Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
2.
PLoS One ; 2(12): e1313, 2007 Dec 12.
Article in English | MEDLINE | ID: mdl-18074035

ABSTRACT

BACKGROUND: Nitrosylcobalamin (NO-Cbl) is a chemotherapeutic pro-drug derived from vitamin B12 that preferentially delivers nitric oxide (NO) to tumor cells, based upon increased receptor expression. NO-Cbl induces Apo2L/TRAIL-mediated apoptosis and inhibits survival signaling in a variety of malignant cell lines. Chemotherapeutic agents often simultaneously induce an apoptotic signal and activation of NF-kappaB, which has the undesired effect of promoting cell survival. The specific aims of this study were to 1) measure the anti-tumor effects of NO-Cbl alone and in combination with conventional chemotherapeutic agents, and to 2) examine the mechanism of action of NO-Cbl as a single agent and in combination therapy. METHODOLOGY: Using anti-proliferative assays, electrophoretic mobility shift assay (EMSA), immunoblot analysis and kinase assays, we demonstrate an increase in the effectiveness of chemotherapeutic agents in combination with NO-Cbl as a result of suppressed NF-kappaB activation. RESULTS: Eighteen chemotherapeutic agents were tested in combination with NO-Cbl, in thirteen malignant cell lines, resulting in a synergistic anti-proliferative effect in 78% of the combinations tested. NO-Cbl pre-treatment resulted in decreased NF-kappaB DNA binding activity, inhibition of IkappaB kinase (IKK) enzymatic activity, decreased AKT activation, increased caspase-8 and PARP cleavage, and decreased cellular XIAP protein levels. CONCLUSION: The use of NO-Cbl to inhibit survival signaling may enhance drug efficacy by preventing concomitant activation of NF-kappaB or AKT.


Subject(s)
Antineoplastic Agents/pharmacology , Nitroso Compounds/pharmacology , Signal Transduction/drug effects , Vitamin B 12/analogs & derivatives , Apolipoproteins/physiology , Apoptosis/drug effects , Apoptosis/physiology , Cell Line, Tumor , Electrophoretic Mobility Shift Assay , Enzyme Activation , Humans , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TNF-Related Apoptosis-Inducing Ligand/physiology , Vitamin B 12/pharmacology , X-Linked Inhibitor of Apoptosis Protein/metabolism
3.
J Biol Chem ; 282(21): 15349-56, 2007 05 25.
Article in English | MEDLINE | ID: mdl-17379600

ABSTRACT

We previously showed that inositol hexakisphosphate kinase 2 (IHPK2) functions as a growth-suppressive and apoptosis-enhancing kinase during cell stress. Overexpression of IHPK2 sensitized ovarian carcinoma cell lines to the growth-suppressive and apoptotic effects of interferon beta (IFN-beta), IFN-alpha2, and gamma-irradiation. Expression of a kinase-dead mutant abrogated 50% of the apoptosis induced by IFN-beta. Because the kinase-dead mutant retained significant response to cell stressors, we hypothesized that a portion of the death-promoting function of IHPK2 was independent of its kinase activity. We now demonstrate that IHPK2 binds to tumor necrosis factor (TNF) receptor-associated factor (TRAF) 2 and interferes with phosphorylation of transforming growth factor beta-activated kinase 1 (TAK1), thereby inhibiting NF-kappaB signaling. IHPK2 contains two sites required for TRAF2 binding, Ser-347 and Ser-359. Compared with wild type IHPK2-transfected cells, cells expressing S347A and S359A mutations displayed 3.5-fold greater TAK1 activation following TNF-alpha. This mutant demonstrated a 6-10-fold increase in NF-kappaB DNA binding following TNF-alpha compared with wild type IHPK2-expressing cells in which NF-kappaB DNA binding was inhibited. Cells transfected with wild type IHPK2 or IHPK2 mutants that lacked S347A and S359A mutations displayed enhanced terminal deoxynucleotidyltransferase-mediated dUTP nick end-labeling staining following TNF-alpha. We believe that IHPK2-TRAF2 binding leads to attenuation of TAK1- and NF-kappaB-mediated signaling and is partially responsible for the apoptotic activity of IHPK2.


