Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
ACS Omega ; 7(27): 23978-23987, 2022 Jul 12.
Article in English | MEDLINE | ID: mdl-35847311

ABSTRACT

Deposition in fuel cooling systems remains a challenge to the development of active cooling technologies for air-breathing engines. We experimentally and numerically investigated the influence of the secondary flow and heat-transfer characteristics of supercritical kerosene in a coiled tube on oxidation deposition. A coiled heated tube reactor (3000 mm long, 23 cycles) under constant heat flux and flow rate was applied to simulate the conditions of the fuel side in the heat exchanger of an aero-engine cooling system. The coupling characteristics of coking distribution with the development of secondary flow were studied along the whole pipe. The dynamic pressure, temperature, and velocity were analyzed in two specific circular cross sections located in the bend of the tube. The secondary flows induced in the coiled tube greatly enhance the heat transfer and slightly decrease the deposition rate, resulting in linear wall temperature profiles and a uniform coking distribution along the tube compared to the long straight tube. There is no obvious heat-transfer enhancement or deterioration in the whole coiled tube. The modified heat-transfer correlation of the supercritical RP-3 in the coiled tube was fitted at different flow rates and heat fluxes with an error of ±10%.

2.
Mol Cancer Res ; 20(3): 456-467, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34782371

ABSTRACT

The relationship between the checkpoint kinase Chk1 and the STAT3 pathway was examined in multiple myeloma cells. Gene expression profiling of U266 cells exposed to low (nmol/L) Chk1 inhibitor [PF-477736 (PF)] concentrations revealed STAT3 pathway-related gene downregulation (e.g., BCL-XL, MCL-1, c-Myc), findings confirmed by RT-PCR. This was associated with marked inhibition of STAT3 Tyr705 (but not Ser727) phosphorylation, dimerization, nuclear localization, DNA binding, STAT3 promoter activity by chromatin immunoprecipitation assay, and downregulation of STAT-3-dependent proteins. Similar findings were obtained in other multiple myeloma cells and with alternative Chk1 inhibitors (e.g., prexasertib, CEP3891). While PF did not reduce GP130 expression or modify SOCS or PRL-3 phosphorylation, the phosphatase inhibitor pervanadate antagonized PF-mediated Tyr705 dephosphorylation. Significantly, PF attenuated Chk1-mediated STAT3 phosphorylation in in vitro assays. Surface plasmon resonance analysis suggested Chk1/STAT3 interactions and PF reduced Chk1/STAT3 co-immunoprecipitation. Chk1 CRISPR knockout or short hairpin RNA knockdown cells also displayed STAT3 inactivation and STAT3-dependent protein downregulation. Constitutively active STAT3 diminished PF-mediated STAT3 inactivation and downregulate STAT3-dependent proteins while significantly reducing PF-induced DNA damage (γH2A.X formation) and apoptosis. Exposure of cells with low basal phospho-STAT3 expression to IL6 or human stromal cell conditioned medium activated STAT3, an event attenuated by Chk1 inhibitors. PF also inactivated STAT3 in primary human CD138+ multiple myeloma cells and tumors extracted from an NSG multiple myeloma xenograft model while inhibiting tumor growth. IMPLICATIONS: These findings identify a heretofore unrecognized link between the Chk1 and STAT3 pathways and suggest that Chk1 pathway inhibitors warrant attention as novel and potent candidate STAT3 antagonists in myeloma.


Subject(s)
Checkpoint Kinase 1/metabolism , Multiple Myeloma , Apoptosis , Cell Line, Tumor , DNA/metabolism , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , STAT3 Transcription Factor/metabolism
3.
Cancer Res ; 78(11): 3075-3086, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29559471

ABSTRACT

Inhibitors targeting BCL-2 apoptotic proteins have significant potential for the treatment of acute myeloid leukemia (AML); however, complete responses are observed in only 20% of patients, suggesting that targeting BCL-2 alone is insufficient to yield durable responses. Here, we assessed the efficacy of coadministration of the PI3K/mTOR inhibitor GDC-0980 or the p110ß-sparing PI3K inhibitor taselisib with the selective BCL-2 antagonist venetoclax in AML cells. Tetracycline-inducible downregulation of BCL-2 significantly sensitized MV4-11 and MOLM-13 AML cells to PI3K inhibition. Venetoclax/GDC-0980 coadministration induced rapid and pronounced BAX mitochondrial translocation, cytochrome c release, and apoptosis in various AML cell lines in association with AKT/mTOR inactivation and MCL-1 downregulation; ectopic expression of MCL-1 significantly protected cells from this regimen. Combined treatment was also effective against primary AML blasts from 17 patients, including those bearing various genetic abnormalities. Venetoclax/GDC-0980 markedly induced apoptosis in primitive CD34+/38-/123+ AML cell populations but not in normal hematopoietic progenitor CD34+ cells. The regimen was also active against AML cells displaying intrinsic or acquired venetoclax resistance or tumor microenvironment-associated resistance. Either combinatorial treatment markedly reduced AML growth and prolonged survival in a systemic AML xenograft mouse model and diminished AML growth in two patient-derived xenograft models. Venetoclax/GDC-0980 activity was partially diminished in BAK-/- cells and failed to induce apoptosis in BAX-/- and BAX-/-BAK-/- cells, whereas BIM-/- cells were fully sensitive. Similar results were observed with venetoclax alone in in vitro and in vivo systemic xenograft models. Collectively, these studies demonstrate that venetoclax/GDC-0980 exhibits potent anti-AML activity primarily through BAX and, to a lesser extent, BAK. These findings argue that dual BCL-2 and PI3K inhibition warrants further evaluation in AML.Significance: Combined treatment with clinically relevant PI3K and BCL-2 inhibitors may prove effective in the treatment of acute myeloid leukemia. Cancer Res; 78(11); 3075-86. ©2018 AACR.


