMERON EE, BACHMAN KE, MYOHANEN S et al.: Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nat. Genet. (1999) 21:103–107.
  • ZHU WG, OTTERSON Gk The interaction of histone deacetylase inhibitors and DNA methyltransferase inhibitors in the treatment of human cancer cells. Curr. Med. Chem. Anti-Cane. Agents (2003) 3:187–199.
  • APARICIO A, WEBER JS: Review of theclinical experience with 5-azacytidine and 5-aza-2'-deoxycytidine in solid tumors. Curr. Opin. Investig Drugs (2002) 3:627–633.
  • INSINGA A, PELICCI PG, INUCCI S: Leukemia-associated fusion proteins. Multiple mechanisms of action to drive cell transformation. Cell Cycle (2005) 4:67–69.
  • HE LZ, TOLENTINO T, GRAYSON P et al.: Histone deacetylase inhibitors induce remission in transgenic models of therapy-resistant acute promyelocytic leukemia. J. Clin. Invest. (2001) 108:1321–1330.
  • JING Y, XIA L, WAXMAN S: Targeted removal of PML-RARalpha protein is required prior to inhibition of histone deacetylase for overcoming all-trans retinoic acid differentiation resistance in acute promyelocytic leukemia. Blood (2002) 100:1008–1013.
  • ZHOU DC, KIM SH, DING Wet aL: Frequent mutations in the ligand-binding domain of PML-RARalpha after multiple relapses of acute promyelocytic leukemia: analysis for functional relationship to response to all-trans retinoic acid and histone deacetylase inhibitors in vitro and in vivo. Blood (2002) 99:1356–1363.
  • VAN LINT C, EMILIANI S, VERDIN E: The expression of a small fraction of cellular genes is changed in response to histone hyperacetylation. Gene Expr. (1996) 5:245–253.
  • LEE H, LEE S, BAEK M et ell.: Expression profile analysis of trichostatin A in human gastric cancer cells. Biotech. Lett. (2002) 24:377–381.
  • BUTLER LM, ZHOU X, XU WS et al.: The histone deacetylase inhibitor SAHA arrests cancer cell growth, up-regulates thioredoxin-binding protein-2, and down-regulates thioredoxin. Proc. Natl. Acad. Sci. USA (2002) 99:11700–11705.
  • GLASER KB, STAVER MJ, WARING JF et al.: Gene expression profiling of multiple histone deacetylase (HDAC) inhibitors: defining a common gene set produced by HDAC inhibition in T24 and MDA carcinoma cell lines. Mol. Cancer Ther. (2003) 2:151–163.
  • LI H, WI] X: Histone deacetylase inhibitor, Trichostatin A, activates p21WAF1/CIP1 expression through downregulation of c-myc and release of the repression of c-myc from the promoter in human cervical cancer cells. Biochem. Biophys. Res. Commun. (2004) 324:860-867.This study provides useful insights into the mechanisms involved in the regulation of p21wArlicIP1 by HDACIs.
  • QIAN DZ, WANG X, KACHHAP SK et al.: The histone deacetylase inhibitor NVP-LAQ824 inhibits angiogenesis and has a greater antitumor effect in combination with the vascular endothelial growth factor receptor tyrosine kinase inhibitor PTK787/ZK222584.Cancer Res. (2004) 64:6626–6634.
  • MARKS Pk RICHON VM, MILLER T, KELLY WK: Histone deacetylase inhibitors. Adv. Cancer Res. (2004) 91:137–168.
  • BEHREND L, HENDERSON G, ZWACKA RM: Reactive oxygen species in oncogenic transformation. Biochem. Soc. Mans. (2003) 31:1441–1444.
  • IRANI K, XIA Y, ZWEIER JL et aL: Mitogenic signaling mediated by oxidants in Ras-transformed fibroblasts. Science (1997) 275:1649–1652.
