Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer Ther ; 5(12): 3096-104, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17172412

ABSTRACT

Human enhancer of zeste 2 (EZH2) protein belongs to the multiprotein polycomb repressive complex 2, which also includes suppressor of zeste 12 (SUZ12) and embryonic ectoderm development (EED). The polycomb repressive complex 2 complex possesses histone methyltransferase activity mediated by the Su(var)3-9, enhancer of zeste, and trithorax domain of EZH2, which methylates histone H3 on lysine (K)-27 (H3K27). In the present studies, we determined that treatment with the hydroxamate histone deacetylase inhibitor LBH589 or LAQ824 depleted the protein levels of EZH2, SUZ12, and EED in the cultured (K562, U937, and HL-60) and primary human acute leukemia cells. This was associated with decreased levels of trimethylated and dimethylated H3K27, with concomitant depletion of the homeobox domain containing HOXA9 and of MEIS1 transcription factors. Knockdown of EZH2 by EZH2 small interfering RNA also depleted SUZ12 and EED, inhibited histone methyltransferase activity, and reduced trimethylated and dimethylated H3K27 levels, with a concomitant loss of clonogenic survival of the cultured acute myelogenous leukemia (AML) cells. EZH2 small interfering RNA sensitized the AML cells to LBH589-mediated depletion of EZH2, SUZ12, and EED; loss of clonogenic survival; and LBH589-induced differentiation of the AML cells. These findings support the rationale to test anti-EZH2 treatment combined with hydroxamate histone deacetylase inhibitors as an antileukemia epigenetic therapy, especially against AML with coexpression of EZH2, HOXA9, and MEIS1 genes.


Subject(s)
DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Leukemia/drug therapy , Leukemia/metabolism , Repressor Proteins/genetics , Transcription Factors/metabolism , Acetylation/drug effects , Acute Disease , Carrier Proteins/genetics , Chronic Disease , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Down-Regulation/drug effects , Enhancer of Zeste Homolog 2 Protein , HL-60 Cells , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Indoles , K562 Cells , Leukemia/enzymology , Leukemia/genetics , Neoplasm Proteins , Nuclear Proteins/genetics , Panobinostat , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Protein Methyltransferases , RNA, Small Interfering/genetics , Repressor Proteins/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , U937 Cells
2.
Clin Cancer Res ; 12(19): 5869-78, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-17020995

ABSTRACT

PURPOSE: We determined the effects of vorinostat [suberoylanilide hydroxamic acid (SAHA)] and/or dasatinib, a dual Abl/Src kinase (tyrosine kinase) inhibitor, on the cultured human (K562 and LAMA-84) or primary chronic myelogenous leukemia (CML) cells, as well as on the murine pro-B BaF3 cells with ectopic expression of the unmutated and kinase domain-mutant forms of Bcr-Abl. EXPERIMENTAL DESIGN: Following exposure to dasatinib and/or vorinostat, apoptosis, loss of clonogenic survival, as well as the activity and levels of Bcr-Abl and its downstream signaling proteins were determined. RESULTS: Treatment with dasatinib attenuated the levels of autophosphorylated Bcr-Abl, p-CrkL, phospho-signal transducer and activator of transcription 5 (p-STAT5), p-c-Src, and p-Lyn; inhibited the activity of Lyn and c-Src; and induced apoptosis of the cultured CML cells. Combined treatment of cultured human CML and BaF3 cells with vorinostat and dasatinib induced more apoptosis than either agent alone, as well as synergistically induced loss of clonogenic survival, which was associated with greater depletion of Bcr-Abl, p-CrkL, and p-STAT5 levels. Cotreatment with dasatinib and vorinostat also attenuated the levels of Bcr-AblE255K and Bcr-AblT315I and induced apoptosis of BaF3 cells with ectopic expression of the mutant forms of Bcr-Abl. Finally, cotreatment of the primary CML cells with vorinostat and dasatinib induced more loss of cell viability and depleted Bcr-Abl or Bcr-AblT315I, p-STAT5, and p-CrkL levels than either agent alone. CONCLUSIONS: As shown here, the preclinical in vitro activity of vorinostat and dasatinib against cultured and primary CML cells supports the in vivo testing of the combination in imatinib mesylate-sensitive and imatinib mesylate-resistant CML cells.


