Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
1.
Invest New Drugs ; 38(1): 1-9, 2020 02.
Article in English | MEDLINE | ID: mdl-30877426

ABSTRACT

PLK1 has an important role in the regulation of cell cycle and represents an important target for cancer treatment. This enzyme belongs to the Polo-like kinases family, which is characterized by a regulatory domain named Polo-box domain (PBD). Rather than regular kinase inhibitors, this domain provides high selectivity to PLK1. Here, we report on four novel PLK1 PBD inhibitors identified by cytotoxicity screening and fluorescence polarization assay of a chemical library of natural and semisynthetic compounds. These compounds revealed two- to three-fold higher selectivity to the PDB of PLK1 than to those of the related family members, PLK2 and PLK3. These four substances inhibited tumor cell growth of sensitive CCRF-CEM and multidrug-resistant CEM/ADR5000 leukemia cells. The tested compounds increased the apoptotic cell fraction, which indicates apoptosis as a major mechanism of cell death. Cell cycle analysis showed compound (5) arrested the cell cycle of CCRF-CEM cells in the G2/M phase, while the other three molecules ((compound (3), compound (4), and compound (6)) exerted pronounced cytotoxicity with an increase of cells in the sub-G1 population. Molecular docking was performed for the understanding of ligand-protein interaction, the tested candidates showed strong binding affinity to PLK1 PBD. In conclusion, we identified four new chemical scaffolds that may serve as lead compounds for the development of selective PLK1 inhibitors in the future.


Subject(s)
Biological Products/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Cell Proliferation , High-Throughput Screening Assays/methods , Leukemia, T-Cell/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Apoptosis , Cell Cycle , Humans , Leukemia, T-Cell/enzymology , Leukemia, T-Cell/pathology , Molecular Docking Simulation , Protein Binding , Protein Kinase Inhibitors/chemistry , Tumor Cells, Cultured , Polo-Like Kinase 1
2.
Anticancer Res ; 37(10): 5415-5423, 2017 10.
Article in English | MEDLINE | ID: mdl-28982851

ABSTRACT

Unopposed phosphoinositide 3-kinase (PI3K) activity and 3-phosphoinositide production in Jurkat cells, due to a mutation in the phosphatase and tensin homolog deleted on chromosome 10 (PTEN) tumor-suppressor protein, results in deregulation of PH domain-containing proteins including the serine/threonine kinase PKB. In Jurkat cells, PKB is constitutively active and phosphorylated at the activation-loop residue (Thr308). 3-Phosphoinositide-dependent protein kinase-1 (PDK1), an enzyme that also contains a PH domain, catalyses Thr308 phosphorylation of PKB in addition to other kinase families such as PKC isoforms. It is unknown, however, whether the loss of PTEN in Jurkat cells also results in unregulated PDK1 activity and whether such loss has an impact on activation-loop phosphorylation of other PDK1 substrates e.g. PKC. In this study, we addressed whether loss of PTEN in Jurkat cells affects PDK1 catalytic activity and intracellular localization. We demonstrated that reducing the level of 3-phosphoinositides in Jurkat cells with pharmacological inhibitors of PI3K or expression of PTEN does not affect PDK1 activity or its intracellular localization. We conclude, therefore, that although Jurkat cells lack PTEN expression, only a subset of pathways downstream of PDK1 are perturbed as a consequence of PTEN loss.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/metabolism , Leukemia, T-Cell/enzymology , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Catalysis , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Jurkat Cells , Leukemia, T-Cell/genetics , Leukemia, T-Cell/pathology , PTEN Phosphohydrolase/genetics , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Time Factors , Transfection
3.
Biomed Res Int ; 2017: 9872073, 2017.
Article in English | MEDLINE | ID: mdl-28785594

ABSTRACT

Pterostilbene is a natural 3,5-dimethoxy analog of trans-resveratrol that has been reported to have antitumor, antioxidant, and anti-inflammatory effects. T-cell leukemia/lymphoma is one of the more aggressive yet uncommon non-Hodgkin lymphomas. Although there has been increasing research into T-cell leukemia/lymphoma, the molecular mechanisms of the antitumor effects of pterostilbene against this malignancy are still largely unknown. The aim of this study is to confirm the effects of pterostilbene in T-cell leukemia/lymphoma. Jurkat and Hut-78 cells treated with pterostilbene were evaluated for cell proliferation using Cell Counting Kit-8, and apoptosis, cell cycle progression, reactive oxygen species generation, and mitochondrial membrane potential were analyzed using flow cytometry. The level of protein expression was detected by western blot. The results demonstrated that pterostilbene significantly inhibited the growth of T-cell leukemia/lymphoma cell lines in vitro and induced apoptosis in a dose- and time-dependent manner. Moreover, pterostilbene treatment markedly induced S-phase cell cycle arrest, which was accompanied by downregulation of cdc25A, cyclin A2, and CDK2. Pterostilbene also induced the generation of reactive oxygen species and the loss of mitochondrial membrane potential and inhibited ERK1/2 phosphorylation. Taken together, our study demonstrated the potential of pterostilbene to be an effective treatment for T-cell leukemia/lymphoma.