  • CHUNG YM, BAE YS, LEE SY: Molecular ordering of ROS production, mitochondrial changes, and caspase activation during sodium salicylate-induced apoptosis. Free Radic. Biol. Med. (2003) 34:434–442.
  • CURTIN JF, DONOVAN M, COTTER TG: Regulation and measurement of oxidative stress in apoptosis. J. ImmunoL Methods (2002) 265:49–72.
  • CHEN QM, BARTHOLOMEW JC, CAMPISI J et al.: Molecular analysis of H202-induced senescent-like growth arrest in normal human fibroblasts: p53 and Rb control G1 arrest but not cell replication. Biochem. J. (1998) 332(Pt 1):43–50.
  • MOREIRA JM, SCHEIPERS P, SORENSEN P: The histone deacetylase inhibitor Trichostatin A modulates CD4+ T cell responses. BMC Cancer (2003) 3:30–47.
  • RUEFLI AA, AUSSERLECHNER MJ, BERNHARD D et al.: The histone deacetylase inhibitor and chemotherapeutic agent suberoylanilide hydroxamic acid (SAHA) induces a cell-death pathway characterized by cleavage of Bid and production of reactive oxygen species. Proc. NatL Acad. Sci. USA (2001) 98:10833-10838.One of the first studies identifying induction of oxidative damage as an important component of HDACI-mediated lethality.
  • LOUIS M, ROSATO RR, BRAULT L et al.: The histone deacetylase inhibitor sodium butyrate induces breast cancer cell apoptosis through diverse cytotoxic actions including glutathione depletion and oxidative stress. Int. J. Oncol. (2004) 25:1701–1711.
  • YU C, SUBLER M, RAHMANI M et al.:Induction of apoptosis in BCR/ABL+ cells by histone deacetylase inhibitors involves reciprocal effects on the RAF/MEK/ERK and JNK pathways. Cancer Biol. Ther. (2003) 2:544–551.
  • LUCAS DM, DAVIS ME, PARTHUN MR et al.: The histone deacetylase inhibitor MS-275 induces caspase-dependent apoptosis in B-cell chronic lymphocytic leukemia cells. Leukemia (2004) 18:1207–1214.
  • FERNANDEZ-CHECA JC: Redox regulation and signaling lipids in mitochondrial apoptosis. Biochem. Biophys. Res Commun. (2003) 304:471–479.
  • POWIS G, MONTFORT WR: Properties and biological activities of thioredoxins. Ann. Rev. PharmacoL ToxicoL (2001) 41:261–295.
  • FERNANDEZ-CHECA JC, KAPLOWITZ N, GARCIA-RUIZ C et aL: GSH transport in mitochondria: defense against TNF-induced oxidative stress and alcohol-induced defect. Am. J. Physiol (1997) 273:G7–17.
  • LOUIS M, ROSATO RR, BATTAGLIA E et al.: Modulation of sensitivity to doxorubicin by the histone deacetylase inhibitor sodium butyrate in breast cancer cells. Int. J. OncoL (2005) 26:1569–1574.
  • UNGERSTEDT JS, SOWA Y, XU WS et al.: Role of thioredoxin in the response of normal and transformed cells to histone deacetylase inhibitors. Proc. Natl. Acad. Sci. USA (2005) 102:673-678.Provides a theoretical basis for HDACI-associated selectivity by demonstrating that normal but not neoplastic cells respond to these agents by upregulating thioredoxin, resulting in escape from oxidative injury.
  • DICKINSON DA, FORMAN HJ: Glutathione in defense and signaling: lessons from a small thiol. Ann. N Y Acad. Sci. (2002) 973:488–504.
  • FILOMENI G, ROTILIO G, CIRIOLO MR: Glutathione disulfide induces apoptosis in U937 cells by a redox-mediated p38 MAP kinase pathway. FASEB J. (2003) 17:64–66.
