Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Int J Oncol ; 57(4): 1057-1071, 2020 10.
Article in English | MEDLINE | ID: mdl-32945449

ABSTRACT

Menin­mixed­lineage leukemia (MLL) inhibitors have potential for use as therapeutic agents for MLL­rearranged leukemia. They are also effective against solid cancers, such as breast cancer. The present study demonstrated that menin­MLL inhibitors, such as MI­463, unexpectedly induced the ferroptotic cell death of several cancer cell lines. MI­463 at a double­digit nM concentration markedly decreased the viable number of OVCAR­8 ovarian cancer cells for 3 days. Ferrostatin­1 (a ferroptosis inhibitor) almost completely abrogated the MI­463­induced decrease in viable cell numbers. Furthermore, the cancer cell­killing activity was inhibited by N­acetylcysteine [a scavenger of reactive oxygen species (ROS)], deferoxamine (DFO, an iron chelator), PD146176 (a specific inhibitor of arachidonate 15­lipoxygenase), idebenone (a membrane­permeable analog of CoQ10) and oleic acid [a monounsaturated fatty acid and one of the end products of stearoyl­CoA desaturase 1 (SCD1)], whereas Z­VAD­FMK (an apoptosis inhibitor) had a negligible effect on cell death. It was also found that MI­463 in combination with auranofin (a thioredoxin reductase inhibitor) synergistically increased cancer the death of breast, ovarian, pancreatic and lung cancer cell lines (88%, 14/16 cell lines). The synergistic induction of cell death was abrogated by ferroptosis inhibitor and DFO. Inhibitors of SCD1, similar to MI­463, also enhanced cancer cell death synergistically with auranofin, while inhibitors of SCD1 and MI­463 did not additively induce cell death. Treatment with zinc protoporphyrin­9, a specific inhibitor of heme oxygenase­1 (HO­1), markedly attenuated the cell death induced by MI­463 plus auranofin. On the whole, these results suggest that the MI­463­induced decrease in cell viability may be at least partly associated with the inhibition of SCD1 activity. In addition, the potent induction of HO­1 contributed to the synergistic effects of MI­463 plus auranofin. Therefore, menin­MLL inhibitors, such as MI­463, in combination with auranofin represent an effective therapeutic approach for several types of cancer via the induction of ferroptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Auranofin/pharmacology , Ferroptosis/drug effects , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Indoles/pharmacology , Myeloid-Lymphoid Leukemia Protein/antagonists & inhibitors , Neoplasms/drug therapy , Proto-Oncogene Proteins/antagonists & inhibitors , Antirheumatic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Humans , Neoplasms/metabolism , Neoplasms/pathology , Reactive Oxygen Species/metabolism
2.
Ann Clin Lab Sci ; 48(3): 314-322, 2018 May.
Article in English | MEDLINE | ID: mdl-29970434

ABSTRACT

OBJECTIVE: An aberrant production of inflammatory cytokines, with resultant febrile response, is a cardinal finding of hemophagocytic syndrome. A role of inflammatory cytokines on phagocytosis and clearance of apoptotic cells or particles has been shown, but effects of cytokines or hyperthermia on phagocytosis of viable blood cells were not fully understood. We examined effects of cytokines and hyperthermia on phagocytosis, and externalization of phosphatidylserine on the surface of phagocytosed blood cells, to clarify the pathophysiology of hemophagocytic syndrome. METHODS: THP-1 macrophage cells were incubated with non-opsonized and opsonized sheep erythrocytes (SE) in the presence of tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), macrophage-colony stimulating factor (M-CSF), interleukin (IL)-6, IL-10 or IL-18, and phagocytic activity was analyzed. Co-operative effect between cytokines was also examined. In addition, SE were incubated at 37 or 39°C, and phagocytic activity was analyzed. After treatment of SE with cytokine or hyperthermia, phosphatidylserine expression of the cell surface was analyzed by detecting Annexin V-positive cells. RESULTS: IL-6, IL-10 and IL-18 significantly increased phagocytosis of non-opsonized SE, but IFN-γ suppressed it. Phagocytosis of opsonized SE was significantly increased by any of the cytokines. IFN-γ suppressed and IL-10 enhanced phagocytic activity induced by other cytokines in non-opsonized SE, while in opsonized SE, both cytokines co-operated with other cytokines to enhance phagocytosis. Incubation of SE at a high temperature (39°C) resulted in increased phagocytic activity, as compared to SE incubated at 37°C. Cytokines and a high temperature did not increase the number of Annexin V-positive SE. CONCLUSIONS: IL-6, IL-10 and IL-18 can augment phagocytosis of viable blood cells, whether cells are opsonized or not. Hyperthermia also enhances phagocytosis. These in vitro data suggest that therapy for targeting cytokine (IL-6, IL-10 or IL-18) by using biologics or small molecule drugs may be beneficial for the treatment of hemophagocytic syndrome. Unlike the case of apoptotic cells, phagocytosis of viable blood cells seems to be mediated via phosphatidylserine-independent manner.