Subject(s)
Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Leukemia, T-Cell/enzymology , Leukemia, T-Cell/pathology , MAP Kinase Signaling System/drug effects , Stilbenes/pharmacology , Caspases/metabolism , Cell Proliferation/drug effects , Humans , Indazoles/pharmacology , Jurkat Cells , Membrane Potential, Mitochondrial/drug effects , Phosphorylation/drug effects , Piperazines/pharmacology , Reactive Oxygen Species/metabolism
4.
Oncotarget ; 8(15): 24706-24718, 2017 Apr 11.
Article in English | MEDLINE | ID: mdl-28160557

ABSTRACT

Many human cancers have been reported to have enhanced expression of the immune checkpoint molecule programmed death-ligand 1 (PD-L1), which binds to programmed cell death-1 (PD-1) expressed on immune cells. PD-L1/PD-1 plays a role in inhibition of antitumor immunity by inducing T cell apoptosis and tolerance. Thus, it is crucial to elucidate mechanisms of PD-L1 expression on cancer cells. ERO1-α is an oxidase located in the endoplasmic reticulum. It is overexpressed in a variety of tumor types and it plays a role in disulfide bond formation in collaboration with PDI. Here, we investigated the influence of ERO1-α on expression of PD-L1 and immune escape. We demonstrated that ERO1-α augmented the expression of PD-L1 via facilitation of oxidative protein folding within PD-L1. In addition, we showed that overexpression of ERO1-α increased HIF-1α protein expression, resulting in an increase of PD-L1 mRNA as well as protein. In clinical cases, we observed that the expression of ERO1-α in triple negative breast cancer was related to the expression of PD-L1. Moreover, apoptosis of Jurkat leukemia T cells, which express PD-1, induced by tumor PD-L1 was inhibited when ERO1-α was depleted. The results suggest that targeting ERO1-α in tumor cells can be a novel approach for cancer immunotherapy. Therefore, the role of ERO1-α in tumor-mediated immunosuppression should be further explored.


Subject(s)
B7-H1 Antigen/immunology , Breast Neoplasms/immunology , Leukemia, T-Cell/immunology , Membrane Glycoproteins/immunology , Oxidoreductases/immunology , Apoptosis/immunology , B7-H1 Antigen/biosynthesis , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Interferon-gamma/pharmacology , Jurkat Cells , Leukemia, T-Cell/enzymology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/biosynthesis , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/biosynthesis , Protein Folding , Transfection , Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/immunology , Tumor Escape , Up-Regulation/drug effects , Up-Regulation/immunology
5.
Adv Biol Regul ; 60: 115-121, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26527132

ABSTRACT

Replicative stress (RS) is a cell-intrinsic phenomenon enhanced by oncogenic transformation. Checkpoint kinase 1 (CHK1) is a key component of the ATR-dependent DNA damage response pathway that protects cells from RS by preventing replication fork collapse and activating homologous DNA repair. Taking this knowledge into account, one would predict CHK1 behaves strictly as a tumor suppressor. However, the reality seems far more complex. CHEK1 loss-of-function mutations have not been found in human tumors, and transgenic expression of Chek1 in mice promotes oncogene-induced transformation through RS inhibition. Moreover, CHK1 is overexpressed in various human cancers and CHK1 inhibitors have been developed as sensitizers to enhance the cytotoxicity of DNA damage-inducing chemotherapies. Here, we summarize the literature on the involvement of CHK1 in cancer progression, including our recent observation that CHK1 sustains T-cell acute lymphoblastic leukemia (T-ALL) cell viability. We also debate the importance of identifying patients that could benefit the most from treatment with CHK1 inhibitors, taking T-ALL as a model, and propose possible markers of therapeutic response.


