Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Anticancer Res ; 39(7): 3579-3584, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31262882

ABSTRACT

BACKGROUND/AIM: Neuroblastoma (NB) is the most common extracranial solid tumor in childhood; treatments with greater effectiveness are required for NB, especially in advanced cases. This study aimed at evaluating the combined effect of anaplastic lymphoma kinase (ALK) inhibitor alectinib and histone deacetylase inhibitor vorinostat on NB cell lines harboring wild-type or mutated ALK. MATERIALS AND METHODS: Cytotoxicity was examined using the 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyltetrazolium bromide assay. Protein expression was analyzed using western blotting. RESULTS: Combination treatment with alectinib and vorinostat had a synergistic effect on growth inhibition of the NB cell line with ALK R1275Q mutation. Cleavage of caspase-3 and poly-(ADP-ribose) polymerase increased, indicating enhanced caspase-dependent apoptosis. In addition, this combination reduced the protein levels of MYCN proto-oncogene and nuclear factor kappa B, both of which are important for NB tumorigenesis and progression. CONCLUSION: Combined treatment with alectinib and vorinostat might be a novel therapeutic option for NB harboring the ALK R1275Q mutation.


Subject(s)
Anaplastic Lymphoma Kinase/antagonists & inhibitors , Carbazoles/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Neuroblastoma/drug therapy , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Vorinostat/pharmacology , Anaplastic Lymphoma Kinase/genetics , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Humans , Mutation , N-Myc Proto-Oncogene Protein/metabolism , NF-kappa B/metabolism , Neuroblastoma/genetics , Neuroblastoma/metabolism , Proto-Oncogene Mas
2.
Anticancer Res ; 35(12): 6679-84, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26637884

ABSTRACT

BACKGROUND: Bendamustine is effective in B-cell malignancies, including mantle cell lymphoma (MCL), alone and in combination with other agents. This study investigated the combination effect of bendamustine and the Bruton tyrosine kinase (BTK) inhibitor PCI-32765 on MCL cell death and the underlying mechanisms. MATERIALS AND METHODS: Cytotoxicity was examined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MIT) assay. Apoptosis was assessed by annexin V/propidium iodide staining and protein expression was analyzed by western blotting. RESULTS: When combined with bendamustine, PCI-32765 showed a synergistic effect on growth inhibition of the MCL cell line Jeko-1. Cleavage of caspase-3 and poly-(ADP-ribose) polymerase was increased, indicating enhanced apoptosis induction. In addition, this combination decreased the protein expression of cyclin D1. Phosphorylated v-akt murine thymoma viral oncogene homolog 1 (AKT) (Ser473) was also down-regulated, suggesting a suppression of the phosphatidylinositol 3-kinase/AKT signaling pathway. CONCLUSION: Combination treatment with bendamustine and a BTK inhibitor may be effective in MCL therapy.


Subject(s)
Bendamustine Hydrochloride/therapeutic use , Lymphoma, Mantle-Cell/drug therapy , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Adenine/analogs & derivatives , Bendamustine Hydrochloride/administration & dosage , Bendamustine Hydrochloride/pharmacology , Cell Proliferation , Humans , Lymphoma, Mantle-Cell/pathology , Piperidines , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Signal Transduction
3.
Apoptosis ; 20(7): 975-85, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25835755

ABSTRACT

Mantle cell lymphoma (MCL) is a B cell malignancy characterized by aberrant expression of cyclin D1 due to a t(11;14) translocation. MCL is refractory to conventional chemotherapy, and treatment remains challenging. We investigated the efficacy of the histone deacetylase (HDAC) inhibitor vorinostat combined with one of several B-cell receptor (BCR) signaling inhibitors on MCL cell death and the underlying mechanisms, using MCL cell lines. The Bruton's tyrosine kinase inhibitor PCI-32765 and the spleen tyrosine kinase inhibitor R406 showed synergistic effects with vorinostat on growth inhibition. Treatment with PCI-32765 or R406 alone induced 27.3 ± 2.1 or 25.1 ± 3.2% apoptosis. When combined with vorinostat, these apoptotic fractions significantly increased to 50.8 ± 4.9 and 63.1 ± 5.0%, respectively. Activation of caspase-3 and poly-(ADP-ribose) polymerase cleavage were markedly increased. We performed gene expression profiling following treatment with the combination of vorinostat and individual BCR signaling inhibitors using a microarray, and differentially expressed genes were identified. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis showed that the nuclear factor (NF)-κB signaling pathway was significantly enriched following treatment with the combination of vorinostat and R406. Protein expression analysis confirmed the down-regulation of NF-κB1/p105 and cyclin D1, suggesting inhibition of the NF-κB pathway. Taken together, the combination of an HDAC inhibitor and a BCR signaling inhibitor may be a novel therapeutic strategy for MCL.