Subject(s)
Cytokines/pharmacology , Erythrocytes/pathology , Hyperthermia, Induced/adverse effects , Lymphohistiocytosis, Hemophagocytic/physiopathology , Phagocytosis/physiology , Phosphatidylserines/metabolism , Animals , Cells, Cultured , Erythrocytes/drug effects , Humans , Lymphohistiocytosis, Hemophagocytic/etiology , Macrophages/drug effects , Macrophages/pathology , Phagocytosis/drug effects , Sheep
3.
Oncol Lett ; 15(3): 3240-3244, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29435064

ABSTRACT

Chemotherapy and radiotherapy are the most common approaches in cancer therapy. They may kill cancer cells through the generation of high levels of reactive oxygen species (ROS), which leads to oxidative DNA damage. However, tumor resistance to ROS is a problem in cancer therapy. MTH1 sanitizes oxidized dNTP pools to prevent the incorporation of damaged bases during DNA replication. Although MTH1 is non-essential in normal cells, cancer cells require MTH1 activity to avoid the incorporation of oxidized dNTPs, which would result in DNA damage and cell death. By targeting a redox-adaptation mechanism, MTH1 inhibition represents a novel therapeutic strategy against cancer. However, recent reports have indicated that growth inhibition by MTH1 inhibitors may be due to off-target cytotoxic effects. TH588, one of the first-in-class MTH1 inhibitors, kills cancer cells by an off-target effect. However, a low concentration of TH588 may effectively inhibit MTH1 activity without inhibiting cell proliferation. Phenethyl isothiocyanate (PEITC) is a dietary anticarcinogenic compound and an inducer of ROS. In the present study, it has been demonstrated that combined treatment with PEITC and TH588 effectively inhibited the growth of pancreatic cancer MIAPaCa-2 and Panc-1 cells. The antioxidant N-acetylcysteine negated this synergistic growth inhibition. PEITC and TH588 cooperatively induced the formation of 8-oxo-deoxyguanine in nuclei and pH2AX foci, a marker of DNA damage. However, the combined effects are not associated with MTH1 mRNA expression in several cancer cell lines, suggesting that the possibility of an off-target effect of TH588 cannot be eliminated. These results suggest that the combination of PEITC and TH588 has potential as a novel therapeutic strategy against pancreatic cancer.

4.
Int J Oncol ; 52(3): 1011-1022, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29393418

ABSTRACT

Pancreatic cancer is one of the most lethal types of cancer with a mortality rate of almost 95%. Treatment with current chemotherapeutic drugs has limited success due to poor responses. Therefore, the development of novel drugs or effective combination therapies is urgently required. Piperlongumine (PL) is a natural product with cytotoxic properties restricted to cancer cells by significantly increasing intracellular reactive oxygen species (ROS) levels. In the present study, we demonstrated that PL induced cancer cell death through, at least in part, the induction of ferroptosis, as the cancer cell-killing activity was inhibited by the antioxidant, N­acetylcysteine, ferroptosis inhibitors (ferrostatin­1 and liproxstatin­1) and the iron chelator, deferoxamine (DFO), but not by the apoptosis inhibitor, Z-VAD-FMK, or the necrosis inhibitor, necrostatin­1. Cotylenin A (CN­A; a plant growth regulator) exhibits potent antitumor activities in several cancer cell lines, including pancreatic cancer cell lines. We found that CN­A and PL synergistically induced the death of pancreatic cancer MIAPaCa­2 and PANC­1 cells for 16 h. CN­A enhanced the induction of ROS by PL for 4 h. The synergistic induction of cell death was also abrogated by the ferroptosis inhibitors and DFO. The present results revealed that clinically approved sulfasalazine (SSZ), a ferroptosis inducer, enhanced the death of pancreatic cancer cells induced by PL and the combined effects were abrogated by the ferroptosis inhibitors and DFO. SSZ further enhanced the cancer cell-killing activities induced by combined treatment with PL plus CN­A. On the other hand, the synergistic induction of cell death by PL and CN­A was not observed in mouse embryonic fibroblasts (MEFs), and SSZ did not enhance the death of MEFs induced by PL plus CN­A. These results suggest that the triple combined treatment with PL, CN­A and SSZ is highly effective against pancreatic cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Death/drug effects , Dioxolanes/pharmacology , Iron/metabolism , Pancreatic Neoplasms/drug therapy , Acetylcysteine/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cyclohexylamines/pharmacology , Deferoxamine/pharmacology , Dioxolanes/therapeutic use , Diterpenes/pharmacology , Diterpenes/therapeutic use , Drug Synergism , Fibroblasts , Humans , Mice , Pancreatic Neoplasms/pathology , Phenylenediamines/pharmacology , Quinoxalines/pharmacology , Reactive Oxygen Species/metabolism , Spiro Compounds/pharmacology , Sulfasalazine/pharmacology , Sulfasalazine/therapeutic use
5.
Leuk Res ; 66: 39-48, 2018 03.
Article in English | MEDLINE | ID: mdl-29407582