Subject(s)
Checkpoint Kinase 1/metabolism , Genes, Tumor Suppressor , Leukemia, T-Cell/enzymology , Oncogenes , Animals , Checkpoint Kinase 1/genetics , DNA Replication , Humans , Leukemia, T-Cell/genetics , Mice
6.
J Med Chem ; 58(11): 4812-21, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25974739

ABSTRACT

Histone deacetylase inhibitors (HDACi) target abnormal epigenetic states associated with a variety of pathologies, including cancer. Here, the development of a prodrug of the canonical broad-spectrum HDACi suberoylanilide hydroxamic acid (SAHA) is described. Although hydroxamic acids are utilized universally in the development of metalloenzyme inhibitors, they are considered to be poor pharmacophores with reduced activity in vivo. We developed a prodrug of SAHA by appending a promoiety, sensitive to thiols, to the hydroxamic acid warhead (termed SAHA-TAP). After incubation of SAHA-TAP with an HDAC, the thiol of a conserved HDAC cysteine residue becomes covalently tagged with the promoiety, initiating a cascade reaction that leads to the release of SAHA. Mass spectrometry and enzyme kinetics experiments validate that the cysteine residue is covalently appended with the TAP promoiety. SAHA-TAP demonstrates cytotoxicity activity against various cancer cell lines. This strategy represents an original prodrug design with a dual mode of action for HDAC inhibition.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Prodrugs/pharmacology , Repressor Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Blotting, Western , Cells, Cultured , Chromatography, Liquid , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases , Humans , Hydroxamic Acids/chemistry , Leukemia, T-Cell/drug therapy , Leukemia, T-Cell/enzymology , Leukemia, T-Cell/pathology , Mice , Models, Molecular , Molecular Structure , Prodrugs/chemistry , Structure-Activity Relationship , Tandem Mass Spectrometry , Vorinostat
7.
Biochem Biophys Res Commun ; 453(3): 569-75, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25285634

ABSTRACT

Present-day oncology sees at least two-thirds of cancer patients receiving radiation therapy as a part of their anticancer treatment. The objectives of the current study were to investigate the effects of the small molecule inhibitors of Wee1 kinase II (681641) and Rad51 (RI-1) on cell cycle progression, DNA double-strand breaks repair and apoptosis following ionizing radiation exposure in human leukemic T-cells Jurkat and MOLT-4. Pre-treatment with the Wee1 681641 or Rad51 RI-1 inhibitor alone increased the sensitivity of Jurkat cells to irradiation, however combining both inhibitors together resulted in a further enhancement of apoptosis. Jurkat cells pre-treated with inhibitors were positive for γH2AX foci 24h upon irradiation. MOLT-4 cells were less affected by inhibitors application prior to ionizing radiation exposure. Pre-treatment with Rad51 RI-1 had no effect on apoptosis induction; however Wee1 681641 increased ionizing radiation-induced cell death in MOLT-4 cells.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , DNA Damage/radiation effects , Leukemia, T-Cell/enzymology , Nuclear Proteins/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Rad51 Recombinase/antagonists & inhibitors , DNA Repair , Humans , Jurkat Cells , Leukemia, T-Cell/genetics , Leukemia, T-Cell/pathology , Protein Kinase Inhibitors/pharmacology , Radiation, Ionizing
8.
Clin Lab ; 60(1): 1-8, 2014.
Article in English | MEDLINE | ID: mdl-24600969

ABSTRACT

BACKGROUND: AKT1 is a member of the serine/threoine AGC protein kinase family involved in cancer's metabolism, growth, proliferation, and survival. It is a potential target for cancer gene therapy. METHODS: In the present study, we used DNAzyme and siRNA technology to inhibit AKT1 expression and evaluated the effects of DNAzymes and siRNA as therapeutic agents to treat leukemic cells. We designed two AKT1 specific DNAzymes (DRz1 and DRz2) and siRNA to test their effects on the apoptosis of leukemic cells. RESULTS: Here, we showed that DRz1 could down-regulate the expression of AKT1 in both mRNA and protein levels, hence significantly inhibiting growth and inducing apoptosis of Jurkat cells. CONCLUSIONS: These results provide a significant insight into the potential anticarcinogenic action of DNAzyme against AKT1 which might be used as a valuable therapy to leukemia.


Subject(s)
Cell Division , DNA, Catalytic/metabolism , Leukemia, T-Cell/pathology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Base Sequence , Cell Line, Tumor , DNA Primers , Humans , Leukemia, T-Cell/enzymology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction
9.
Apoptosis ; 19(1): 224-40, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24166139