Subject(s)
Apoptosis/physiology , Leukemia, Myeloid, Acute/metabolism , Mitochondria/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Down-Regulation/drug effects , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mitochondria/drug effects , Pyrimidines/pharmacology , Sulfonamides/pharmacology , U937 Cells
4.
Stat Methods Med Res ; 26(3): 1261-1280, 2017 Jun.
Article in English | MEDLINE | ID: mdl-25744107

ABSTRACT

Drug combination is a critically important therapeutic approach for complex diseases such as cancer and HIV due to its potential for efficacy at lower, less toxic doses and the need to move new therapies rapidly into clinical trials. One of the key issues is to identify which combinations are additive, synergistic, or antagonistic. While the value of multidrug combinations has been well recognized in the cancer research community, to our best knowledge, all existing experimental studies rely on fixing the dose of one drug to reduce the dimensionality, e.g. looking at pairwise two-drug combinations, a suboptimal design. Hence, there is an urgent need to develop experimental design and analysis methods for studying multidrug combinations directly. Because the complexity of the problem increases exponentially with the number of constituent drugs, there has been little progress in the development of methods for the design and analysis of high-dimensional drug combinations. In fact, contrary to common mathematical reasoning, the case of three-drug combinations is fundamentally more difficult than two-drug combinations. Apparently, finding doses of the combination, number of combinations, and replicates needed to detect departures from additivity depends on dose-response shapes of individual constituent drugs. Thus, different classes of drugs of different dose-response shapes need to be treated as a separate case. Our application and case studies develop dose finding and sample size method for detecting departures from additivity with several common (linear and log-linear) classes of single dose-response curves. Furthermore, utilizing the geometric features of the interaction index, we propose a nonparametric model to estimate the interaction index surface by B-spine approximation and derive its asymptotic properties. Utilizing the method, we designed and analyzed a combination study of three anticancer drugs, PD184, HA14-1, and CEP3891 inhibiting myeloma H929 cell line. To our best knowledge, this is the first ever three drug combinations study performed based on the original 4D dose-response surface formed by dose ranges of three drugs.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Drug Combinations , Research Design , Benzopyrans/administration & dosage , Benzopyrans/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Humans , Nitriles/administration & dosage , Nitriles/pharmacology , Sample Size , Statistics, Nonparametric
5.
Biom J ; 59(1): 9-24, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27185067

ABSTRACT

Few articles have been written on analyzing three-way interactions between drugs. It may seem to be quite straightforward to extend a statistical method from two-drugs to three-drugs. However, there may exist more complex nonlinear response surface of the interaction index (II) with more complex local synergy and/or local antagonism interspersed in different regions of drug combinations in a three-drug study, compared in a two-drug study. In addition, it is not possible to obtain a four-dimensional (4D) response surface plot for a three-drug study. We propose an analysis procedure to construct the dose combination regions of interest (say, the synergistic areas with II≤0.9). First, use the model robust regression method (MRR), a semiparametric method, to fit the entire response surface of the II, which allows to fit a complex response surface with local synergy/antagonism. Second, we run a modified genetic algorithm (MGA), a stochastic optimization method, many times with different random seeds, to allow to collect as many feasible points as possible that satisfy the estimated values of II≤0.9. Last, all these feasible points are used to construct the approximate dose regions of interest in a 3D. A case study with three anti-cancer drugs in an in vitro experiment is employed to illustrate how to find the dose regions of interest.


Subject(s)
Biometry/methods , Data Interpretation, Statistical , Drug Interactions , Drug Synergism , Pharmacology/methods , Humans
6.
Blood ; 124(17): 2687-97, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25208888

ABSTRACT

Bim contributes to resistance to various standard and novel agents. Here we demonstrate that Bim plays a functional role in bortezomib resistance in multiple myeloma (MM) cells and that targeting Bim by combining histone deacetylase inhibitors (HDACIs) with BH3 mimetics (eg, ABT-737) overcomes bortezomib resistance. BH3-only protein profiling revealed high Bim levels (Bim(hi)) in most MM cell lines and primary CD138(+) MM samples. Whereas short hairpin RNA Bim knockdown conferred bortezomib resistance in Bim(hi) cells, adaptive bortezomib-resistant cells displayed marked Bim downregulation. HDACI upregulated Bim and, when combined with ABT-737, which released Bim from Bcl-2/Bcl-xL, potently killed bortezomib-resistant cells. These events were correlated with Bim-associated autophagy attenuation, whereas Bim knockdown sharply increased autophagy in Bim(hi) cells. In Bim(low) cells, autophagy disruption by chloroquine (CQ) was required for HDACI/ABT-737 to induce Bim expression and lethality. CQ also further enhanced HDACI/ABT-737 lethality in bortezomib-resistant cells. Finally, HDACI failed to diminish autophagy or potentiate ABT-737-induced apoptosis in bim(-/-) mouse embryonic fibroblasts. Thus, Bim deficiency represents a novel mechanism of adaptive bortezomib resistance in MM cells, and Bim-targeting strategies combining HDACIs (which upregulate Bim) and BH3 mimetics (which unleash Bim from antiapoptotic proteins) overcomes such resistance, in part by disabling cytoprotective autophagy.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , Autophagy/physiology , Membrane Proteins/metabolism , Multiple Myeloma/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Autophagy/drug effects , Autophagy/genetics , Bcl-2-Like Protein 11 , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Cells, Cultured , Drug Resistance, Neoplasm/drug effects , Fluorescent Antibody Technique , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/administration & dosage , Hydroxamic Acids/pharmacology , Immunoblotting , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Pyrazines/pharmacology , RNA Interference , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays
7.
Mol Cell Biol ; 34(18): 3435-49, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25002530