Subject(s)
Apoptosis/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Lymphoma, Mantle-Cell/pathology , Protein Kinase Inhibitors/pharmacology , Receptors, Antigen, B-Cell/metabolism , Adenine/analogs & derivatives , Caspases/metabolism , Cell Line, Tumor , Cyclin D1/metabolism , Drug Synergism , Humans , NF-kappa B/metabolism , Oxazines/pharmacology , Piperidines , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Receptors, Antigen, B-Cell/antagonists & inhibitors , Vorinostat
4.
Nagoya J Med Sci ; 74(3-4): 261-71, 2012 Aug.
Article in English | MEDLINE | ID: mdl-23092099

ABSTRACT

Cytokine-dependent cell lines have been used to analyze the cytokine-induced cellular signaling and the mechanism of oncogenesis. In the current study, we analyzed MOTN-1 and PLT-2 cell lines established from different stages of a T-cell large granular lymphocyte leukemia patient (Daibata et al. 2004). MOTN-1 is IL-2-dependent derived from the chronic phase, whereas IL-2-independent PLT-2 is from the aggressive and terminal stage. They shared considerable chromosome abnormalities and the pattern of T-cell receptor rearrangement, presuming that the cytokine independence of PLT-2 was due to the additive genetic abnormality. Besides IL-2, IL-15 supported MOTN-1 cell growth, because these receptors share beta- and gamma-subunits. IL-2 activated ERK, AKT and STAT pathway of MOTN-1. STAT3 pathway of PLT-2 was also activated by IL-2, suggesting intact IL-2 induces signal transduction of PLT-2. However, ERK1/2 but not AKT, was continuously activated in PLT-2, consistent with the increased Ras-activity of PLT-2. Sequence analysis revealed KRAS G12A mutation but not NRAS and HRAS mutation of PLT-2 but not MOTN-1. Another signaling molecule affecting Ras-signaling pathway, SHP2, which has been frequently mutated in juvenile myelomonocytic leukemia (JMML), did not show mutation. Moreover, MEK inhibitor, PD98059, as well as farnesylation inhibitor inhibited PLT-2 cell growth. Using NIH3T3 and MOTN-1, ERK activation, increased cell proliferation and survival by KRAS G12A were shown, suggesting the important role of KRAS G12A in IL-2-independent growth of PLT-2. Taken together, KRAS G12A is important for IL-2-independent growth of PLT-2 cells and suggests the possibility of involvement of KRAS mutation with disease progression.


Subject(s)
Interleukin-2/pharmacology , Leukemia, Large Granular Lymphocytic/metabolism , Proto-Oncogene Proteins/genetics , ras Proteins/genetics , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Flavonoids/pharmacology , Humans , Interleukin-15/pharmacology , Leukemia, Large Granular Lymphocytic/genetics , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
5.
Genes Chromosomes Cancer ; 51(11): 1024-36, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22833470

ABSTRACT

Prospero-related homeobox 1 (PROX1) is important for embryonic organ formation and differentiation, and changes in PROX1 activity were recently associated with cancer. To address the PROX1 roles in tumorigenesis, we established cells stably overexpressing PROX1 using the human cervical cancer cell line, HeLa. Overexpression of PROX1 reduced cell proliferation and the rate of tumor formation as compared with controls. Comparison of gene expression profiles between PROX1-overexpressing and mock-transfected cells revealed that the expression of protein kinase C ßII (PRKCB2) is down-regulated in PROX1-overexpressing cells. A PRKCB inhibitor suppressed cell growth of control cells more than PROX1-expressing cells. Analysis of the 5'-promoter of PRKCB revealed that a region between -110 bp and the first exon contains two Sp1 binding sites and is important for transcriptional regulation of PRKCB. The inhibition of Sp1 transcription factor resulted in down-regulation of PRKCB2 protein levels. Treatment with a demethylating agent, 5-aza-2'-deoxycytidine, restored PRKCB2 mRNA expression in PROX1-expressing cells, suggesting that the 5'-promoter of PRKCB is methylated in these cells. Actually, it was found that a CpG island in this region, in particular a CpG site overlapping with the distal Sp1 site, was hypermethylated and direct Sp1 binding to this region was inhibited in PROX1-overexpressing cells. Thus, the suppressive effect of PROX1 on cell growth and tumor formation might be partially mediated by PRKCB2 via altered methylation of its promoter.