  • ARNER ES, HOLMGREN A: Physiological functions of thioredoxin and thioredoxin reductase. Eur. j Biochem. (2000) 267:6102–6109.
  • KWON SH, AHN SH, KIM YK et al.: Apicidin, a histone deacetylase inhibitor, induces apoptosis and Fas/Fas ligand expression in human acute promyelocytic leukemia cells. J. Biol Chem. (2002) 277:2073–2080.
  • GLICK RD, SWENDEMAN SL, COFFEY DC et al.: Hybrid polar histone deacetylase inhibitor induces apoptosis and CD95/CD95 ligand expression in human neuroblastoma. Cancer Res. (1999) 59:4392–4399.
  • •This work was among the firsts to describe HDACI-mediated induction of proteins involved in the extrinsic, receptor-mediated apoptotic pathway.
  • KIM YH, PARK JW, LEE JY KWON TK: Sodium butyrate sensitizes TRAIL-mediated apoptosis by induction of transcription from the DR5 gene promoter through Spl sites in colon cancer cells. Carcinogenesis (2004) 25: 1813-1820.
  • WATANABE K, OKAMOTO K, YONEHARA S: Sensitization of osteosarcoma cells to death receptor-mediated apoptosis by HDAC inhibitors through downregulation of cellular FLIP. Cell Death Differ. (2005) 12:10–18.
  • NAKATA S, YOSHIDA T, HORINAKA M et al.: Histone deacetylase inhibitors upregulate death receptor 5/ TRAIL-R2 and sensitize apoptosis induced by TRAIL/AP02-L in human malignant tumor cells. Oncogene (2004) 23:6261–6271.
  • INSINGA A, MONESTIROLI S, RONZONI S et al.: Inhibitors of histone deacetylases induce tumor-selective apoptosis through activation of the death receptor pathway. Nat. Med. (2005) 11:71–76.
  • ••This and the following study implicateinduction of death receptor pathways as a possible basis for the selective toxicity of HDACIs towards neoplastic cells.
  • NEBBIOSO A, CLARKE N, VOLTZ E et al.: Tumor-selective action of HDAC inhibitors involves TRAIL induction in acute myeloid leukemia cells. Nat. Med. (2005) 11:77–84.
  • ••Important study demonstrating thatinduction of genes related to the TRAIL pathway may be also involved in the specificity of HDACI-induced lethality in cancer cells.
  • INOUE S, MACFARLANE M, HARPER N et al.: Histone deacetylase inhibitors potentiate TNF-related apoptosis-inducing ligand (TRAM-induced apoptosis in lymphoid malignancies. Cell Death Differ. (2004) 11 (Supp1.2): S193–S206.
  • CHOPIN V, SLOMIANNY C, HONDERMARCK H, LE B, X: Synergistic induction of apoptosis in breast cancer cells by cotreatment with butyrate and TNF-alpha, TRAIL, or anti-Fas agonist antibody involves enhancement of death receptors' signaling and requires P21(waf1). Exp. Cell Res. (2004) 298:560–573.
  • ROSATO RR, ALMENARA JA, DAI Y, GRANTS: Simultaneous activation of the intrinsic and extrinsic pathways by histone deacetylase (HDAC) inhibitors and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) synergistically induces mitochondrial damage and apoptosis in human leukemia cells. Mol Cancer Ther. (2003) 2:1273–1284.
  • SIRULNIK A, MELNICK A, ZELENT A, LICHT JD: Molecular pathogenesis of acute promyelocytic leukaemia and APL variants. Best. Pract. Res Clin Haematol (2003) 16:387–408.
  • MELNICK A, CARLILE GW, MCCONNELL MJ et al.: AML-1 /ETO fusion protein is a dominant negative inhibitor of transcriptional repression by the promyelocytic leukemia zinc finger protein. Blood (2000) 96:3939–3947.