Subject(s)
Apoptosis , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Phosphotransferases (Phosphate Group Acceptor)/metabolism , TNF Receptor-Associated Factor 2/metabolism , Amino Acid Substitution , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/radiation effects , Cell Line, Tumor , Gamma Rays , Gene Expression , Humans , Interferon-alpha/pharmacology , Interferon-beta/pharmacology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/radiation effects , Mice , Mutation, Missense , NF-kappa B/metabolism , Phosphotransferases (Phosphate Group Acceptor)/genetics , Protein Binding/genetics , TNF Receptor-Associated Factor 2/genetics
4.
Biochem J ; 385(Pt 2): 595-603, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15634191

ABSTRACT

Previously, we have reported that overexpression of IHPK2 (inositol hexakisphosphate kinase 2) sensitized NIH-OVCAR-3 ovarian carcinoma cell lines to the growth-suppressive and apoptotic effects of IFN-beta (interferon-beta) treatment and gamma-irradiation. In the present study, we demonstrate that Apo2L/TRAIL (Apo2L/tumour-necrosis-factor-related apoptosis-inducing ligand) is a critical mediator of IFN-induced apoptosis in these cells. Compared with IFN-alpha2, IFN-beta is a more potent inducer of Apo2L/TRAIL and IHPK2 activity. Overexpression of IHPK2 converts IFN-alpha2-resistant cells into cells that readily undergo apoptosis in response to IFN-alpha2. In untreated cells transfected with IHPK2-eGFP (where eGFP stands for enhanced green fluorescent protein), the fusion protein is localized to the cytoplasm and perinuclear region. After treatment with IFN-beta, IHPK2-eGFP translocated to the nucleus. In cells transfected with mutant IHPK2-NLS-eGFP (where NLS stands for nuclear localization sequence), containing point mutations in the NLS, the fusion protein remained trapped in the cytoplasm, even after IFN-beta treatment. Cells expressing mutant NLS mutation were more resistant to IFN-beta. The IC50 value of IHPK2-expressing cells was 2-3-fold lower than vector control. The IC50 value of NLS-mutant-expressing cells was 3-fold higher than vector control. Blocking antibodies to Apo2L/TRAIL or transfection with a dominant negative Apo2L/TRAIL receptor (DR5Delta) inhibited the antiproliferative effects of IFN-beta. Thus overexpression of IHPK2 enhanced apoptotic effects of IFN-beta, and expression of the NLS mutant conferred resistance to IFN-beta. Apo2L/TRAIL expression and nuclear localization of IHPK2 are both required for the induction of apoptosis by IFN-beta in ovarian carcinoma.


Subject(s)
Active Transport, Cell Nucleus/physiology , Apoptosis/physiology , Carcinoma/metabolism , Interferon-beta/physiology , Membrane Glycoproteins/metabolism , Ovarian Neoplasms/metabolism , Phosphotransferases (Phosphate Group Acceptor)/metabolism , Tumor Necrosis Factor-alpha/metabolism , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Carcinoma/pathology , Cell Line, Tumor , Cell Proliferation , Female , Humans , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/immunology , Ovarian Neoplasms/pathology , Phosphotransferases (Phosphate Group Acceptor)/biosynthesis , Protein Transport/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
5.
J Biol Chem ; 278(41): 39461-9, 2003 Oct 10.
Article in English | MEDLINE | ID: mdl-12881518

ABSTRACT

We have previously demonstrated the anti-tumor activity of nitrosylcobalamin (NO-Cbl), an analog of vitamin B12 that delivers nitric oxide (NO) and increases the expression of tumor necrosis factor-related apoptosis-inducing ligand (Apo2L/TRAIL) and its receptors in human tumors. The specific aim of this study was to examine whether NO-Cbl could sensitize drug-resistant melanomas to Apo2L/TRAIL. Antiproliferative effects of NO-Cbl and Apo2L/TRAIL were assessed in malignant melanomas and non-tumorigenic melanocyte and fibroblast cell lines. Athymic nude mice bearing human melanoma A375 xenografts were treated with NO-Cbl and Apo2L/TRAIL. Apoptosis was measured by TUNEL and confirmed by examining levels and activity of key mediators of apoptosis. The activation status of NF-kappa B was established by assaying DNA binding, luciferase reporter activity, the phosphorylation status of I kappa B alpha, and in vitro IKK activity. NO-Cbl sensitized Apo2L/TRAIL-resistant melanoma cell lines to growth inhibition by Apo2L/TRAIL but had minimal effect on normal cell lines. NO-Cbl and Apo2L/TRAIL exerted synergistic anti-tumor activity against A375 xenografts. Treatment with NO-Cbl followed by Apo2L/TRAIL induced apoptosis in Apo2L/TRAIL-resistant tumor cells, characterized by cleavage of caspase-3, caspase-8, and PARP. NO-Cbl inhibited IKK activation, characterized by decreased phosphorylation of I kappa B alpha and inhibition of NF-kappa B DNA binding activity. NO-Cbl suppressed Apo2L/TRAIL- and TNF-alpha-mediated activation of a transfected NF-kappa B-driven luciferase reporter. XIAP, an inhibitor of apoptosis, was inactivated by NO-Cbl. NO-Cbl treatment rendered Apo2L/TRAIL-resistant malignancies sensitive to the anti-tumor effects of Apo2L/TRAIL in vitro and in vivo. The use of NO-Cbl and Apo2L/TRAIL capitalizes on the tumor-specific properties of both agents and represents a promising anti-cancer combination.