ABSTRACT

The NM23 gene is overexpressed in many hematological malignancies and its overexpression predicts poor treatment outcomes. NM23 overexpression is thought to suppress myeloid differentiation of leukemia cells, but the molecular mechanism is unknown. In breast cancer cells, the lysophosphatidic acid (LPA) receptor EDG2/lpa1 was downregulated by NM23-H1 overexpression, and this reciprocal expression pattern was associated with suppressed or induced cell motility/metastasis. Here, we examined the relationship between EDG2 and NM23 expression during myeloid differentiation of leukemia cells. NM23 expression decreased and EDG2 expression increased during all-trans retinoic acid (ATRA)-induced myeloid differentiation of HL-60, NB4, and THP-1 leukemia cells. Moreover, this inverse correlation was more evident when myeloid differentiation was enhanced by ellagic acid, an inhibitor of NM23 activity. In contrast, there was no inverse correlation between EDG2 and NM23 expression during erythroid differentiation of HEL and K562 cells. ATRA plus LPA enhanced the motility of leukemia cells as well as breast cancer cells in an EDG2-dependent manner. These results suggest a common molecular mechanism between myeloid differentiation of leukemia cells and migration of breast cancer cells depending on NM23 and EDG2 expression levels.


Subject(s)
Cell Differentiation , Down-Regulation , Gene Expression Regulation, Leukemic , Leukemia/metabolism , Myeloid Cells/metabolism , NM23 Nucleoside Diphosphate Kinases/biosynthesis , Neoplasm Proteins/biosynthesis , Receptors, Lysophosphatidic Acid/biosynthesis , Up-Regulation , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , HL-60 Cells , Humans , K562 Cells , Leukemia/genetics , Leukemia/pathology , Lysophospholipids/pharmacology , MCF-7 Cells , Myeloid Cells/pathology , NM23 Nucleoside Diphosphate Kinases/genetics , Receptors, Lysophosphatidic Acid/genetics , Tretinoin/pharmacology , U937 Cells
6.
Oncol Rep ; 36(2): 968-76, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27375275

ABSTRACT

The treatment of pancreatic cancer, one of the most aggressive gastrointestinal tract malignancies, with current chemotherapeutic drugs has had limited success due to its chemoresistance and poor prognosis. Therefore, the development of new drugs or effective combination therapies is urgently needed. Cotylenin A (CN-A) (a plant growth regulator) is a potent inducer of differentiation in myeloid leukemia cells and exhibits potent antitumor activities in several cancer cell lines. In the present study, we demonstrated that CN-A and phenethyl isothiocyanate (PEITC), an inducer of reactive oxygen species (ROS) and a dietary anticarcinogenic compound, synergistically inhibited the proliferation of MIAPaCa-2, PANC-1 and gemcitabine-resistant PANC-1 cells. A combined treatment with CN-A and PEITC also effectively inhibited the anchorage-independent growth of these cancer cells. The combined treatment with CN-A and PEITC strongly induced cell death within 1 day at concentrations at which CN-A or PEITC alone did not affect cell viability. A combined treatment with synthetic CN-A derivatives (ISIR-005 and ISIR-042) or fusicoccin J (CN-A-related natural product) and PEITC did not have synergistic effects on cell death. The combined treatment with CN-A and PEITC synergistically induced the generation of ROS. Antioxidants (N-acetylcysteine and trolox), ferroptosis inhibitors (ferrostatin-1 and liproxstatin), and the lysosomal iron chelator deferoxamine canceled the synergistic cell death. Apoptosis inhibitors (Z-VAD-FMK and Q-VD-OPH) and the necrosis inhibitor necrostatin-1s did not inhibit synergistic cell death. Autophagy inhibitors (3-metyladenine and chloroquine) partially prevented cell death. These results show that synergistic cell death induced by the combined treatment with CN-A and PEITC is mainly due to the induction of ferroptosis. Therefore, the combination of CN-A and PEITC has potential as a novel therapeutic strategy against pancreatic cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Death/drug effects , Cyclohexylamines/metabolism , Diterpenes/pharmacology , Isothiocyanates/pharmacology , Pancreatic Neoplasms/drug therapy , Phenylenediamines/metabolism , Acetylcysteine/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Antioxidants/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Glycosides/pharmacology , Humans , Pancreatic Neoplasms/metabolism , Quinolines/pharmacology , Reactive Oxygen Species/metabolism
7.
Int J Oncol ; 47(2): 473-80, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26046133