ABSTRACT

Treatment of Jurkat T cells with the microtubule-depolymerizing agent nocodazole (NOC) caused prometaphase arrest and apoptosis. NOC-induced mitochondrial apoptotic events including Bak activation, Δψm loss, cytochrome c release, and caspase cascade activation were blocked by Bcl-2 overexpression. However, mitotic arrest, Cdc25C activation, upregulation of cyclin B1 levels, Cdk1 activation, Bcl-2 phosphorylation at Thr-56 and Ser-70, and Bim phosphorylation were retained. The treatment of Jurkat T cells concomitantly with NOC and the G1/S-blocking agent hydroxyurea resulted in G1/S arrest and complete abrogation of all apoptotic events. The association of Bcl-2 with Bim or Bak declined after the prometaphase arrest-dependent phosphorylation of Bcl-2 and Bim, whereas the association of Bcl-2 with Bax remained relatively constant. Although Bax was redistributed from the cytosol to the mitochondria, resulting in an increase in the mitochondrial level of Bax following NOC treatment, the subcellular localization of Bcl-2, Bim, Bak and apoptosis-inducing factor was confined to the mitochondrial fraction irrespective of NOC treatment. Experiments using selective caspase inhibitors showed that mitochondria-dependent activation of caspase-9 and -3 was crucial for NOC-induced apoptosis. NOC-induced phosphorylation of Bcl-2 and Bim, Δψm loss, and mitochondria-dependent apoptotic events were significantly suppressed by a Cdk1 inhibitor roscovitine, but not by the JNK inhibitor SP600125 or the p38 MAPK inhibitor SB203580. These results show that the prometaphase arrest-dependent phosphorylation of Bcl-2 and Bim, which was mediated by Cdk1, could reduce the association of Bcl-2 with Bak or Bim to allow Bak activation and mitochondrial apoptotic events in Jurkat T cells exposed to NOC.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Jurkat Cells/drug effects , Leukemia, T-Cell/enzymology , Membrane Proteins/metabolism , Nocodazole/pharmacology , Prometaphase/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Line, Tumor , Humans , Jurkat Cells/cytology , Jurkat Cells/metabolism , Leukemia, T-Cell/drug therapy , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/physiopathology , M Phase Cell Cycle Checkpoints/drug effects , Membrane Proteins/genetics , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , bcl-2 Homologous Antagonist-Killer Protein/genetics
10.
Chem Biol Interact ; 205(3): 198-211, 2013 Oct 05.
Article in English | MEDLINE | ID: mdl-23911876

ABSTRACT

In this study the role of PI3K/Akt signaling pathway in arsenic trioxide (ATO)-treated parental Jurkat cells and also in derived ATO-resistant clones grown in the presence of given ATO concentration was investigated. ATO-resistant clones (cultured for 8-12weeks in the presence of 1, 2.5 and 5µM ATO) were characterized by high viability in the presence of ATO but slower growth rate compared to the parental cells. Morphological and functional characterization of derived ATO-resistant clones revealed that they did not differ fundamentally from parental Jurkat cells in terms of cell size, level of GSH, the lysosomal fluorescence or CD95/Fas surface antigen expression. However, a slight increase in the mitochondrial potential (JC-1 staining) was detected in the clones compared to parental Jurkat cells. Side population analysis (Vybrant DyeCycle Violet™ staining) in ATO resistant clones did not indicate any enrichment withcancer stem cells. Akt1/2, AktV or wortmannin inhibitors decreased viability of ATO-resistant clones grown in the presence of ATO, with no effect on ATO-treated parental cells. Flow cytometry analysis showed that ATO decreased the level of p-Akt in ATO-treated parental cells, while the resistant clones exhibited higher levels of p-Akt immunostaining than parental Jurkat cells. Expression analysis of 84 genes involved in the PI3K/Akt pathway revealed that this pathway was predominantly active in ATO-resistant clones. c-JUN seems to play a key role in the induction of cell death in ATO-treated parental Jurkat cells, as dose-dependent strong up-regulation of JUN was specific for the ATO-treated parental Jurkat cells. On the other hand, changes in expression of cyclin D1 (CCND1), insulin receptor substrate 1 (IRS1) and protein kinase C isoforms (PRKCZ,PRKCB and PRKCA) may be responsible for the induction of resistance to ATO. The changes in expression of growth factor receptor-bound protein 10 (GRB10) observed in ATO-resistant clones suggest a possibility of induction of different mechanisms in development of resistance to ATO depending on the drug concentration and thus involvement of different signaling mediators.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenicals/pharmacology , Leukemia, T-Cell/drug therapy , Oxides/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/physiology , Arsenic Trioxide , Cell Survival/drug effects , Clone Cells , Flow Cytometry , Gene Expression Profiling , Humans , Jurkat Cells , Leukemia, T-Cell/enzymology , Leukemia, T-Cell/pathology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , RNA, Neoplasm/chemistry , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction
11.
Food Chem Toxicol ; 50(8): 2916-22, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22613213