Subject(s)
Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Membrane Glycoproteins/pharmacology , NF-kappa B/metabolism , Nitroso Compounds/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Vitamin B 12/analogs & derivatives , Vitamin B 12/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Base Sequence , Binding Sites/genetics , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , Humans , I-kappa B Kinase , Melanoma, Experimental/pathology , Mice , Mice, Nude , Neoplasm Transplantation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Signal Transduction/drug effects , TNF-Related Apoptosis-Inducing Ligand , Transplantation, Heterologous
6.
J Interferon Cytokine Res ; 23(1): 3-10, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12639293

ABSTRACT

Angiogenesis is an absolute requirement for tumor growth and metastasis. The purpose of this study was to evaluate the antiangiogenic activity of interferon-alpha2b (IFN-alpha2b) and thalidomide, as single agents and in combination. The murine dermis model was used to assess tumor-induced angiogenesis in nude mice. Human ACHN (renal), NIH-OVCAR-3 (ovarian), LNCaP (prostate), and SK-Mel-1 (melanoma) tumor cells were inoculated intradermally into the flanks of nude mice. IFN-alpha2b and thalidomide, administered daily, were effective inhibitors of angiogenesis induced by all four tumor types. The combination of IFN-alpha2b and thalidomide caused a synergistic decrease in mean vessel count in tumors that were resistant to the antiproliferative effects of IFN-alpha2b and thalidomide in vitro. This enhanced suppression of angiogenesis translated into synergistic antitumor activity in a xenograft model. Pegylated IFN-alpha (PEG-IFN-alpha2b) (10(6) U) administered once in 10 days was as effective as daily IFN-alpha2b treatment (10(6) U x 10 days). IFN-alpha2b and thalidomide have potentiated antiangiogenic activity when used in combination. A single dose of PEG-IFN-alpha2b (10(6) U) was as effective at suppressing vessel growth as an equivalent dose of IFN-alpha2b given daily for 10 days.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Interferon-alpha/administration & dosage , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/prevention & control , Polyethylene Glycols , Thalidomide/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Cell Division/drug effects , Drug Administration Schedule , Drug Synergism , Female , Humans , Interferon alpha-2 , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/pathology , Recombinant Proteins , Transplantation, Heterologous , Tumor Cells, Cultured
7.
J Natl Cancer Inst ; 94(13): 1010-9, 2002 Jul 03.
Article in English | MEDLINE | ID: mdl-12096086