  • DAI Y, RAHMANI M, GRANTS: An intact NF-lcappaB pathway is required for histone deacetylase inhibitor-induced G1 arrest and maturation in U937 human myeloid leukemia cells. Cell Cycle (2003) 2:467–472.
  • MAYO MW, DENLINGER CE, BROAD RM et al.: Ineffectiveness of histone deacetylase inhibitors to induce apoptosis involves the transcriptional activation of NF-kappa B through the Akt pathway. J. Biol. Chem. (2003) 278:18980–18989.
  • •This study provides evidence that activation of NF-x13 by HDACIs operating through an Akt-dependent pathway opposes the lethal actions of this class of agents.
  • CATLEY L, WEISBERG E, TM YT et al.: NVP-LAQ824 is a potent novel histone deacetylase inhibitor with significant activity against multiple myeloma. Blood (2003) 102:2615–2622.
  • RUNDALL BK, DENLINGER CE, JONES DR: Combined histone deacetylase and NF-lcappaB inhibition sensitizes non-small cell lung cancer to cell death. Surgery (2004) 136:416–425.
  • CHEN LF, GREENE WC: Shaping the nuclear action of NF-lcappaB. Nat. Rev. Mol Cell. Biol. (2004) 5:392–401.
  • GHOSH S, KARIN M: Missing pieces in the NF-lcappaB puzzle. Cell (2002) 109(Suppl.):S81–S96.
  • KARIN M, LIN A: NF-lcappaB at the crossroads of life and death. Nat. Immunol. (2002) 3:221–227.
  • LEZOUALC'H F, SAGARA Y, HOLSBOER F, BEHL C: High constitutive NF-lcappaB activity mediates resistance to oxidative stress in neuronal cells. J. Neurosci. (1998) 18:3224–3232.
  • STORZ P, TOKER A: Protein kinase D mediates a stress-induced NF-kappaB activation and survival pathway. EMBO J. (2003) 22:109–120.
  • QUIVY V, VAN LINT C: Regulation at multiple levels of NF-lcappaB-mediated transactivation by protein acetylation. Biochem. PharmacoL (2004) 68:1221–1229.
  • ORLOWSKI RZ, BALDWIN AS, JR.: NF-lcappaB as a therapeutic target in cancer. Trends MoL Med. (2002) 8:385–389.
  • FRENCH LE, TSCHOPP J: The TRAIL to selective tumor death. Nat. Med. (1999) 5:146–147.
  • BALDWIN AS: Control of oncogenesis and cancer therapy resistance by the transcription factor NF-kappaB. Clin. Invest. (2001) 107:241–246.
  • AMIT S, BEN NERIAH NF-lcappaB activation in cancer: a challenge for ubiquitination- and proteasome-based therapeutic approach. Semin. Cancer Biol. (2003) 13:15–28.
  • STEHLIK C, DE MARTIN R, KUMABASHIRI I et al.: Nuclear factor (NF)-kappaB-regulated X chromosome-linked iap gene expression protects endothelial cells from tumor necrosis factor alpha-induced apoptosis. J. Exp. Med. (1998) 188:211–216.
  • GRUMONT RJ, ROURKE IJ, GERONDAKIS S: Rd-dependent induction of Al transcription is required (1999) 13:400–411.
  • ZONG WX, EDELSTEIN LC, CHEN C et al.: The prosurvival Bc1-2 homolog Bfl-1/ Al is a direct transcriptional target of NF-lcappaB that blocks TNFalpha-induced
  • KREUZ S, SIEGMUND D, SCHEURICH P, WAJANT H: NF-lcappaB inducers upregulate cFLIP, a damage and apoptosis in human leukemia cells by flavopiridol and the histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA). Leukemia (2002) 16:1331–1343.
  • DE AW, JR., MUELLER-DIECKMANN HJ, SCHULZE-GAHMEN U et aL: Structural basis for specificity and potency of a flavonoid inhibitor of human CDK2, a cell cycle kinase. Proc. Nail. Acad. Sci. USA (1996) 93:2735–2740.