ABSTRACT

In selective autophagy, the adaptor protein SQSTM1/p62 plays a critical role in recognizing/loading cargo (e.g., malfolded proteins) into autophagosomes for lysosomal degradation. Here we report that whereas SQSTM1/p62 levels fluctuated in a time-dependent manner during autophagy, inhibition or knockdown of Cdk9/cyclin T1 transcriptionally downregulated SQSTM1/p62 but did not affect autophagic flux. These interventions, or short hairpin RNA (shRNA) directly targeting SQSTM1/p62, resulted in cargo loading failure and inefficient autophagy, phenomena recently described for Huntington's disease neurons. These events led to the accumulation of the BH3-only protein NBK/Bik on endoplasmic reticulum (ER) membranes, most likely by blocking loading and autophagic degradation of NBK/Bik, culminating in apoptosis. Whereas NBK/Bik upregulation was further enhanced by disruption of distal autophagic events (e.g., autophagosome maturation) by chloroquine (CQ) or Lamp2 shRNA, it was substantially diminished by inhibition of autophagy initiation (e.g., genetically by shRNA targeting Ulk1, beclin-1, or Atg5 or pharmacologically by 3-methyladenine [3-MA] or spautin-1), arguing that NBK/Bik accumulation stems from inefficient autophagy. Finally, NBK/Bik knockdown markedly attenuated apoptosis in vitro and in vivo. Together, these findings identify novel cross talk between autophagy and apoptosis, wherein targeting SQSTM1/p62 converts cytoprotective autophagy to an inefficient form due to cargo loading failure, leading to NBK/Bik accumulation, which triggers apoptosis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Protein Transport/drug effects , Adaptor Proteins, Signal Transducing/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/genetics , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Cells, Cultured , Cyclin T/metabolism , Cyclin-Dependent Kinase 9/metabolism , Cycloheximide/pharmacology , Flavonoids/pharmacology , Gene Expression Regulation , Humans , Indoles , Leupeptins/pharmacology , Membrane Proteins/genetics , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Piperidines/pharmacology , Pyrazines/pharmacology , Pyrroles/pharmacology , RNA, Small Interfering/pharmacology , Sequestosome-1 Protein
8.
PLoS One ; 9(3): e89064, 2014.
Article in English | MEDLINE | ID: mdl-24594907

ABSTRACT

The anti-apoptotic protein Mcl-1 plays a major role in multiple myeloma (MM) cell survival as well as bortezomib- and microenvironmental forms of drug resistance in this disease. Consequently, there is a critical need for strategies capable of targeting Mcl-1-dependent drug resistance in MM. The present results indicate that a regimen combining Chk1 with MEK1/2 inhibitors effectively kills cells displaying multiple forms of drug resistance stemming from Mcl-1 up-regulation in association with direct transcriptional Mcl-1 down-regulation and indirect disabling of Mcl-1 anti-apoptotic function through Bim up-regulation and increased Bim/Mcl-1 binding. These actions release Bak from Mcl-1, accompanied by Bak/Bax activation. Analogous events were observed in both drug-naïve and acquired bortezomib-resistant MM cells displaying increased Mcl-1 but diminished Bim expression, or cells ectopically expressing Mcl-1. Moreover, concomitant Chk1 and MEK1/2 inhibition blocked Mcl-1 up-regulation induced by IL-6/IGF-1 or co-culture with stromal cells, effectively overcoming microenvironment-related drug resistance. Finally, this regimen down-regulated Mcl-1 and robustly killed primary CD138+ MM cells, but not normal hematopoietic cells. Together, these findings provide novel evidence that this targeted combination strategy could be effective in the setting of multiple forms of Mcl-1-related drug resistance in MM.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Multiple Myeloma/drug therapy , Multiple Myeloma/enzymology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Protein Kinase Inhibitors/therapeutic use , Protein Kinases/metabolism , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Benzamides/pharmacology , Benzamides/therapeutic use , Boronic Acids/pharmacology , Bortezomib , Cell Line, Tumor , Checkpoint Kinase 1 , Cytoprotection/drug effects , Down-Regulation/drug effects , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Membrane Proteins/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein Binding/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/metabolism , Pyrazines/pharmacology , Syndecan-1/metabolism , Up-Regulation/drug effects , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
9.
Mol Cancer Ther ; 12(6): 878-89, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23536721