ABSTRACT

Inophyllin A (INO-A), a pyranoxanthone isolated from the roots of Calophyllum inophyllum represents a new xanthone with potential chemotherapeutic activity. In this study, the molecular mechanism of INO-A-induced cell death was investigated in Jurkat T lymphoblastic leukemia cells. Assessment of phosphatidylserine exposure confirmed apoptosis as the primary mode of cell death in INO-A-treated Jurkat cells. INO-A treatment for only 30 min resulted in a significant increase of tail moment which suggests that DNA damage is an early apoptotic signal. Further flow cytometric assessment of the superoxide anion level confirmed that INO-A induced DNA damage was mediated with a concomitant generation of reactive oxygen species (ROS). Investigation on the thiols revealed an early decrease of free thiols in 30 min after 50 µM INO-A treatment. Using tetramethylrhodamine ethyl ester, a potentiometric dye, the loss of mitochondrial membrane potential (MPP) was observed in INO-A-treated cells as early as 30 min. The INO-A-induced apoptosis progressed with the simultaneous activation of caspases-2 and -9 which then led to the processing of caspase-3. Taken together, these data demonstrate that INO-A induced early oxidative stress, DNA damage and loss of MMP which subsequently led to the activation of an intrinsic pathway of apoptosis in Jurkat cells.


Subject(s)
Apoptosis/drug effects , Leukemia, T-Cell/metabolism , Oxidative Stress/drug effects , Xanthones/pharmacology , Caspases/metabolism , DNA Damage , Enzyme Activation , Flow Cytometry , Humans , Jurkat Cells , Leukemia, T-Cell/enzymology , Leukemia, T-Cell/pathology , Reactive Oxygen Species/metabolism
12.
Biochem Biophys Res Commun ; 410(3): 422-7, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21669187

ABSTRACT

Canertinib is a novel ErbB-receptor inhibitor currently in clinical development for the treatment of solid tumors overexpressing ErbB-receptors. We have recently demonstrated that canertinib displays anti-proliferative and pro-apoptotic effects in human myeloid leukemia cells devoid of ErbB-receptors. The mechanism mediating these effects are however unknown. In this study, we show that canertinib is able to act as a multi-kinase inhibitor by inhibition of several intracellular kinases involved in T-cell signaling such as Akt, Erk1/2 and Zap-70, and reduced Lck protein expression in the human T-cell leukemia cell line Jurkat. Treatment with canertinib at a concentration of 2 µM caused accumulation of Jurkat cells in the G(1) cell cycle phase and increased doses induced apoptosis in a time-dependent manner. Apoptotic signs of treated cells were detected by Annexin V staining and cleavage of PARP, caspase-3, -8, -9, -10 and Bid. A subset of the pro-apoptotic signals mediated by canertinib could be significantly reduced by specific caspase inhibitors. Taken together, these results demonstrate the dual ability of canertinib to downregulate important signaling pathways and to activate caspase-mediated intrinsic apoptosis pathway in human T-cell leukemia cells.


Subject(s)
Apoptosis , Caspases/metabolism , Leukemia, T-Cell/enzymology , Morpholines/pharmacology , Protein Kinase Inhibitors/pharmacology , Humans , Jurkat Cells , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Antigen, T-Cell/metabolism , ZAP-70 Protein-Tyrosine Kinase/metabolism
13.
Autophagy ; 6(6): 711-24, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20543569

ABSTRACT

Suberoylanilide hydroxamic acid (SAHA) is a newly emerging histone deacetylase inhibitor (HDACi) and has been approved in phase II clinical trials for treating patients with cutaneous T-cell lymphoma. Autophagy is a conserved self-digestion process that degrades cytoplasmic materials and recycles long-lived proteins and organelles within cells. In this study, we demonstrate that SAHA stimulates autophagy in Jurkat T-leukemia cells, which was evidenced by the appearance of autophagic vacuoles, formation of acidic vesicular organelles, recruitment of LC3-II to the autophagosomes and conversion of LC3-I to LC3-II . Moreover, SAHA treatment upregulated expression of Beclin 1 and Atg7 and promoted formation of the Atg12-Atg5 conjugate. Furthermore, inhibition of autophagy by chloroquine (CQ) enhanced SAHA-induced apoptosis. To determine the underlying mechanism of SAHA-induced autophagy, two complementary proteomic approaches (2-DE and SILAC), coupled with ESI-Q-TOF MS/MS analysis are utilized to profile differentially expressed proteins between control and SAHA-treated Jurkat T-leukemia cells. In total, 72 proteins were identified with significant alterations. Cluster analysis of the changed proteins reveal several groups of enzymes associated with energy metabolism, anti-oxidative stress and cellular redox control, which suggested an abnormal reactive oxygen species (ROS) production in SAHA-treated Jurkat T-leukemia cells. These observations were further confirmed by ROS chemiluminescence assay. Mechanistic studies revealed that SAHA-triggered autophagy was mediated by ROS production, which could be attenuated by N-acetyl cysteine (NAC), a ROS inhibitor. Finally, we illustrated that Akt-mTOR signaling, a major suppressive cascade of autophagy, was inactivated by SAHA treatment. Taken together, our study identifies autophagy as a reaction to counter increased ROS and is thus involved as a cellular prosurvival mechanism in response to SAHA treatment.