ABSTRACT

Although all-trans retinoic acid (ATRA) is a standard and effective drug used for differentiation therapy in acute promyelocytic leukemia, ATRA-resistant leukemia cells ultimately emerge during this treatment. Therefore, the development of new drugs or effective combination therapy is urgently needed. We demonstrate that the combined treatment of vitamin K2 and cotylenin A synergistically induced monocytic differentiation in HL-60 cells. This combined treatment also synergistically induced NBT-reducing activity and non-specific esterase-positive cells as well as morphological changes to monocyte/macrophage-like cells. Vitamin K2 and cotylenin A cooperatively inhibited the proliferation of HL-60 cells in short-term and long-term cultures. This treatment also induced growth arrest at the G1 phase. Although 5 µg/ml cotylenin A or 5 µM vitamin K2 alone reduced c-MYC gene expression in HL-60 cells to approximately 45% or 80% that of control cells, respectively, the combined treatment almost completely suppressed c-MYC gene expression. We also demonstrated that the combined treatment of vitamin K2 and cotylenin A synergistically induced the expression of cyclin G2, which had a positive effect on the promotion and maintenance of cell cycle arrest. These results suggest that the combination of vitamin K2 and cotylenin A has therapeutic value in the treatment of acute myeloid leukemia.


Subject(s)
Cyclin G2/genetics , Diterpenes/pharmacology , Leukemia/genetics , Monocytes/drug effects , Proto-Oncogene Proteins c-myc/genetics , Vitamin K 2/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , HL-60 Cells , Humans , Leukemia/drug therapy , Monocytes/cytology
8.
Int J Oncol ; 46(2): 841-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25405645

ABSTRACT

Arsenic trioxide (ATO) is an approved treatment for acute promyelocytic leukemia (APL). It has also shown potential for treatment of multiple myeloma and various solid tumors including breast cancer. The requirement of high, toxic concentrations for the induction of apoptosis in non-APL and solid tumor cells is a major limitation for its use in other hematological malignancies and solid tumors. We have examined whether inducers of differentiation of leukemia cells can control the growth of solid tumor cells. In the present study, we found that cotylenin A, a plant growth regulator and a potent inducer of differentiation in myeloid leukemia cells, significantly potentiated both ATO-induced inhibition of cell growth in a liquid culture, and ATO-induced inhibition of anchorage-independent growth in a semi-solid culture in human breast cancer MCF-7 and MDA-MB-231 cells. ISIR-005 (a synthetic cotylenin A-derivative) was also able to enhance ATO-induced growth inhibition. The combined treatment with cotylenin A and ATO induced cleaved caspase-7 in MCF-7 cells at the concentrations which ATO alone scarcely induced and cotylenin A alone only weakly induced. Expression of survivin in MCF-7 cells was markedly decreased with the presence of both cotylenin A and ATO, although the expression of survivin was only slightly decreased by cotylenin A or ATO alone. The pretreatment with N-acetylcysteine significantly reduced the combination treatment-induced cell growth inhibition. These data suggest that induction of cleaved caspase-7, inhibition of survivin and oxidative responses are important events in the corporative inhibition in the growth of MCF-7 cells induced by both cotylenin A and ATO. Furthermore, we found that the combined treatment with cotylenin A and ATO also could be effective in suppressing the invasive capacity of MDA-MB-231 cells determined with the impedance-based xCELLigence Real-Time Cell Analysis technology. These results suggest that cotylenin A is an attractive enhancer for the ATO-induced anticancer activities in human breast cancer.


Subject(s)
Apoptosis/drug effects , Arsenicals/administration & dosage , Breast Neoplasms/drug therapy , Diterpenes/administration & dosage , Oxides/administration & dosage , Arsenic Trioxide , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology
9.
Int J Oncol ; 42(2): 767-75, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23255002