  • CHAO SH, FUJINAGA K, MARION JE et al.: Flavopiridol inhibits P-TEFB and blocks HIV-1 replication. J. Biol. Chem. (2000) 275:28345–28348.
  • CHAO SH, PRICE DH: Flavopiridol inactivates P-TEFb and blocks most RNA polymerase II transcription in vivo. J. Biol. Chem. (2001) 276:31793–31799.
  • SAVICKIENE J, TREIGYTE G, PIVORIUNAS A et al.: Spl and NF-{lcappa}B transcription factor activity in the regulation of the p21 and FasL promoters during promyelocytic leukemia cell monocytic differentiation and its associated apoptosis. Ann. N Y Acad. Sci. (2004) 1030:569-577.This study investigated the activity of Spl and NF-x13 in relation to the control of p21/WAF1/CIP1 and FasL genes during HDACI-mediated monocytic differentiation and apoptosis.
  • TANG G, MINEMOTO Y, DIBLING B et al.: Inhibition of JNK activation through NF-lcappaB target genes. Nature (2001) 414:313–317.
  • TAKADA Y, AGGARWAL BB: Flavopiridol inhibits NF-lcappaB activation induced by various carcinogens and inflammatory agents through inhibition of IlcappaBalpha kinase and p65 phosphorylation: abrogation of cyclin D1, cyclooxygenase-2, and matrix metalloprotease-9.j Biol. Chem. (2004) 279:4750–4759.
  • CARTEE L, WANG Z, DECKER RH et al.: The cyclin-dependent kinase inhibitor (CDKI) flavopiridol disrupts phorbol 12-myristate 13-acetate-induced differentiation and CDKI expression while enhancing apoptosis in human myeloid leukemia cells. Cancer Res. (2001) 61:2583–2591.
  • KITADA S, ZAPATA JM, ANDREEFF M, REED JC: Protein kinase inhibitors flavopiridol and 7-hydroxy-staurosporine down-regulate antiapoptosis proteins in B-cell chronic lymphocytic leukemia. Blood (2000) 96:393–397.
  • ROSATO RR, DM Y, ALMENARA JA et al.: Potent antileukemic interactions between flavopiridol and TRAIL/Apo2L involve flavopiridol-mediated XIAP downregulation. Leukemia (2004) 18: 1780-1788.
  • NGUYEN DM, SCHRUMP WD, TSAI WS et al.: Enhancement of depsipeptide-mediated apoptosis of lung or esophageal cancer cells by flavopiridol: activation of the mitochondria-dependent death-signaling pathway. J. Thorac. Cardiovasc. Surg. (2003) 125:1132–1142.
  • NGUYEN DM, SCHRUMP WD, CHEN GA et al.: Abrogation of p21 expression by flavopiridol enhances depsipeptide-mediated apoptosis in malignant pleural mesothelioma cells. Clin. Cancer Res. (2004) 10:1813–1825.
  • DENLINGER CE, KELLER MD, MAYO MW et al.: Combined proteasome and histone deacetylase inhibition in non-small cell lung cancer. J. Thorac. Cardiovasc. Surg. (2004) 127:1078–1086.
  • YU C, RAHMANI M, CONRAD D et al.: The proteasome inhibitor bortezomib interacts synergistically with histone deacetylase inhibitors to induce apoptosis in Bcr/Abl+ cells sensitive and resistant to 5T1571.Blood (2003) 102:3765–3774.
  • PEI XY, DAI Y, GRANT S: Synergistic induction of oxidative injury and apoptosis in human multiple myeloma cells by the proteasome inhibitor bortezomib and histone deacetylase inhibitors. Clin. Cancer Res. (2004) 10:3839–3852.
  • YEUNG F, HOBERG JE, RAMSEY CS et al.: Modulation of NF-lcappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J. (2004) 23:2369–2380.