ABSTRACT

Interactions between the novel Chk1 inhibitor MK-8776 and the histone deacetylase (HDAC) inhibitor (HDACI) vorinostat were examined in human leukemia cells harboring wild-type (wt) or deficient p53. MK-8776 synergistically potentiated vorinostat-mediated apoptosis in various p53-wt or -deficient leukemia cell lines, whereas p53 knockdown by short hairpin RNA (shRNA) sensitized p53-wt cells to lethality of this regimen. Leukemia cell lines carrying FLT3-ITD were also sensitive to the MK-8776/vorinostat regimen. Synergistic interactions were associated with inhibition of Chk1 activity, interference with the intra-S-phase checkpoint, disruption of DNA replication, and downregulation of proteins involved in DNA replication (e.g., Cdt1) and repair (e.g., CtIP and BRCA1), resulting in sharp increases in DNA damage, reflected by enhanced γ-H2A.X formation, and apoptosis. Moreover, leukemia cells expressing kinase-dead Chk1 (D130A) or Chk1 shRNA were significantly more sensitive to HDACIs compared with their wt counterparts and displayed downregulation of CtIP and BRCA1 phosphorylation following HDACI exposure. Finally, the MK-8776/vorinostat regimen was active in primary acute myelogenous leukemia (AML) blasts, particularly against the CD34(+)/CD38(-)/CD123(+) population enriched for leukemia-initiating cells. In contrast, identical regimens were relatively sparing toward normal cord blood CD34(+) cells. Together, these findings indicate that the novel Chk1 inhibitor MK-8776 markedly potentiates HDACI lethality in leukemia cells displaying various genetic backgrounds through mechanisms involving disruption of the intra-S checkpoint, DNA replication, and DNA repair. They also argue that leukemic cells, including those bearing oncogenic mutations associated with poor prognosis, for example, p53 deletion/mutation or FLT3-ITD, may also be susceptible to this strategy.


Subject(s)
Apoptosis/drug effects , Histone Deacetylase Inhibitors/administration & dosage , Leukemia/drug therapy , Protein Kinases/metabolism , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Bone Marrow Cells/pathology , Cells, Cultured , Checkpoint Kinase 1 , DNA Repair/drug effects , DNA Replication/drug effects , Gene Expression Regulation, Leukemic , Histone Deacetylases/biosynthesis , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/administration & dosage , Leukemia/metabolism , Leukemia/pathology , Protein Kinases/chemistry , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , S Phase Cell Cycle Checkpoints/drug effects , Tumor Suppressor Protein p53/antagonists & inhibitors , Vorinostat
10.
Cancer Res ; 72(16): 4225-37, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22693249

ABSTRACT

BH3 mimetic drugs induce cell death by antagonizing the activity of antiapoptotic Bcl-2 family proteins. Cyclin-dependent kinase (CDK) inhibitors that function as transcriptional repressors downregulate the Bcl-2 family member Mcl-1 and increase the activity of selective BH3 mimetics that fail to target this protein. In this study, we determined whether CDK inhibitors potentiate the activity of pan-BH3 mimetics directly neutralizing Mcl-1. Specifically, we evaluated interactions between the prototypical pan-CDK inhibitor flavopiridol and the pan-BH3 mimetic obatoclax in multiple myeloma (MM) cells in which Mcl-1 is critical for survival. Coadministration of flavopiridol and obatoclax synergistically triggered apoptosis in both drug-naïve and drug-resistant MM cells. Mechanistic investigations revealed that flavopiridol inhibited Mcl-1 transcription but increased transcription of Bim and its binding to Bcl-2/Bcl-xL. Obatoclax prevented Mcl-1 recovery and caused release of Bim from Bcl-2/Bcl-xL and Mcl-1, accompanied by activation of Bax/Bak. Whether administered singly or in combination with obatoclax, flavopiridol also induced upregulation of multiple BH3-only proteins, including BimEL, BimL, Noxa, and Bik/NBK. Notably, short hairpin RNA knockdown of Bim or Noxa abrogated lethality triggered by the flavopiridol/obatoclax combination in vitro and in vivo. Together, our findings show that CDK inhibition potentiates pan-BH3 mimetic activity through a cooperative mechanism involving upregulation of BH3-only proteins with coordinate downregulation of their antiapoptotic counterparts. These findings have immediate implications for the clinical trial design of BH3 mimetic-based therapies that are presently being studied intensively for the treatment of diverse hematopoietic malignancies, including lethal multiple myeloma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , Flavonoids/pharmacology , Multiple Myeloma/drug therapy , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Biomimetic Materials/administration & dosage , Biomimetic Materials/pharmacology , Cell Line, Tumor , Drug Synergism , Flavonoids/administration & dosage , Humans , Indoles , Membrane Proteins/metabolism , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Mitochondria/drug effects , Multiple Myeloma/enzymology , Myeloid Cell Leukemia Sequence 1 Protein , Peptide Fragments/metabolism , Piperidines/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrroles/administration & dosage , Up-Regulation/drug effects , Xenograft Model Antitumor Assays , bcl-X Protein/metabolism
11.
Blood ; 118(19): 5189-200, 2011 Nov 10.
Article in English | MEDLINE | ID: mdl-21911831

ABSTRACT

Effects of Chk1 and MEK1/2 inhibition were investigated in cytokinetically quiescent multiple myeloma (MM) and primary CD138(+) cells. Coexposure to the Chk1 and MEK1/2 inhibitors AZD7762 and selumetinib (AZD6244) robustly induced apoptosis in various MM cells and CD138(+) primary samples, but spared normal CD138(-) and CD34(+) cells. Furthermore, Chk1/MEK1/2 inhibitor treatment of asynchronized cells induced G(0)/G(1) arrest and increased apoptosis in all cell-cycle phases, including G(0)/G(1). To determine whether this regimen is active against quiescent G(0)/G(1) MM cells, cells were cultured in low-serum medium to enrich the G(0)/G(1) population. G(0)/G(1)-enriched cells exhibited diminished sensitivity to conventional agents (eg, Taxol and VP-16) but significantly increased susceptibility to Chk1 ± MEK1/2 inhibitors or Chk1 shRNA knock-down. These events were associated with increased γH2A.X expression/foci formation and Bim up-regulation, whereas Bim shRNA knock-down markedly attenuated lethality. Immunofluorescent analysis of G(0)/G(1)-enriched or primary MM cells demonstrated colocalization of activated caspase-3 and the quiescent (G(0)) marker statin, a nuclear envelope protein. Finally, Chk1/MEK1/2 inhibition increased cell death in the Hoechst-positive (Hst(+)), low pyronin Y (PY)-staining (2N Hst(+)/PY(-)) G(0) population and in sorted small side-population (SSP) MM cells. These findings provide evidence that cytokinetically quiescent MM cells are highly susceptible to simultaneous Chk1 and MEK1/2 inhibition.