ABSTRACT

Cotylenin A, a plant growth regulator, and rapa-mycin, an inhibitor of the mammalian target of rapamycin, are potent inducers of differentiation in myeloid leukemia cells and also synergistically inhibit the proliferation of several human breast cancer cell lines including MCF-7 in vitro and in vivo. However, the mechanisms of the combined effects of cotylenin A and rapamycin are still unknown. Activated Akt induced by rapamycin has been suggested to attenuate the growth-inhibitory effects of rapamycin, serving as a negative feedback mechanism. In this study, we found that cotylenin A could suppress rapamycin-induced phosphorylation of Akt (Ser473) in MCF-7 cells and lung carcinoma A549 cells and that cotylenin A also enhanced the rapamycin-induced growth inhibition of MCF-7 and A549 cells. ISIR-005 (a synthetic cotylenin A-derivative) was able to enhance rapamycin­induced growth inhibition and could also markedly inhibit rapamycin-induced phosphorylation of Akt. We also found that the HSP90 inhibitor 17-allylamino-17-demethoxygeldanamycin (17-AAG) or arsenic trioxide (ATO) in combination with rapamycin markedly inhibited the growth of MCF-7 cells and 17-AAG or ATO suppressed rapamycin-induced phosphorylation of Akt. The PI3K inhibitor LY294002 also suppressed rapamycin-induced phosphorylation of Akt and combined treatment showed synergistic growth inhibition of MCF-7 cells. Rapamycin inhibited growth more significantly in Akt siRNA-transfected MCF-7 cells than in control siRNA-transfected MCF-7 cells. These results suggest that the inhibition of rapamycin-induced Akt phosphorylation by cotylenin A correlates with their effective growth inhibition of cancer cells.


Subject(s)
Breast Neoplasms/drug therapy , Diterpenes/administration & dosage , Lung Neoplasms/drug therapy , Sirolimus/administration & dosage , Benzoquinones/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Chromones/pharmacology , Drug Synergism , Female , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Lactams, Macrocyclic/pharmacology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MCF-7 Cells , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors
10.
Adv Hematol ; 2012: 879368, 2012.
Article in English | MEDLINE | ID: mdl-21941554

ABSTRACT

An elevated serum level of NM23-H1 protein is a poor prognostic factor in patients with various hematologic malignancies. The extracellular NM23-H1 protein promotes the in vitro growth and survival of acute myelogenous leukemia (AML) cells and inversely inhibits the in vitro survival of normal peripheral blood monocytes in primary culture at concentrations equivalent to the levels found in the serum of AML patients. The growth and survival promoting activity to AML cells is associated with cytokine production and activation of mitogen-activated protein kinases (MAPKs) and signal transducers and activators of transcription (STAT) signaling pathways. Inhibitors specific for MAPK signaling pathways inhibit the growth/survival-promoting activity of NM23-H1. These findings indicate a novel biological action of extracellular NM23-H1 and its association with poor prognosis. These results suggest an important role of extracellular NM23-H1 in the malignant progression of leukemia and a potential therapeutic target for these malignancies.

11.
Int J Hematol ; 92(1): 136-43, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20559758

ABSTRACT

All-trans retinoic acid (ATRA) is a standard drug used for differentiation therapy in acute promyelocytic leukemia. To potentiate this therapy, we examined the effect of ellagic acid (EA), a natural polyphenolic compound with antiproliferative and antioxidant properties, on the growth and differentiation of HL-60 acute myeloid leukemia cells. EA was found to induce apoptosis, which was blocked by pan-caspase inhibitor, Z-VAD-FMK. EA activated the caspase-3 pathway and enhanced the expressions of myeloid differentiation markers (CD11b, MRP-14 protein, granulocytic morphology) induced by ATRA treatment. These results indicate that EA is a potent apoptosis inducer and also effectively potentiates ATRA-induced myeloid differentiation of HL-60 cells.


Subject(s)
Apoptosis/drug effects , Cell Differentiation/drug effects , Ellagic Acid/pharmacology , Tretinoin/pharmacology , Antineoplastic Agents/pharmacology , Drug Synergism , Flavonoids , HL-60 Cells , Humans , Phenols , Polyphenols
12.
Int J Hematol ; 90(2): 143-152, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19655221

ABSTRACT

An elevated serum level of NM23-H1 protein is found in acute myelogenous leukemia (AML) and predicts a poor treatment outcome for AML patients. To investigate the potential pathological link between the elevated serum level of this protein and poor prognosis, we examined the extracellular effects of recombinant NM23-H1 protein on the in vitro survival of primary cultured normal peripheral blood mononuclear cells (PBMNC) at concentrations equivalent to the levels found in the serum of AML patients. Extracellular NM23-H1 protein inhibited the in vitro survival of PBMNC and promoted the production of various cytokines, such as GM-CSF and IL-1beta, which in fact promoted the growth of primary cultured AML cells. These findings indicate a novel biological action of extracellular NM23-H1 and its association with poor prognosis of patients with elevated serum levels of NM23-H1 protein. These results suggest an important role of extracellular NM23-H1 in the malignant progression of leukemia and a potential therapeutic target for these malignancies.