  • OGRETMEN B, HANNUN YA: Biologically active sphingolipids in cancer pathogenesis and treatment. Nat. Rev. Cancer (2004) 4:604–616.
  • BEKTAS M, SPIEGEL S: Glycosphingolipids and cell death. Glycoconj. J. (2004) 20:39–47.
  • HANNUN YA, OBEID LM: The Ceramide-centric universe of lipid-mediated cell regulation: stress encounters of the lipid kind. J. Biol. Chem. (2002) 277:25847–25850.
  • CATZ SD, JOHNSON JL: Transcriptional Oncogene (2001) 20:7342–7351.
  • SUZUKI M, SHINOHARA F, SATO K et al.: Interleukin-1 betaconverting enzyme subfamily inhibitors prevent induction of CD86 molecules by butyrate through a CREB-dependent mechanism in HL60 cells. Immunology (2003) 108:375–383.
  • GAO N, DAI Y, RAHMANI M et al.: Contribution of disruption of the nuclear factor-lcappaB pathway to induction of apoptosis in human leukemia cells by (2004) 66:956–963.
  • YIN L, LAEVSKY G, GIARDINA C: Butyrate suppression of colonocyte NF-kappa B activation and cellular proteasome activity. J. Biol. Chem. (2001) 276:44641–44646.
  • MORI N, MATSUDA T, TADANO M et al.: Apoptosis induced by the histone deacetylase inhibitor FR901228 in human T-cell leukemia virus Type 1-infected T-cell lines and primary adult T-cell leukemia
  • ROSATO RR, ALMENARA JA, CARTEE Let al.: The cyclin-dependent kinase inhibitor flavopiridol disrupts sodium butyrate-induced p21WAF1/CIP1 expression and maturation while reciprocally potentiating apoptosis in human leukemia cells. MoL
  • ALMENARA J, ROSATO R, GRANTS: Synergistic induction of mitochondrial
  • PAYNE SG, MILSTIEN S, SPIEGEL S: Sphingosine-l-phosphate: dual messenger functions. FEBS Lett. (2002) 531:54–57.
  • PETTUS BJ, BIELAWSKI J, PORCELLI AM et al.: The sphingosine kinase 1/sphingosine-1-phosphate pathway mediates COX-2 induction and PGE2 production in response to TNF-alpha. FASEB J. (2003) 17:1411–1421.
  • STUNFF HL, MILSTIEN S, SPIEGEL S: Generation and metabolism of bioactive sphingosine-l-phosphate. J. Cell Biochem. (2004) 92:882–899.
  • DING WX, YIN XM: Dissection of the multiple mechanisms of TNF-alpha-induced apoptosis in liver injury. J. Cell Md. Med. (2004) 8:445–454.
  • BOSE R, VERHEIJ M, HAIMOVITZ-FRIEDMAN A et al: Ceramide synthase mediates daunorubicin-induced apoptosis: an alternative mechanism for generating death signals. Cell (1995) 82:405–414.
  • PERRY DK, CARTON J, SHAH AK et al: Serine palmitoyltransferase regulates de novo ceramide generation during etoposide-induced apoptosis. J. Biol. Chem. (2000) 275:9078–9084.
  • CHAUVIER D, MORJANI H, MANFAIT M: Ceramide involvement in homocamptothecin- and camptothecin-induced cytotoxicity and apoptosis in colon HT29 cells. Int. J. Oncol (2002) 20:855–863.
  • BISWAL SS, DATTA K, ACQUAAH-MENSAH GK, KEHRER JP: Changes in ceramide and sphingomyelin following fludarabine treatment of human chronic B-cell leukemia cells. Toxicology (2000) 154:45–53.
  • KOLESNICK RN, KRONKE M: Regulation of ceramide production and apoptosis. Ann. Rev. Physiol (1998) 60:643–665.