Subject(s)
MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Protein Kinase Inhibitors/therapeutic use , Protein Kinases/metabolism , Apoptosis/drug effects , Benzimidazoles/administration & dosage , Benzimidazoles/therapeutic use , Cell Line, Tumor , Checkpoint Kinase 1 , DNA Damage , G1 Phase , Humans , Interleukin-6/metabolism , Multiple Myeloma/pathology , Protein Kinase Inhibitors/administration & dosage , Resting Phase, Cell Cycle , Syndecan-1/metabolism , Thiophenes/administration & dosage , Thiophenes/therapeutic use , Urea/administration & dosage , Urea/analogs & derivatives , Urea/therapeutic use
12.
J Biol Chem ; 286(39): 34036-50, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21816815

ABSTRACT

Post-translational modifications of RelA play an important role in regulation of NF-κB activation. We previously demonstrated that in malignant hematopoietic cells, histone deacetylase inhibitors (HDACIs) induced RelA hyperacetylation and NF-κB activation, attenuating lethality. We now present evidence that IκB kinase (IKK) ß-mediated RelA Ser-536 phosphorylation plays a significant functional role in promoting RelA acetylation, inducing NF-κB activation, and limiting HDACI lethality in human multiple myeloma (MM) cells. Immunoblot profiling revealed that although basal RelA phosphorylation varied in MM cells, Ser-536 phosphorylation correlated with IKK activity. Exposure to the pan-HDACIs vorinostat or LBH-589 induced phosphorylation of IKKα/ß (Ser-180/Ser-181) and RelA (Ser-536) in MM cells, including cells expressing an IκBα "super-repressor," accompanied by increased RelA nuclear translocation, acetylation, DNA binding, and transactivation activity. These events were substantially blocked by either pan-IKK or IKKß-selective inhibitors, resulting in marked apoptosis. Consistent with these events, inhibitory peptides targeting either the NF-κB essential modulator (NEMO) binding domain for IKK complex formation or RelA phosphorylation sites also significantly increased HDACI lethality. Moreover, IKKß knockdown by shRNA prevented Ser-536 phosphorylation and significantly enhanced HDACI susceptibility. Finally, introduction of a nonphosphorylatable RelA mutant S536A, which failed to undergo acetylation in response to HDACIs, impaired NF-κB activation and increased cell death. These findings indicate that HDACIs induce Ser-536 phosphorylation of the NF-κB subunit RelA through an IKKß-dependent mechanism, an action that is functionally involved in activation of the cytoprotective NF-κB signaling cascade primarily through facilitation of RelA acetylation rather than nuclear translocation.


Subject(s)
Apoptosis/drug effects , Cell Nucleus/metabolism , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , I-kappa B Kinase/metabolism , Multiple Myeloma/metabolism , Neoplasm Proteins/metabolism , Transcription Factor RelA/metabolism , Acetylation/drug effects , Active Transport, Cell Nucleus/drug effects , Amino Acid Substitution , Cell Line, Tumor , Cell Nucleus/genetics , Gene Knockdown Techniques , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/genetics , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/genetics , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Mutation, Missense , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factor RelA/genetics , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Vorinostat
13.
Br J Haematol ; 153(2): 222-35, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21375523

ABSTRACT

Interactions between the histone deacetylase inhibitor belinostat and the proteasome inhibitor bortezomib were investigated in acute myeloid leukaemia (AML) and acute lymphoblastic leukaemia (ALL) cells. Co-administration of sub-micromolar concentrations of belinostat with low nanomolar concentrations of bortezomib sharply increased apoptosis in both AML and ALL cell lines and primary blasts. Synergistic interactions were associated with interruption of both canonical and non-canonical nuclear factor (NF)-κB signalling pathways, e.g. accumulation of the phosphorylated (S32/S36) form of IκBα, diminished belinostat-mediated RelA/p65 hyperacetylation (K310), and reduced processing of p100 into p52. These events were accompanied by down-regulation of NF-κB-dependent pro-survival proteins (e.g. XIAP, Bcl-xL). Moreover, belinostat/bortezomib co-exposure induced up-regulation of the BH3-only pro-death protein Bim. Significantly, shRNA knock-down of Bim substantially reduced the lethality of belinostat/bortezomib regimens. Administration of belinostat ± bortezomib also induced hyperacetylation (K40) of α-tubulin, indicating histone deacetylase inhibitor 6 inhibition. Finally, in contrast to the pronounced lethality of belinostat/bortezomib toward primary leukaemia blasts, equivalent treatment was relatively non-toxic to normal CD34(+) cells. Together, these findings indicate that belinostat and bortezomib interact synergistically in both cultured and primary AML and ALL cells, and raise the possibilities that up-regulation of Bim and interference with NF-κB pathways contribute to this phenomenon. They also suggest that combined belinostat/bortezomib regimens warrant further attention in acute leukaemias.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis Regulatory Proteins/metabolism , Boronic Acids/pharmacology , Hydroxamic Acids/pharmacology , Leukemia, Myeloid, Acute/metabolism , Membrane Proteins/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins/metabolism , Pyrazines/pharmacology , Transcription Factor RelA/metabolism , Acetylation/drug effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Bcl-2-Like Protein 11 , Boronic Acids/therapeutic use , Bortezomib , Drug Synergism , Female , HL-60 Cells , Histone Deacetylase 6 , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/therapeutic use , I-kappa B Kinase/metabolism , Jurkat Cells , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Male , Phosphorylation/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Pyrazines/therapeutic use , Signal Transduction/drug effects , Sulfonamides , Tubulin/metabolism , U937 Cells , X-Linked Inhibitor of Apoptosis Protein/metabolism , bcl-X Protein/metabolism
14.
Blood ; 117(6): 1947-57, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21148814