Subject(s)
Cell Survival/immunology , Leukemia, Monocytic, Acute/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukocytes, Mononuclear/cytology , NM23 Nucleoside Diphosphate Kinases/blood , Autoantibodies/blood , Cell Survival/drug effects , Cytokines/blood , Cytokines/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/blood , Humans , In Vitro Techniques , Leukemia, Monocytic, Acute/immunology , Leukemia, Monocytic, Acute/pathology , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/pathology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , NM23 Nucleoside Diphosphate Kinases/genetics , NM23 Nucleoside Diphosphate Kinases/pharmacology , Prognosis , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Tumor Cells, Cultured
13.
Cancer Sci ; 100(10): 1885-94, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19664043

ABSTRACT

An elevated serum level of NM23-H1 protein is found in acute myelogenous leukemia (AML), and predicts a poor treatment outcome in AML patients. To investigate the potential pathological link between the elevated serum level of this protein and poor prognosis, we examined the extracellular effects of recombinant NM23-H1 protein on the in vitro growth and survival of primary cultured AML cells at concentrations equivalent to the levels found in the serum of AML patients. Extracellular NM23-H1 protein promoted the in vitro growth and survival of AML cells and this activity was associated with the cytokine production and activation of the MAPK and signal transducers and activators of transcription signaling pathways. Inhibitors specific to MAPK signaling pathways inhibited the growth- and survival-promoting activity of NM23-H1. These findings indicate the novel biological action of extracellular NM23-H1 and its association with poor prognosis, and suggest an important role for extracellular NM23-H1 in the malignant progression of leukemia and a potential therapeutic target for these malignancies.


Subject(s)
Cell Proliferation , Extracellular Fluid/chemistry , Leukemia, Myeloid, Acute/metabolism , NM23 Nucleoside Diphosphate Kinases/metabolism , Signal Transduction/physiology , Biomarkers, Tumor/analysis , Blotting, Western , Cell Line, Tumor , Cell Survival/physiology , Cytokines/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , NM23 Nucleoside Diphosphate Kinases/genetics , Prognosis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
14.
Cancer Sci ; 99(8): 1693-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18754885

ABSTRACT

Cotylenin A, a plant growth regulator, and rapamycin, an inhibitor of mammalian target of rapamycin (mTOR), are potent inducers of differentiation of myeloid leukemia cells. Recently, we found that cotylenin A and rapamycin effectively inhibited the proliferation of several human breast cancer cell lines including MCF-7. Herein, we demonstrate that cotylenin A and rapamycin rapidly and markedly induced the cyclin G2 gene expression in several cancer cells including MCF-7 cells. The growth arrest of the MCF-7 cells at the G1 phase, induced by the treatment with cotylenin A and rapamycin or the culture in low serum medium, markedly induced the cyclin G2 gene expression. Anticancer drugs including doxorubicin, etoposide and 5-fluorouracil also induced cyclin G2 expression during induction of growth arrest of the MCF-7 cell at the G1 phase or G2/M phase. Ectopically inducible cyclin G2 expression potently inhibited the proliferation of MCF-7 cells. Furthermore, cyclin G2 knockdown induced by cyclin G2 small interfering RNA markedly reduced the potency of cotylenin A plus rapamycin to induce growth inhibition. Taken together, our results suggest that cotylenin A and rapamycin induce inhibition of cancer cell growth through the induction of cyclin G2.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Cyclins/biosynthesis , Diterpenes/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Sirolimus/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Cyclin G2 , Gene Expression/drug effects , Humans
15.
Leuk Lymphoma ; 47(4): 733-40, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16690533

ABSTRACT

Cotylenin A, which has been isolated as a plant growth regulator, potently induces the differentiation of human myeloid leukemia cells. Treatment of HL-60 cells with a combination of transforming growth factor (TGF)-beta and 1alpha, 25-dihydroxyvitamin D(3) (VD3) resulted in increased differentiation compared to separate treatments, but TGF-beta did not affect the cotylenin A-induced differentiation of HL-60 cells. It is possible that the signal transduction pathway used by cotylenin A for inducing the differentiation of leukemia cells is the same as that used by TGF-beta. However, cotylenin A did not affect the expression of TGF superfamily or Smad genes in HL-60 cells. Treatment with neutralizing anti-TGF-beta antibody or an inhibitor of TGF-beta signaling did not inhibit cotylenin A-induced differentiation, although VD3-induced differentiation was significantly suppressed by these treatments. The subcellular distribution of Smad3 was also unaffected by cotylenin A. These results suggest that the cotylenin A-induced differentiation of leukemia cells is independent of the TGF-beta signaling system, although TGF-beta acts as an autocrine mediator of the growth arrest and differentiation of leukemia cells induced by VD3 and other inducers.