  • MORALES A, COLELL A, MARI M et al: Glycosphingolipids and mitochondria: role in apoptosis and disease. G/yeoconj J. (2004) 20:579–588.
  • KURITA-OCHIAI T, AMANO S, FUKUSHIMA K, OCHIAI K: Cellular Events Involved in Butyric Acid-Induced T Cell Apoptosis. j Immunol. (2003) 171:3576–3584.
  • MAGGIO SC, ROSATO RR, KRAMER LB et al: The histone deacetylase inhibitor MS-275 interacts synergistically with fludarabine to induce apoptosis in human leukemia cells.Cancer Res. (2004) 64:2590–2600.
  • •Demonstrated the involvement of ceramide in the mechanism of HDACI-induced lethality in human leukaemia cells.
  • RAHMANI M, REESE E, DAI Yet al: Coadministration of Histone Deacetylase Inhibitors and Perifosine Synergistically Induces Apoptosis in Human Leukemia Cells through Akt and ERK1/2 Inactivation and the Generation of Ceramide and Reactive Oxygen Species. Cancer Res. (2005) 65:2422–2432.
  • EICKHOFF B, RULLER S, LAUE T et al.: Trichostatin A modulates expression of p2lwafl/cipl, Bc1-xL, ID1, ID2, ID3, CRAB2, GATA-2, hsp86 and TFIID/ TAFII31 mRNA in human lung adenocarcinoma cells. Biol. Chem. (2000) 381:107–112.
  • EICKHOFF B, GERMEROTH L, STAHL C et al: Trichostatin A-mediated regulation of gene expression and protein kinase activities: reprogramming tumor cells for ribotoxic stress-induced apoptosis. Biol. Chem. (2000) 381:1127–1132.
  • SREEDHAR AS, CSERMELY P: Heat shock proteins in the regulation of apoptosis: new strategies in tumor therapy: a comprehensive review. Pharmacol Ther. (2004) 101:227–257.
  • BLAGOSKLONNY MV: Hsp-90-associated oncoproteins: multiple targets of geldanamycin and its analogs. Leukemia (2002) 16:455–462.
  • KIMURA E, ENNS RE, ALCARAZ JE et al.: Correlation of the survival of ovarian cancer patients with mRNA expression of the 60-1cD heat-shock protein HSP-60.J. Clin. Oncol (1993) 11:891–898.
  • CIOCCA DR, CLARK GM, TANDON AK et al: Heat shock protein hsp70 in patients with axillary lymph node-negative breast cancer: prognostic implications. J. Natl. Cancer Inst. (1993) 85:570–574.
  • SANTAROSA M, FAVARO D, QUAIA M, GALLIGIONI E: Expression of heat shock protein 72 in renal cell carcinoma: possible role and prognostic implications in cancer patients. Eur. .1.Cancer (1997) 33:873–877.
  • TAKAYAMA S, REED JC, HOMMA S: Heat-shock proteins as regulators of apoptosis. Oncogene (2003) 22:9041–9047.
  • BAGATELL R, WHITESELL L: Altered Hsp90 function in cancer: a unique therapeutic opportunity. MoL Cancer Ther. (2004) 3:1021–1030.
  • CREAGH EM, SHEEHAN D, COTTER TG: Heat shock proteins-modulators of apoptosis in tumour cells. Leukemia (2000) 14:1161–1173.
  • BELIAKOFF J, WHITESELL L: Hsp90: an emerging target for breast cancer therapy. Anticancer Drugs (2004) 15:651–662.
  • YU X, GUO ZS, MARCU MG et al: Modulation of p53, ErbB1, ErbB2, and Raf-1 expression in lung cancer cells by depsipeptide FR901228.J. Natl. Cancer Inst. (2002) 94:504–513.
  • •One of the first reports demonstrating disruption of Hsp90 function by HDAC-mediated acetylation.
  • NECKERS L, SCHULTE TW, MIMNAUGH E: Geldanamycin as a potential anti-cancer agent: its molecular target and biochemical activity. Invest. New Drugs (1999) 17:361–373.