ABSTRACT

BACKGROUND: The ubiquitous plasma membrane transcobalamin II receptor (TC II-R) mediates uptake of cobalamin (Cbl; vitamin B12), an essential micronutrient. Tumors often require more Cbl than normal tissue, and increased Cbl uptake may result from increased TC II-R expression. To examine whether Cbl could therefore be used as a carrier molecule to target a chemotherapy drug, we tested an analogue of Cbl with nitric oxide as a ligand, nitrosylcobalamin (NO-Cbl). Because interferon beta (IFN-beta) has antitumor effects and increases expression of some membrane receptors, we examined whether it may enhance the effects of NO-Cbl. METHODS: Antiproliferative effects of NO-Cbl were assessed in 24 normal and cancer cell lines. Xenograft tumors of human ovarian cancer NIH-OVCAR-3 cells were established in athymic nude mice, and tumor growth was monitored after treatment with NO-Cbl and IFN-beta, both individually and concomitantly. TC II-R expression and apoptosis was monitored in vitro and in vivo. RNA protection assays and mitochondrial membrane potential assays were used to distinguish the extrinsic and intrinsic apoptotic pathways, respectively. RESULTS: Cancer cell lines were more sensitive to NO-Cbl (with ID(50)s [the dose that inhibits growth by 50%] as low as 2 microM) than normal cell lines (with ID(50)s of 85-135 microM). Single-agent NO-Cbl and IFN-beta treatment of NIH-OVCAR-3 xenografts induced tumor regression, whereas combination treatment induced tumor eradication. IFN-beta treatment increased TC II-R expression in vitro and uptake of [(57)Co]cobalamin in vivo. Compared with NIH-OVCAR-3 cells treated with NO-Cbl, cells treated with NO-Cbl and IFN-beta were more apoptotic and expressed higher mRNA levels of various apoptosis-associated genes. No changes in mitochondrial membrane potential were observed in cells treated with NO-Cbl. CONCLUSION: NO-Cbl inhibited tumor growth in vivo by activating the extrinsic apoptotic pathway. The increased expression of TC II-R induced by IFN-beta resulted in enhanced antitumor effects with NO-Cbl both in vitro and in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/therapy , Interferon-beta/therapeutic use , Melanoma/therapy , Nitroso Compounds/pharmacology , Ovarian Neoplasms/therapy , Receptors, Cell Surface/metabolism , Vitamin B 12/analogs & derivatives , Vitamin B 12/pharmacology , Animals , Annexin A5/metabolism , Antineoplastic Agents/therapeutic use , Apoptosis/physiology , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Division/drug effects , Combined Modality Therapy , Drug Synergism , Female , Humans , Immunoenzyme Techniques , Male , Melanoma/metabolism , Melanoma/pathology , Membrane Potentials , Mice , Mice, Nude , Mitochondria/metabolism , Nitric Oxide/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Rhodamines , Ribonuclease, Pancreatic/metabolism , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
8.
Oncogene ; 21(12): 1882-9, 2002 Mar 14.
Article in English | MEDLINE | ID: mdl-11896621

ABSTRACT

We recently identified inositol hexakisphosphate kinase 2 (IP6K2) as a positive regulator of apoptosis. Overexpression of IP6K2 enhances apoptosis induced by interferon-beta (IFN-beta) and cytotoxic agents in NIH-OVCAR-3 ovarian carcinoma cells. In this study, we contrast and compare IFN-beta and radiation-induced death, and show that IP6K2 expression sensitizes tumor cells. Unirradiated NIH-OVCAR-3 cells transfected with IP6K2 formed fewer colonies compared to unirradiated vector-expressing cells. IP6K2 overexpression caused increased radiosensitivity, evidenced by decreased colony forming units (CFU). Both IFN-beta and radiation induced caspase 8. IFN-beta, but not gamma-irradiation, induced TRAIL in NIH-OVCAR-3 cells. Gamma irradiation, but not IFN-beta, induced DR4 mRNA. Apoptotic effects of IFN-beta or gamma-irradiation were blocked by expression of a dominant negative mutant death receptor 5 (DR5Delta) or by Bcl-2. Caspase-8 mRNA induction was more pronounced in IP6K2-expressing cells compared to vector-expressing cells. These data suggest that overexpression of IP6K2 enhances sensitivity of some ovarian carcinomas to radiation and IFN-beta. IP6K2 may function to enhance the expression and/or function of caspase 8 and DR4 following cell injury. Both IFN-beta and gamma-irradiation induce apoptosis through the extrinsic, receptor-mediated pathway, IFN-beta through TRAIL, radiation through DR4, and both through caspase 8. The function of both death inducers is positively regulated by IP6K2.


Subject(s)
Ovarian Neoplasms/enzymology , Phosphotransferases (Phosphate Group Acceptor)/physiology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/radiation effects , Caspase 8 , Caspase 9 , Caspases/genetics , Caspases/metabolism , Female , Gamma Rays , Gene Expression Regulation, Enzymologic , Humans , Interferon-beta/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/radiotherapy , Proto-Oncogene Proteins c-bcl-2/physiology , RNA, Messenger/metabolism , Radiation-Sensitizing Agents , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Tumor Necrosis Factor/physiology , Ribonucleases/genetics , Ribonucleases/metabolism , Transfection , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/radiation effects , Tumor Stem Cell Assay
SELECTION OF CITATIONS
SEARCH DETAIL
...