ABSTRACT

Ras/MEK/ERK pathway activation represents an important compensatory response of human multiple myeloma (MM) cells to checkpoint kinase 1 (Chk1) inhibitors. To investigate the functional roles of Src in this event and potential therapeutic significance, interactions between Src and Chk1 inhibitors (eg, UCN-01 or Chk1i) were examined in vitro and in vivo. The dual Src/Abl inhibitors BMS354825 and SKI-606 blocked Chk1-inhibitor-induced extracellular signal-regulated kinase 1/2 (ERK1/2) activation, markedly increasing apoptosis in association with BimEL up-regulation, p34(cdc2) activation, and DNA damage in MM cell lines and primary CD138(+) MM samples. Loss-of-function Src mutants (K297R, K296R/Y528F) or shRNA knock-down of Src prevented the ERK1/2 activation induced by Chk1 inhibitors and increased apoptosis. Conversely, constitutively active Ras or mitogen-activated protein kinase/ERK kinase 1 (MEK1) significantly diminished the ability of Src inhibitors to potentiate Chk1-inhibitor lethality. Moreover, Src/Chk1-inhibitor cotreatment attenuated MM-cell production of vascular endothelial growth factor and other angiogenic factors (eg, ANG [angiogenin], TIMP1/2 [tissue inhibitor of metalloproteinases 1/2], and RANTES [regulated on activation normal T-cell expressed and secreted]), and inhibited in vitro angiogenesis. Finally, coadministration of BMS354825 and UCN-01 suppressed human MM tumor growth in a murine xenograft model, increased apoptosis, and diminished angiogenesis. These findings suggest that Src kinase is required for Chk1-inhibitor-mediated Ras → ERK1/2 signaling activation, and that disruption of this event sharply potentiates the anti-MM activity of Chk1 inhi-bitors in vitro and in vivo.


Subject(s)
Apoptosis/physiology , Multiple Myeloma/pathology , Multiple Myeloma/physiopathology , Protein Kinases/physiology , src-Family Kinases/antagonists & inhibitors , Aniline Compounds/pharmacology , Animals , Apoptosis/drug effects , CDC2 Protein Kinase/metabolism , Cell Line, Tumor , Checkpoint Kinase 1 , DNA Damage , Dasatinib , Female , Gene Knockdown Techniques , Humans , In Vitro Techniques , MAP Kinase Signaling System/drug effects , Mice , Mice, Nude , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Mutation , Neovascularization, Pathologic/drug therapy , Nitriles/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Quinolines/pharmacology , RecQ Helicases/antagonists & inhibitors , Thiazoles/pharmacology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/physiology , src-Family Kinases/genetics , src-Family Kinases/physiology
15.
Br J Haematol ; 151(1): 70-83, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20701602

ABSTRACT

Interactions between the nuclear factor (NF)-κB inhibitor parthenolide and the pan-histone deacetylase inhibitors (HDACIs) vorinostat and LBH589 were investigated in human acute myeloid leukaemia (AML) cells, including primary AML blasts. Co-administration of parthenolide blocked HDACI-mediated phosphorylation/activation of IKK and RelA/p65 in association with increased JNK1 activation in various AML cell types. These events were accompanied by an increase in apoptosis in multiple AML cell lines (e.g. U937, HL-60, NB4, MV-4-11, and MOLM-13). Significantly, parthenolide also increased HDACI-mediated cell death in haematopoietic cells transduced with the MLL-MLLT1 fusion gene, which exhibit certain leukaemia-initiating cell characteristics, as well as primary AML blasts. Exposure to parthenolide/HDACI regimens clearly inhibited the growth of AML-colony-forming units but was relatively sparing toward normal haematopoietic progenitors. Notably, blockade of c-Jun N-terminal kinase (JNK) signalling by either pharmacological inhibitors or genetic means (e.g. dominant-negative JNK1 or JNK1 shRNA) diminished parthenolide/HDACI-mediated lethality. Moreover, dominant-negative MKK7, but not dominant-negative MKK4/SEK1, blocked JNK1 activation and apoptosis induced by parthenolide/HDACI regimens. Together, these findings indicate that parthenolide potentiates HDACI lethality in human AML cells through a process involving NF-κB inhibition and subsequent MKK7-dependent activation of the SAPK/JNK pathway. They also raise the possibility that this strategy may target leukaemic progenitor cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Leukemia, Myeloid, Acute/pathology , NF-kappa B/antagonists & inhibitors , Apoptosis/drug effects , Drug Evaluation, Preclinical/methods , Drug Synergism , Enzyme Activation/drug effects , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Leukemia, Myeloid, Acute/metabolism , Sesquiterpenes/administration & dosage , Sesquiterpenes/pharmacology , Signal Transduction/drug effects , Tumor Cells, Cultured
16.
Mol Cell Biol ; 29(23): 6149-69, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19805519