Subject(s)
Autophagy/drug effects , Hydroxamic Acids/pharmacology , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/pathology , Proteomics/methods , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Apoptosis/drug effects , Autophagy/genetics , Gene Expression Regulation, Leukemic/drug effects , Green Fluorescent Proteins/metabolism , Humans , Immunoblotting , Jurkat Cells , Leukemia, T-Cell/enzymology , Leukemia, T-Cell/genetics , Microtubule-Associated Proteins/metabolism , Models, Biological , Neoplasm Proteins/metabolism , Phagosomes/drug effects , Phagosomes/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Vacuoles/drug effects , Vacuoles/metabolism , Vorinostat
14.
Biochem Pharmacol ; 79(3): 321-9, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19737543

ABSTRACT

Stimulation of the cAMP signaling pathway has been shown to induce apoptosis and augment the effects of glucocorticoids in inducing apoptosis in leukemic cells. We recently reported that in primary B cell chronic lymphocytic leukemic (B-CLL) cells, apoptosis could be induced by stimulating the cAMP signaling pathway with a phosphodiesterase4 (PDE4) inhibitor alone; while in contrast, in the CEM T leukemic cell line, PDE4 inhibitors alone were ineffective, and concurrent stimulation of adenylyl cyclase was required to see effects [Tiwari et al. (2005)]. We report here that in the CEM and Jurkat T leukemic cell lines, the most abundantly expressed PDEs are PDE3B, PDE4A, PDE4D, PDE7A, and PDE8A. Selective inhibition of PDE3, PDE4 or PDE7 alone produces little effect on cell viability, but inhibition of all three of these PDEs together dramatically enhances glucocorticoid-induced apoptosis in CEM cells, and overcomes glucocorticoid resistance in a glucocorticoid-resistant CEM cell line. These studies indicate that for some leukemic cell types, a desired therapeutic effect may be achieved by inhibiting more than one form of PDE.


Subject(s)
Apoptosis/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 7/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Glucocorticoids/pharmacology , Leukemia, T-Cell/enzymology , Phosphodiesterase 3 Inhibitors , Phosphodiesterase 4 Inhibitors , Apoptosis/physiology , Cyclic Nucleotide Phosphodiesterases, Type 3/physiology , Cyclic Nucleotide Phosphodiesterases, Type 4/physiology , Cyclic Nucleotide Phosphodiesterases, Type 7/physiology , Drug Resistance, Neoplasm/physiology , Drug Synergism , Humans , Jurkat Cells , Leukemia, T-Cell/drug therapy , Phosphodiesterase Inhibitors/pharmacology , Tumor Cells, Cultured
15.
Biochem Pharmacol ; 78(2): 191-202, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19442964

ABSTRACT

Na,K-ATPase is a ubiquitous transmembrane protein that regulates and maintains the intracellular Na(+) and K(+) gradient necessary for cell homeostasis. Recently, the importance of this pump in external stimuli-induced leukemia cell apoptosis has been increasingly appreciated, however, the exact role of Na,K-ATPase in mitochondrial apoptotic pathway still remains little understood. In this study, we found mitochondrial toxin rotenone caused a rapid mitochondrial membrane potential (MMP) collapse in Jurkat cells followed by plasma membrane depolarization (PMP). Similar results were also obtained in human U937 cells and non-cancerous mouse primary T cells. Rotenone-induced PMP depolarization occurred before apoptosis and well correlated with Na,K-ATPase impairment. To understand the mechanisms, Jurkat cells with mtDNA depletion and catalase overexpression were used. The results demonstrated that both PMP depolarization and Na,K-ATPase impairment induced by rotenone were regulated by mitochondrial H(2)O(2) and Bcl-2. Finally, Na,K-ATPase suppression by ouabain greatly accelerated and enhanced mitochondrial toxins-induced cells apoptosis in Jurkat, U937 and primary T cells. In sum, by using leukemia cells and mouse primary T cells, we confirmed that mitochondria-to-Na,K-ATPase and PMP depolarization might represent a novel mechanism for mitochondria to amplify death signals in the initiation stage of cells apoptosis induced by mitochondrial toxins.