Subject(s)
Apoptosis/drug effects , Drug Resistance, Neoplasm , Fusion Proteins, bcr-abl/antagonists & inhibitors , Hydroxamic Acids/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols , Benzamides , Dasatinib , Drug Synergism , Fusion Proteins, bcr-abl/metabolism , Histone Deacetylase Inhibitors , Humans , Imatinib Mesylate , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , STAT5 Transcription Factor/metabolism , Tumor Cells, Cultured , Vorinostat , src-Family Kinases/metabolism
3.
Blood ; 108(2): 645-52, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16537804

ABSTRACT

AMN107 (Novartis Pharmaceuticals, Basel, Switzerland) has potent in vitro and in vivo activity against the unmutated and most common mutant forms of Bcr-Abl. Treatment with the histone deacetylase inhibitor LBH589 (Novartis) depletes Bcr-Abl levels. We determined the effects of AMN107 and/or LBH589 in Bcr-Abl-expressing human K562 and LAMA-84 cells, as well as in primary chronic myelogenous leukemia (CML) cells. AMN107 was more potent than imatinib mesylate (IM) in inhibiting Bcr-Abl tyrosine kinase (TK) activity and attenuating p-STAT5, p-AKT, Bcl-x(L), and c-Myc levels in K562 and LAMA-84 cells. Cotreatment with LBH589 and AMN107 exerted synergistic apoptotic effects with more attenuation of p-STAT5, p-ERK1/2, c-Myc, and Bcl-x(L) and increases in p27 and Bim levels. LBH589 attenuated Bcr-Abl levels and induced apoptosis of mouse pro-B BaF3 cells containing ectopic expression of Bcr-Abl or the IM-resistant, point-mutant Bcr-AblT315I and Bcr-AblE255K. Treatment with LBH589 also depleted Bcr-Abl levels and induced apoptosis of IM-resistant primary human CML cells, including those with expression of Bcr-AblT315I. As compared with either agent alone, cotreatment with AMN107 and LBH589 induced more loss of cell viability of primary IM-resistant CML cells. Thus, cotreatment with LBH589 and AMN107 is active against cultured or primary IM-resistant CML cells, including those with expression of Bcr-AblT315I.


Subject(s)
Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia/pathology , Pyrimidines/pharmacology , Animals , Apoptosis/drug effects , Benzamides , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Synergism , Histone Deacetylase Inhibitors , Humans , Hydroxamic Acids/pharmacology , Imatinib Mesylate , Indoles , Leukemia/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Panobinostat , Piperazines/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Tumor Cells, Cultured
4.
Cancer Res ; 65(22): 10536-44, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16288046

ABSTRACT

17-Allylamino-demethoxy geldanamycin (17-AAG) inhibits the chaperone association of heat shock protein 90 (hsp90) with the heat shock factor-1 (HSF-1), which induces the mRNA and protein levels of hsp70. Increased hsp70 levels inhibit death receptor and mitochondria-initiated signaling for apoptosis. Here, we show that ectopic overexpression of hsp70 in human acute myelogenous leukemia HL-60 cells (HL-60/hsp70) and high endogenous hsp70 levels in Bcr-Abl-expressing cultured CML-BC K562 cells confers resistance to 17-AAG-induced apoptosis. In HL-60/hsp70 cells, hsp70 was bound to Bax, inhibited 17-AAG-mediated Bax conformation change and mitochondrial localization, thereby inhibiting the mitochondria-initiated events of apoptosis. Treatment with 17-AAG attenuated the levels of phospho-AKT, AKT, and c-Raf but increased hsp70 levels to a similar extent in the control HL-60/Neo and HL-60/hsp70 cells. Pretreatment with 17-AAG, which induced hsp70, inhibited 1-beta-D-arabinofuranosylcytosine or etoposide-induced apoptosis in HL-60 cells. Stable transfection of a small interfering RNA (siRNA) to hsp70 completely abrogated the endogenous levels of hsp70 and blocked 17-AAG-mediated hsp70 induction, resulting in sensitizing K562/siRNA-hsp70 cells to 17-AAG-induced apoptosis. This was associated with decreased binding of Bax to hsp70 and increased 17-AAG-induced Bax conformation change. 17-AAG-mediated decline in the levels of AKT, c-Raf, and Bcr-Abl was similar in K562 and K562/siRNA-hsp70 cells. Cotreatment with KNK437, a benzylidine lactam inhibitor of hsp70 induction and thermotolerance, attenuated 17-AAG-mediated hsp70 induction and increased 17-AAG-induced apoptosis and loss of clonogenic survival of HL-60 cells. Collectively, these data indicate that induction of hsp70 attenuates the apoptotic effects of 17-AAG, and abrogation of hsp70 induction significantly enhances the antileukemia activity of 17-AAG.


Subject(s)
HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/biosynthesis , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Rifabutin/analogs & derivatives , Apoptosis/drug effects , Benzhydryl Compounds/pharmacology , Benzoquinones , Combined Modality Therapy , Cytarabine/pharmacology , Drug Synergism , Etoposide/pharmacology , HL-60 Cells , HSP70 Heat-Shock Proteins/genetics , Humans , K562 Cells , Lactams, Macrocyclic , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Protein Conformation/drug effects , Pyrrolidinones/pharmacology , RNA, Small Interfering/genetics , Rifabutin/pharmacology , bcl-2-Associated X Protein/metabolism
5.
Clin Cancer Res ; 11(17): 6382-9, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16144943