ABSTRACT

The Bcl-2 antagonist ABT-737 kills transformed cells in association with displacement of Bim from Bcl-2. The histone deactetylase (HDAC) inhibitor suberoyl bis-hydroxamic acid (SBHA) was employed to determine whether and by what mechanism ABT-737 might interact with agents that upregulate Bim. Expression profiling of BH3-only proteins indicated that SBHA increased Bim, Puma, and Noxa expression, while SBHA concentrations that upregulated Bim significantly potentiated ABT-737 lethality. Concordance between SBHA-mediated Bim upregulation and interactions with ABT-737 was observed in various human leukemia and myeloma cells. SBHA-induced Bim was largely sequestered by Bcl-2 and Bcl-x(L), rather than Mcl-1; ABT-737 attenuated these interactions, thereby triggering Bak/Bax activation and mitochondrial outer membrane permeabilization. Knockdown of Bim (but not Puma or Noxa) by shRNA or ectopic overexpression of Bcl-2, Bcl-x(L), or Mcl-1 diminished Bax/Bak activation and apoptosis. Notably, ectopic expression of these antiapoptotic proteins disabled death signaling by sequestering different proapoptotic proteins, i.e., Bim by Bcl-2, both Bim and Bak by Bcl-x(L), and Bak by Mcl-1. Together, these findings indicate that HDAC inhibitor-inducible Bim is primarily neutralized by Bcl-2 and Bcl-x(L), thus providing a mechanistic framework by which Bcl-2 antagonists potentiate the lethality of agents, such as HDAC inhibitors, which upregulate Bim.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Biphenyl Compounds/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Membrane Proteins/metabolism , Nitrophenols/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins/metabolism , Sulfonamides/pharmacology , bcl-X Protein/metabolism , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Cell Line, Tumor , Humans , Leukemia/metabolism , Leukemia/pathology , Membrane Proteins/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Myeloid Cell Leukemia Sequence 1 Protein , Piperazines/pharmacology , Protein Binding , Protein Transport , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , RNA Interference , Up-Regulation/drug effects
17.
Blood ; 112(6): 2439-49, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18614762

ABSTRACT

The role of the Ras/MEK/ERK pathway was examined in relation to DNA damage in human multiple myeloma (MM) cells exposed to Chk1 inhibitors in vitro and in vivo. Exposure of various MM cells to marginally toxic concentrations of the Chk1 inhibitors UCN-01 or Chk1i modestly induced DNA damage, accompanied by Ras and ERK1/2 activation. Interruption of these events by pharmacologic (eg, the farnesyltransferase inhibitor R115777 or the MEK1/2 inhibitor PD184352) or genetic (eg, transfection with dominant-negative Ras or MEK1 shRNA) means induced pronounced DNA damage, reflected by increased gammaH2A.X expression/foci formation and by comet assay. Increased DNA damage preceded extensive apoptosis. Notably, similar phenomena were observed in primary CD138(+) MM cells. Enforced MEK1/2 activation by B-Raf transfection prevented R115777 but not PD184352 from inactivating ERK1/2 and promoting Chk1 inhibitor-induced gammaH2A.X expression. Finally, coadministration of R115777 diminished UCN-01-mediated ERK1/2 activation and markedly potentiated gammaH2A.X expression in a MM xenograft model, associated with a striking increase in tumor cell apoptosis and growth suppression. Such findings suggest that Ras/MEK/ERK activation opposes whereas its inhibition dramatically promotes Chk1 antagonist-mediated DNA damage. Together, these findings identify a novel mechanism by which agents targeting the Ras/MEK/ERK pathway potentiate Chk1 inhibitor lethality in MM.


Subject(s)
DNA Damage/drug effects , MAP Kinase Signaling System/physiology , Multiple Myeloma/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinases/drug effects , ras Proteins/physiology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis , Checkpoint Kinase 1 , Humans , Mice , Mice, SCID , Multiple Myeloma/pathology , Protein Kinase Inhibitors/therapeutic use , Quinolones/pharmacology , Quinolones/therapeutic use , Staurosporine/analogs & derivatives , Staurosporine/pharmacology , Staurosporine/therapeutic use , Xenograft Model Antitumor Assays
18.
Blood ; 112(3): 793-804, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18505786

ABSTRACT

Interactions between the dual Bcr/Abl and aurora kinase inhibitor MK-0457 and the histone deacetylase inhibitor vorinostat were examined in Bcr/Abl(+) leukemia cells, including those resistant to imatinib mesylate (IM), particularly those with the T315I mutation. Coadministration of vorinostat dramatically increased MK-0457 lethality in K562 and LAMA84 cells. Notably, the MK-0457/vorinostat regimen was highly active against primary CD34(+) chronic myelogenous leukemia (CML) cells and Ba/F3 cells bearing various Bcr/Abl mutations (ie, T315I, E255K, and M351T), as well as IM-resistant K562 cells exhibiting Bcr/Abl-independent, Lyn-dependent resistance. These events were associated with inactivation and down-regulation of wild-type (wt) and mutated Bcr/Abl (particularly T315I). Moreover, treatment with MK-0457 resulted in accumulation of cells with 4N or more DNA content, whereas coadministration of vorinostat markedly enhanced aurora kinase inhibition by MK-0457, and preferentially killed polyploid cells. Furthermore, vorinostat also interacted with a selective inhibitor of aurora kinase A and B to potentiate apoptosis without modifying Bcr/Abl activity. Finally, vorinostat markedly induced Bim expression, while blockade of Bim induction by siRNA dramatically diminished the capacity of this agent to potentiate MK-0457 lethality. Together, these findings indicate that vorinostat strikingly increases MK-0457 activity against IM-sensitive and -resistant CML cells through inactivation of Bcr/Abl and aurora kinases, as well as by induction of Bim.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Hydroxamic Acids/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/pharmacology , Animals , Apoptosis Regulatory Proteins/antagonists & inhibitors , Aurora Kinase A , Aurora Kinases , Bcl-2-Like Protein 11 , Benzamides , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Synergism , Fusion Proteins, bcr-abl/antagonists & inhibitors , Humans , Imatinib Mesylate , Membrane Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pyrimidines , Vorinostat
19.
Mol Cancer Ther ; 7(3): 616-29, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18347148