Subject(s)
Diterpenes/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Transforming Growth Factor beta/metabolism , Cell Differentiation , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , HL-60 Cells , Humans , Immunohistochemistry , Models, Biological , RNA, Messenger/metabolism , Signal Transduction , Smad3 Protein/metabolism , U937 Cells
16.
Cancer Sci ; 96(10): 653-60, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16232196

ABSTRACT

We have previously reported that NM23 genes are overexpressed in various hematological malignancies and that serum NM23-H1 protein levels are useful for predicting patient outcomes. In this study we assessed the clinical implications of serum NM23-H1 protein on neuroblastoma. We examined serum NM23-H1 protein levels in 217 patients with neuroblastoma, including 131 found by mass-screening and 86 found clinically by an enzyme-linked immunosorbent assay, and determined the association between levels of this protein, and known prognostic factors or the clinical outcome. The serum NM23-H1 protein level was higher in neuroblastoma patients than in control children (P < 0.0001). Patients with MYCN amplification had higher serum NM23-H1 levels than those with a single copy of MYCN. Overall survival was assessed in the 86 patients found clinically, and was found to be worse in patients with higher serum MN23-H1 levels (> or = 250 ng/mL) than in those with lower levels (< 250 ng/mL; P = 0.034). The higher level of NM23-H1 was correlated with a worse outcome in patients with a single MYCN copy, or in those younger than 12 months of age. Serum NM23-H1 protein levels may contribute to predictions of clinical outcome in patients with neuroblastoma.


Subject(s)
Biomarkers, Tumor/blood , Gene Dosage , Neuroblastoma/genetics , Neuroblastoma/pathology , Nucleoside-Diphosphate Kinase/blood , Age Factors , Case-Control Studies , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Female , Humans , Infant , Infant, Newborn , Male , N-Myc Proto-Oncogene Protein , NM23 Nucleoside Diphosphate Kinases , Neuroblastoma/therapy , Nuclear Proteins/analysis , Oncogene Proteins/analysis , Prognosis
17.
Biochim Biophys Acta ; 1745(2): 156-65, 2005 Sep 10.
Article in English | MEDLINE | ID: mdl-16129123

ABSTRACT

Cytokinins are important purine derivatives that act as redifferentiation-inducing hormones to control many processes in plants. Cytokinins such as isopentenyladenine (IPA) and kinetin are very effective at inducing the granulocytic differentiation of human myeloid leukemia HL-60 cells. We examined the gene expression profiles associated with exposure to IPA using cDNA microarrays and compared the results with those obtained with other inducers of differentiation, such as all-trans retinoic acid (ATRA), 1 alpha,25-dihydroxyvitamin D3 (VD3) and cotylenin A (CN-A). Many genes were up-regulated, and only a small fraction were down-regulated, upon exposure to the inducers. IPA and CN-A, but not ATRA or VD3, immediately induced the expression of mRNA for the calcium-binding protein S100P. The up-regulation of S100P was confirmed at the protein expression level. We also examined the expression of other S100 proteins, including S100A8, S100A9 and S100A12, and found that IPA preferentially up-regulated S100P at the early stages of differentiation. IPA-induced differentiation of HL-60 cells was suppressed by treatment with antisense oligonucleotides against S100P, suggesting that S100P plays an important role in cell differentiation.


Subject(s)
Calcium-Binding Proteins/biosynthesis , Cell Differentiation/physiology , Cytokinins/pharmacology , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Neoplasm Proteins/biosynthesis , Up-Regulation , Adenine/analogs & derivatives , Adenine/metabolism , Calcitriol/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/physiology , Cell Differentiation/drug effects , Cell Line, Tumor , DNA, Complementary , Diterpenes/metabolism , Gene Expression Regulation, Neoplastic/physiology , HL-60 Cells , Humans , Isopentenyladenosine , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Oligonucleotide Array Sequence Analysis , Tretinoin/metabolism , U937 Cells
18.
Gynecol Oncol ; 99(3): 680-8, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16112179

ABSTRACT

OBJECTIVE: Recently, we found that cotylenin A and IFNalpha synergistically inhibited growth both in vitro and in vivo, and induced apoptosis in human cancer cells. For the clinical application of this combined treatment, suitable cancer targets should be selected. METHODS: We examined the combined effects of these compounds on various types of cancer cells by a human cancer cell line panel assay, and on cancer cells that had been freshly isolated from patients in three-dimensional cultures embedded in collagen gel. RESULTS: In the analysis of 39 cancer cell lines, ovarian cancer cells were highly sensitive to combined treatment with cotylenin A and IFNalpha in inhibiting cell growth. This treatment was also effective toward ovarian cancer cells that were refractory to CDDP, and significantly inhibited the growth of ovarian cancer cells as xenografts without apparent adverse effects. Ovarian cancer cells from the patients were also sensitive to the combined treatment in primary cultures. CONCLUSION: Combined treatment with cotylenin A and IFNalpha may have therapeutic value in treating ovarian cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Diterpenes/pharmacology , Interferon-alpha/pharmacology , Ovarian Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Collagen , Diterpenes/administration & dosage , Drug Screening Assays, Antitumor , Drug Synergism , Female , Humans , Interferon-alpha/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
Br J Haematol ; 128(4): 540-7, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15686465