ABSTRACT

PURPOSE: We determined the effects of suberoylanilide hydroxamic acid (SAHA), a histone deacetylase inhibitor, on hsp90 and its client proteins Her-2, AKT, and c-Raf, as well as evaluated the cytotoxic effects of co-treatment of SAHA with trastuzumab or docetaxel in human breast cancer BT-474 and SKBR-3 cells containing amplification of Her-2. EXPERIMENTAL DESIGN: The cells were treated with SAHA (1.0-5.0 micromol/L) and/or trastuzumab (5-40 microg/mL) or docetaxel (5-20 nmol/L). Following this, apoptosis and the levels of p21(WAF1), p27(KIP1), AKT, c-Raf, and Her-2, as well as of the key regulators of apoptosis were determined. Synergistic interaction between drugs was evaluated by median dose-effect analysis. RESULTS: Treatment with SAHA up-regulated p21(WAF1) and p27(KIP1) levels, increased the percentage of cells in G2-M phase of the cell cycle, as well as induced apoptosis in a dose-dependent manner. This was associated with up-regulation of the pro-death Bak and Bim, as well as with attenuation of the levels of Her-2 and XIAP, survivin, Bcl-2, and Bcl-x(L) proteins. SAHA treatment induced acetylation of hsp90. This reduced the chaperone association of Her-2 with hsp90, promoting polyubiquitylation and degradation of Her-2. SAHA also attenuated the levels of c-Raf and AKT. Co-treatment with SAHA significantly increased trastuzumab or docetaxel-induced apoptosis of BT-474 and SKBR-3 cells. Additionally, median dose-effect analysis revealed that co-treatment with SAHA and trastuzumab or docetaxel induced synergistic cytotoxic effects against the breast cancer cells. CONCLUSIONS: These preclinical findings support the development of SAHA in combination with docetaxel and/or trastuzumab against Her-2-amplified breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Gene Amplification , Hydroxamic Acids/pharmacology , Receptor, ErbB-2/genetics , Signal Transduction/drug effects , Acetylation , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/genetics , Cell Cycle Proteins/metabolism , Cell Division/drug effects , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Docetaxel , Drug Synergism , Flow Cytometry , G2 Phase/drug effects , HSP90 Heat-Shock Proteins/metabolism , Histone Deacetylase Inhibitors , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Proteasome Endopeptidase Complex/drug effects , Protein Serine-Threonine Kinases/metabolism , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Receptor, ErbB-2/metabolism , Survivin , Taxoids/administration & dosage , Trastuzumab , Tumor Cells, Cultured , Tumor Suppressor Proteins/metabolism , Vorinostat , X-Linked Inhibitor of Apoptosis Protein , bcl-X Protein
6.
J Biol Chem ; 280(29): 26729-34, 2005 Jul 22.
Article in English | MEDLINE | ID: mdl-15937340

ABSTRACT

The hydroxamic acid (HAA) analogue pan-histone deacetylase (HDAC) inhibitors (HDIs) LAQ824 and LBH589 have been shown to induce acetylation and inhibit the ATP binding and chaperone function of heat shock protein (HSP) 90. This promotes the polyubiquitylation and degradation of the pro-growth and pro-survival client proteins Bcr-Abl, mutant FLT-3, c-Raf, and AKT in human leukemia cells. HDAC6 is a member of the class IIB HDACs. It is predominantly cytosolic, microtubule-associated alpha-tubulin deacetylase that is also known to promote aggresome inclusion of the misfolded polyubiquitylated proteins. Here we demonstrate that in the Bcr-abl oncogene expressing human leukemia K562 cells, HDAC6 can be co-immunoprecipitated with HSP90, and the knock-down of HDAC6 by its siRNA induced the acetylation of HSP90 and alpha-tubulin. Depletion of HDAC6 levels also inhibited the binding of HSP90 to ATP, reduced the chaperone association of HSP90 with its client proteins, e.g. Bcr-Abl, and induced polyubiquitylation and partial depletion of Bcr-Abl. Conversely, the ectopic overexpression of HDAC6 inhibited LAQ824-induced acetylation of HSP90 and alpha-tubulin and reduced LAQ824-mediated depletion of Bcr-Abl, AKT, and c-Raf. Collectively, these findings indicate that HDAC6 is also an HSP90 deacetylase. Targeted inhibition of HDAC6 leads to acetylation of HSP90 and disruption of its chaperone function, resulting in polyubiquitylation and depletion of pro-growth and pro-survival HSP90 client proteins including Bcr-Abl. Depletion of HDAC6 sensitized human leukemia cells to HAA-HDIs and proteasome inhibitors.


Subject(s)
HSP90 Heat-Shock Proteins/drug effects , Histone Deacetylase Inhibitors , Histone Deacetylases/physiology , Leukemia/pathology , Acetylation/drug effects , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , HSP90 Heat-Shock Proteins/metabolism , Histone Deacetylase 6 , Humans , Hydroxamic Acids/pharmacology , Indoles , K562 Cells , Leukemia/drug therapy , Molecular Chaperones/drug effects , Neoplasm Proteins/analysis , Neoplasm Proteins/drug effects , Panobinostat , Protein Binding , Ubiquitin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...