ABSTRACT

The present studies were initiated to determine in greater molecular detail how MEK1/2 inhibitors [PD184352 and AZD6244 (ARRY-142886)] interact with UCN-01 (7-hydroxystaurosporine) to kill mammary carcinoma cells in vitro and radiosensitize mammary tumors in vitro and in vivo and whether farnesyl transferase inhibitors interact with UCN-01 to kill mammary carcinoma cells in vitro and in vivo. Expression of constitutively activated MEK1 EE or molecular suppression of JNK and p38 pathway signaling blocked MEK1/2 inhibitor and UCN-01 lethality, effects dependent on the expression of BAX, BAK, and, to a lesser extent, BIM and BID. In vitro colony formation studies showed that UCN-01 interacted synergistically with the MEK1/2 inhibitors PD184352 or AZD6244 and the farnesyl transferase inhibitors FTI277 and R115,777 to kill human mammary carcinoma cells. Athymic mice carrying approximately 100 mm(3) MDA-MB-231 cell tumors were subjected to a 2-day exposure of either vehicle, R115,777 (100 mg/kg), the MEK1/2 inhibitor PD184352 (25 mg/kg), UCN-01 (0.2 mg/kg), or either of the drugs in combination with UCN-01. Transient exposure of tumors to R115,777, PD184352, or UCN-01 did not significantly alter tumor growth rate or the mean tumor volume in vivo approximately 15 to 30 days after drug administration. In contrast, combined treatment with R115,777 and UCN-01 or with PD184352 and UCN-01 significantly reduced tumor growth. Tumor cells isolated after combined drug exposure exhibited a significantly greater reduction in plating efficiency using ex vivo colony formation assays than tumor cells that were exposed to either drug individually. Irradiation of mammary tumors after drug treatment, but not before or during treatment, significantly enhanced the lethal effects of UCN-01 and MEK1/2 inhibitor treatment. These findings argue that UCN-01 and multiple inhibitors of the RAS-MEK pathway have the potential to suppress mammary tumor growth, and to interact with radiation, in vitro and in vivo.


Subject(s)
Cell Death/drug effects , MAP Kinase Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Blotting, Western , Cell Line, Tumor , Electrophoresis, Polyacrylamide Gel , Humans , Immunohistochemistry , Staurosporine/analogs & derivatives , Staurosporine/pharmacology
20.
Blood ; 110(6): 2092-101, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17540843

ABSTRACT

The role of Bim in synergistic interactions between UCN-01 and MEK1/2 inhibitors in human multiple myeloma cells was investigated. Exposure of U266 or RPMI8226 cells to UCN-01 resulted in ERK1/2 activation-associated Bim(EL) phosphorylation/down-regulation, events abrogated by MEK1/2 inhibitors. Enforced activation of ERK1/2 by transfection with constitutively active MEK1 diminished the capacity of PD98059 but not PD184352 to block UCN-01-mediated Bim(EL) phosphorylation and to potentiate apoptosis. Cotreatment with MEK1/2 inhibitors increased the association of Bim(EL) with both Bcl-2 and Bcl-x(L) in UCN-01-treated cells, leading to Bax/Bak conformational change and Bax mitochondrial translocation. Down-regulation of Bim(EL) by shRNA substantially diminished UCN-01/MEK inhibitor-mediated Bax/Bak activation and apoptosis. Furthermore, transfection of cells with S65A Bim, a mutant resistant to UCN-01-mediated phosphorylation, significantly sensitized cells to UCN-01 lethality. Conversely, ectopic expression of either Bcl-2 or Bcl-x(L) did not alter UCN-01/MEK1/2 inhibitor-mediated modifications in Bim(EL) phosphorylation but largely prevented cell death. Finally, IL-6 or IGF-1 failed to prevent MEK1/2 inhibitors from blocking UCN-01-induced Bim(EL) phosphorylation/degradation or cell death. Collectively, these findings argue that UCN-01-mediated ERK1/2 activation leads to Bim(EL) phosphorylation/inactivation, resulting in cytoprotection, and that interference with these events by MEK1/2 inhibitors plays a critical role in synergistic induction of apoptosis by these agents.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Membrane Proteins/metabolism , Multiple Myeloma/drug therapy , Proto-Oncogene Proteins/metabolism , Staurosporine/analogs & derivatives , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Benzamides/pharmacology , Blotting, Western , Drug Synergism , Humans , Immunoprecipitation , Insulin-Like Growth Factor I/pharmacology , Interleukin-6/pharmacology , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Phosphorylation/drug effects , Protein Conformation , Protein Kinase C/antagonists & inhibitors , Protein Transport , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/pharmacology , Staurosporine/pharmacology , Subcellular Fractions , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...