ABSTRACT

Cytokinins, purine derivatives that act as hormones to control many processes in plants, are very effective at inducing the granulocytic differentiation of human myeloid leukaemia cells. Isopentenyladenine (IPA), a potent cytokinin, significantly induced the expression of CCAAT/enhancer-binding protein (C/EBP)delta, but not C/EBP alpha protein, whereas all-trans retinoic acid, a well-known inducer of granulocytic differentiation, induced C/EBP alpha but not C/EBP delta protein. Antisense oligonucleotide for C/EBP delta, but not C/EBP alpha or C/EBP beta, effectively suppressed IPA-induced differentiation, suggesting that the expression of C/EBP delta protein is necessary for cytokinin-induced differentiation. Although C/EBP alpha is known to be crucial for granulocytic differentiation, the function of C/EBP delta has not been well documented in the regulation of haematopoiesis. The role of C/EBP delta in the granulocytic differentiation of myeloid leukaemia cells is discussed.


Subject(s)
Adenine/analogs & derivatives , CCAAT-Enhancer-Binding Proteins/metabolism , Cytokinins/pharmacology , Granulocytes/metabolism , Leukemia, Promyelocytic, Acute/metabolism , Transcription Factors/metabolism , Adenine/pharmacology , CCAAT-Enhancer-Binding Protein-delta , CCAAT-Enhancer-Binding Proteins/physiology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Granulocytes/pathology , HL-60 Cells , Humans , Isopentenyladenosine , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Oligonucleotides, Antisense/pharmacology , Transcription Factors/drug effects , Transcription Factors/physiology , Tretinoin/pharmacology , Tumor Cells, Cultured
20.
Breast Cancer Res ; 7(6): R1097-110, 2005.
Article in English | MEDLINE | ID: mdl-16457690

ABSTRACT

INTRODUCTION: Rapamycin, an inhibitor of the serine/threonine kinase target of rapamycin, induces G1 arrest and/or apoptosis. Although rapamycin and its analogues are attractive candidates for cancer therapy, their sensitivities with respect to growth inhibition differ markedly among various cancer cells. Using human breast carcinoma cell line MCF-7 as an experimental model system, we examined the growth-inhibitory effects of combinations of various agents and rapamycin to find the agent that most potently enhances the growth-inhibitory effect of rapamycin. METHOD: We evaluated the growth-inhibitory effect of rapamycin plus various agents, including cotylenin A (a novel inducer of differentiation of myeloid leukaemia cells) to MCF-7 cells, using either MTT assay or trypan blue dye exclusion test. The cell cycle was analyzed using propidium iodide-stained nuclei. Expressions of several genes in MCF-7 cells with rapamycin plus cotylenin A were studied using cDNA microarray analysis and RT-PCR. The in vitro results of MCF-7 cells treated with rapamycin plus cotylenin A were further confirmed in vivo in a mouse xenograft model. RESULTS: We found that the sensitivity of rapamycin to MCF-7 cells was markedly affected by cotylenin A. This treatment induced growth arrest of the cells at the G1 phase, rather than apoptosis, and induced senescence-associated beta-galactosidase activity. We examined the gene expression profiles associated with exposure to rapamycin and cotylenin A using cDNA microarrays. We found that expressions of cyclin G2, transforming growth factor-beta-induced 68 kDa protein, BCL2-interacting killer, and growth factor receptor-bound 7 were markedly induced in MCF-7 cells treated with rapamycin plus cotylenin A. Furthermore, combined treatment with rapamycin and cotylenin A significantly inhibited the growth of MCF-7 cells as xenografts, without apparent adverse effects. CONCLUSION: Rapamycin and cotylenin A cooperatively induced growth arrest in breast carcinoma MCF-7 cells in vitro, and treatment with rapamycin and cotylenin A combined more strongly inhibited the growth of MCF-7 cells as xenografts in vivo than treatment with rapamycin or cotylenin A alone, suggesting that this combination may have therapeutic value in treating breast cancer. We also identified several genes that were markedly modulated in MCF-7 cells treated with rapamycin plus cotylenin A.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/pathology , Carcinoma/pathology , Diterpenes/pharmacology , Sirolimus/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Interactions , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, Neoplasm , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...