  • CITRI A. ALROY I, LAW S et al: Drug-induced ubiquitylation and degradation of ErbB receptor tyrosine kinases: implications for cancer therapy. EMBO J. (2002) 21:2407–2417.
  • FUINO L, BALI P, WITTMANN S et al: Histone deacetylase inhibitor LAQ824 down-regulates Her-2 and sensitizes human breast cancer cells to trastuzumab, taxotere, gemcitabine, and epothilone B. MoL Cancer Ther. (2003) 2:971–984.
  • NIMMANAPALLI R, FUINO L, BALI P et al: Histone deacetylase inhibitor LAQ824 both lowers expression and promotes proteasomal degradation of Bcr-Abl and induces apoptosis of imatinib mesylate-sensitive or -refractory chronic myelogenous leukemia-blast crisis cells. Cancer Res. (2003) 63:5126–5135.
  • •This study demonstrates that HDACI-mediated disruption of Hsp90 function can promote degradation of mutant oncogenic proteins such as Bcr/Abl and in so doing, cooperate with specific kinase inhibitors to enhance leukaemic cell killing.
  • RAHMANI M, YU C, DAI Yet al: Co-administration of the heat shock protein 90 antagonist 17-AAG with SAHA or sodium butyrate synergistically induces apoptosis in human leukemia cells. Cancer Res. (2003) 63:8420–8427.
  • RAHMANI M, REESE E, DAI Y et ell.: Co-treatment with SAHA and 17-AAG synergistically induces apoptosis in Bcr-Abl+ cells sensitive and resistant to STI-571 in association with down-regulation of Bcr-Abl, abrogation of STAT5 activity, and Bax conformational change. Mol. PharmacoL (2005) 67:1166–1176.
  • BECK J, FISCHER T, ROWINSKY E et al.: Phase I pharmacokinetic (PK) and pharmacodynamic (PD) study of LBH589A: A novel histone deacetylase inhibitor. (2004)
  • POROSNICU M, NIMMANAPALLI R, NGUYEN D et al.: Co-treatment with As203 enhances selective cytotoxic effects of STI-571 against Brc-Abl-positive acute leukemia cells. Leukemia (2001) 15:772–778.
  • BALI P, GUO F, FISKUS Wet al.: Mechanisms underlying hydroxamic acid analog (HA) histone deacetylase (HDAC) inhibitors (HDIs)-induced apoptosis: New role of HDAC6 inhibition, acetylation and inhibition of Hsp90 and depletion of pro-growth and pro-survival oncoproteins. Proc. Amer. Assoc. Cancer Res. (2005) 46:769 Abstract 3268.
  • KOVACS JJ, MURPHY PJ, GAILLARD S et al.: HDAC6 regulates Hsp90 acetylation and chaperone-dependent activation of glucocorticoid receptor. MoL Cell (2005) 18:601–607.
  • HUBBERT C, GUARDIOLA A, SHAO R et al.: HDAC6 is a microtubule-associated deacetylase. Nature (2002) 417:455–458.
  • MATSUYAMA A, SHIMAZU T, SUMIDA Yet al.: In vivo destabilization of dynamic microtubules by HDAC6-mediated deacetylation. EMBO J. (2002) 21:6820–6831.
  • HAGGARTY SJ, KOELLER KM, WONG JC et al.: Domain-selective small-molecule inhibitor of histone deacetylase 6 (HDAC6)-mediated tubulin deacetylation. Proc. NatL Acad. Sci. USA (2003) 100:4389–4394.
  • KAWAGUCHI Y, KOVACS JJ, MCLAURIN A et al.: The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell (2003) 115:727–738.
  • •This was the first study to demonstrate that HDAC6 plays a critical role in aggresome formation through the acetylation of dynein, and by extension, the orderly disposition of misfolded proteins.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.