Subject(s)
Apoptosis/drug effects , Cell Membrane/drug effects , Leukemia, T-Cell/enzymology , Leukemia, T-Cell/pathology , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/enzymology , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Animals , Apoptosis/physiology , Cell Membrane/enzymology , Cell Survival/drug effects , Cell Survival/physiology , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/enzymology , Intracellular Membranes/metabolism , Jurkat Cells , Mice , Mice, Transgenic , Mitochondrial Membranes/pathology , Rotenone/toxicity , Sodium-Potassium-Exchanging ATPase/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/enzymology , U937 Cells
16.
Leukemia ; 23(8): 1507-14, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19282831

ABSTRACT

Interactions between inhibitors of the proteasome and histone deacetylases have been examined in human T-leukemia/lymphoma cells both in vitro and in vivo. Co-exposure of cells to bortezomib and suberoylanilide hydroxamic acid (SAHA) synergistically induces T-leukemia/lymphoma cells to undergo apoptosis, consistent with a significant increase in mitochondrial injury and caspase activation. These events are accompanied by inhibition of cyto-protective signaling pathways, including the nuclear factor (NF)-kappaB, Raf-1/mitogen-induced extracellular kinase (MEK)/extracellular signal-related kinase (ERK) and AKT pathways, and activation of stress-related cascades, including the stress-activated kinases c-jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (p38MAPK). Moreover, bortezomib in conjunction with SAHA efficiently induces apoptosis of primary T-leukemia/lymphoma cells and inhibits tumor growth in a murine xenograft model established with subcutaneous injection of Jurkat cells. Taken together, these findings confirm the synergistic anti-tumor effect of the proteasome and histone deacetylase inhibitors, and provide an insight into the future clinical applications of bortezomib-SAHA combining regimen in treating T-cell malignancies.


Subject(s)
Boronic Acids/pharmacology , Histone Deacetylase Inhibitors , Hydroxamic Acids/pharmacology , Leukemia, T-Cell/pathology , Lymphoma, T-Cell/pathology , Neoplasm Proteins/antagonists & inhibitors , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Pyrazines/pharmacology , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Bortezomib , Cell Line, Tumor/drug effects , Cell Line, Tumor/pathology , Drug Synergism , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/physiology , Jurkat Cells/drug effects , Jurkat Cells/enzymology , Jurkat Cells/transplantation , Leukemia, T-Cell/drug therapy , Leukemia, T-Cell/enzymology , Lymphoma, T-Cell/enzymology , Mice , Mice, Nude , Protein Kinases/physiology , Vorinostat , Xenograft Model Antitumor Assays
17.
Cell Res ; 19(3): 358-69, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19048023

ABSTRACT

Methionine adenosyltransferase II (MAT II) is a key enzyme in cellular metabolism and catalyzes the formation of S-adenosylmethionine (SAMe) from L-methionine and ATP. Normal resting T lymphocytes have minimal MAT II activity, whereas activated proliferating T lymphocytes and transformed T leukemic cells show significantly enhanced MAT II activity. This work was carried out to examine the role of MAT II activity and SAMe biosynthesis in the survival of leukemic T cells. Inhibition of MAT II and the resultant decrease in SAMe levels enhanced expression of FasL mRNA and protein, and induced DISC (Death Inducing Signaling Complex) formation with FADD (Fas-associated Death Domain) and procaspase-8 recruitment, as well as concomitant increase in caspase-8 activation and decrease in c-FLIP(s) levels. Fas-initiated signaling induced by MAT II inhibition was observed to link to the mitochondrial pathway via Bid cleavage and to ultimately lead to increased caspase-3 activation and DNA fragmentation in these cells. Furthermore, blocking MAT 2A mRNA expression, which encodes the catalytic subunits of MAT II, using a small-interfering RNA approach enhanced FasL expression and cell death, validating the essential nature of MAT II activity in the survival of T leukemic cells.


Subject(s)
Apoptosis , Caspase 8/metabolism , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Fas Ligand Protein/metabolism , Leukemia, T-Cell/enzymology , Leukemia, T-Cell/pathology , Methionine Adenosyltransferase/antagonists & inhibitors , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 3/metabolism , Cell Survival/drug effects , Cycloleucine/pharmacology , Enzyme Activation/drug effects , Fas-Associated Death Domain Protein/metabolism , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Membrane Potential, Mitochondrial/drug effects , Neutralization Tests , S-Adenosylmethionine/metabolism
18.
Int J Oncol ; 33(5): 1073-9, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18949371

ABSTRACT

Interest in the use of metallic compounds for cancer treatment has been increasing since the discovery of cisplatin. Clinical studies suggest the use of proteasome inhibitors as potential novel anticancer agents. L-glutamine is the most abundant free amino acid in the body, and has been shown to play a regulatory role in several cellular processes, including metabolism, degradation, redox potential and cellular integrity. Although glutamine is reported to play a role in the regulation of apoptosis, the effect of glutamine copper complex on tumor cells and the involved molecular mechanism have not been investigated. Here, for the first time, we report that a newly synthesized L-glutamine-containing copper complex has proteasome-inhibitory activity in human breast cancer and leukemia cells. The inhibition of the tumor proteasomal activity results in the accumulation of ubiquitinated proteins and ubiquitinated form of IkappaB-alpha, a natural proteasome substrate, followed by induction of apoptosis. Furthermore, this glutamine Schiff base copper complex selectively inhibits the proteasomal activity and induces cell death in cultured breast cancer cells, but not normal, immortalized breast cells. Our data suggest that glutamine Schiff base copper complexes have a potential use for to be used in cancer treatment and prevention.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/pathology , Cysteine Proteinase Inhibitors/pharmacology , Glutamine/analogs & derivatives , Leukemia, T-Cell/pathology , Organometallic Compounds/pharmacology , Proteasome Inhibitors , Breast Neoplasms/enzymology , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Glutamine/pharmacology , Humans , I-kappa B Proteins/metabolism , Jurkat Cells , Kinetics , Leukemia, T-Cell/enzymology , NF-KappaB Inhibitor alpha , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism
19.
Mol Cancer ; 7: 57, 2008 Jun 17.
Article in English | MEDLINE | ID: mdl-18559081

ABSTRACT

BACKGROUND: Certain endogenous metabolites can influence the rate of cancer cell growth. For example, diacylglycerol, ceramides and sphingosine, NAD+ and arginine exert this effect by acting as signaling molecules, while carrying out other important cellular functions. Metabolites can also be involved in the control of cell proliferation by directly regulating gene expression in ways that are signaling pathway-independent, e.g. by direct activation of transcription factors or by inducing epigenetic processes. The fact that metabolites can affect the cancer process on so many levels suggests that the change in concentration of some metabolites that occurs in cancer cells could have an active role in the progress of the disease. RESULTS: CoMet, a fully automated Computational Metabolomics method to predict changes in metabolite levels in cancer cells compared to normal references has been developed and applied to Jurkat T leukemia cells with the goal of testing the following hypothesis: Up or down regulation in cancer cells of the expression of genes encoding for metabolic enzymes leads to changes in intracellular metabolite concentrations that contribute to disease progression. All nine metabolites predicted to be lowered in Jurkat cells with respect to lymphoblasts that were examined (riboflavin, tryptamine, 3-sulfino-L-alanine, menaquinone, dehydroepiandrosterone, alpha-hydroxystearic acid, hydroxyacetone, seleno-L-methionine and 5,6-dimethylbenzimidazole), exhibited antiproliferative activity that has not been reported before, while only two (bilirubin and androsterone) of the eleven tested metabolites predicted to be increased or unchanged in Jurkat cells displayed significant antiproliferative activity. CONCLUSION: These results: a) demonstrate that CoMet is a valuable method to identify potential compounds for experimental validation, b) indicate that cancer cell metabolism may be regulated to reduce the intracellular concentration of certain antiproliferative metabolites, leading to uninhibited cellular growth and c) suggest that many other endogenous metabolites with important roles in carcinogenesis are awaiting discovery.


Subject(s)
Cell Proliferation , Leukemia, T-Cell/metabolism , Systems Biology , Antimetabolites, Antineoplastic/pharmacology , Cell Proliferation/drug effects , Disease Progression , Drug Design , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Jurkat Cells , Leukemia, T-Cell/enzymology , Leukemia, T-Cell/genetics , Leukemia, T-Cell/pathology , Reproducibility of Results
20.
Cell Biochem Funct ; 26(1): 87-94, 2008.
Article in English | MEDLINE | ID: mdl-17566139

ABSTRACT

Fatty acid (FA) may disturb the redox state of the cells not only by an increase in reactive oxygen species (ROS) generation but also due to a reduction in antioxidant enzyme activities. The effect of various FAs (palmitic, stearic, oleic, linoleic, gamma-linolenic and eicosapentaenoic acids (EPAs)) on Jurkat and Raji cells, (human T and B leukaemic cell lines was investigated). The following measurements were carried out: FA composition of the cells, cell proliferation and activities of catalase, glutathione peroxidase (GPx) and superoxide dismutase (SOD). The protective effect of alpha-tocopherol on cell death was also investigated. Each cell line presented a specific FA composition. All the tested FAs reduced catalase activity. The toxic effect of FA was abolished by the pre-incubation with physiological concentrations of alpha-tocopherol. The findings support the proposition that the increase in oxidative stress induced by FA partially occurs due to a reduction in catalase activity. In spite of the decrease in the enzyme activity, catalase protein and mRNA levels were not changed, suggesting a post-translational regulation.


Subject(s)
Catalase/antagonists & inhibitors , Catalase/metabolism , Fatty Acids/physiology , Leukemia/enzymology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Fatty Acids/pharmacology , Fatty Acids, Unsaturated/physiology , Humans , Jurkat Cells , Leukemia/metabolism , Leukemia, B-Cell/enzymology , Leukemia, T-Cell/enzymology , Oxidative Stress/drug effects , Oxidative Stress/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...