Subject(s)
DNA Methylation , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Protein Kinase C/genetics , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Proliferation/drug effects , Decitabine , Down-Regulation/drug effects , Gene Expression Profiling , HCT116 Cells , HeLa Cells , Homeodomain Proteins/metabolism , Humans , Indoles/pharmacology , Male , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase C beta , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/metabolism , Tumor Suppressor Proteins/metabolism
6.
J Biochem ; 151(6): 599-610, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22496486

ABSTRACT

Effects of all-trans retinoic acid (ATRA) on sphingomyelinase expression were examined using MCF-7 (ATRA-sensitive) and MDA-MB-231 (ATRA-resistant) breast cancer cells. Increased NSMase activity, NSMase2 mRNA and protein were observed in ATRA-treated MCF-7 but not in ATRA-treated MDA-MB-231. Increased NSMase2 mRNA of ATRA-treated MCF-7 was mostly due to enhanced transcription. Promoter analysis revealed the important 5'-promoter region of NSMase2 between -148 and -42 bp containing three Sp1 sites but no retinoic acid responsive elements. Experiments using mutated Sp1 sites of the NSMase2 promoter, Mithramycin A (a Sp inhibitor) and Sp family over-expression demonstrated the importance of Sp family protein and the three Sp1 sites for ATRA-induced NSMase2 transcription of MCF-7 cells. Although no quantitative change of bound Sp1 on NSMase2 promoter region after ATRA treatment was detected, Sp1 phosphorylation (activation) by ATRA was observed. Interestingly, PKCδ was involved in ATRA-induced increased NSMase2 transcription. ATRA-induced PKCδ phosphorylation and then activated PKCδ phosphorylated Sp1. Chromatin immunoprecipitation (ChIP) assay showed Sp1, RARα and RXRα complex formation in MCF-7 cells regardless of ATRA treatment and ATRA-induced acetylated histone H3 of the 5'-promoter. Thus, NSMase2 mRNA expression enhanced by ATRA was due to increased transcription via phosphorylated Sp1 caused by PKCδ activation, followed by chromatin remodelling with histone H3 acetylation.


Subject(s)
Sphingomyelin Phosphodiesterase/genetics , Transcription, Genetic , Tretinoin/pharmacology , Cell Survival/drug effects , Gene Expression Profiling , Humans , MCF-7 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Structure-Activity Relationship , Transcription, Genetic/drug effects , Tretinoin/therapeutic use
7.
J Biochem ; 151(6): 611-20, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22451680

ABSTRACT

Neutral ceramidase (NCDase) is considered to be a critical enzyme for controlling the turnover of ceramide, an important bioactive lipid, which determines cell's fate. All-trans retinoic acid (ATRA) has been reported to induce neuronal differentiation and cell-cycle arrest [Lopez-Carballo, Moreno, Masia, Perez, and Barettino (Activation of the phosphatidylinositol 3-kinase/Akt signalling pathway by retinoic acid is required for neural differentiation of SH-SY5Y human neuroblastoma cells. J Biol Chem 2002:277:25297-304.)]. In this study, we observed that ATRA-induced cellular ceramide accumulation, cell-growth arrest and differentiation accompanied with down-regulation of NCDase in SH-SY5Y cells, without a decrease in sphingosine or sphingosine 1-phosphate. We examined whether the down-regulation of NCDase was involved in the increase in ceramide and cell differentiation. ATRA was found to down-regulate mRNA, protein and the enzyme activity of NCDase. Interestingly, GATA-2 was also decreased with ATRA treatment, and experiments using its expression vector and siRNA and chromatin immunoprecipitation assay demonstrated GATA-2 acted as transcription-factor of NCDase gene expression. By establishing stable transfectants with decreased NCDase expression and activity, we clarified the significance of NCDase down-regulation for ATRA-induced neuronal differentiation. Those sub-clones showed both increased cellular ceramide and reduced cell growth as well as neuronal differentiation phenotypes. These results demonstrate that down-regulation of NCDase through ATRA-induced GATA-2 decrease plays an important role in induction of ceramide accumulation and neuronal differentiation in SH-SY5Y cells.


Subject(s)
Cell Differentiation/drug effects , Down-Regulation , Neuroblastoma/pathology , Neurons/drug effects , Neutral Ceramidase/metabolism , Tretinoin/pharmacology , Cell Proliferation/drug effects , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , Humans , Neurons/pathology , Neutral Ceramidase/biosynthesis , Neutral Ceramidase/genetics , Structure-Activity Relationship
8.
J Cell Biochem ; 112(11): 3449-58, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21769916

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) is important for the development and maintenance of dopamine neurons (Lin et al. [1993] Science 260: 1130-1132). GDNF is neuroprotective in animal models of Parkinson disease, where dopamine neurons show selective degeneration. We previously reported GDNF-induced SPHK1 gene expression in a neuroblastoma cell line, TGW (Murakami et al. [2007] J Neurochem 102: 1585-1594). In the present study, we focused on the regulatory mechanism of GAP43 (GDNF-induced neuronal phenotype) transcription to further elucidate physiological roles of GDNF-induced SPHK1 expression and activity. Stable wild-type (SPHK1-WT) but not dominant-negative SPHK1 (SPHK1-DN) overexpression increased both control- and GDNF-induced GAP43 expression. SPHK1-WT cells showed enhanced GDNF-induced sphingosine 1-phosphate (S1P) secretion compared with mock- and SPHK1-DN cells. Exogenous S1P also increased GAP43 expression. In TGW cells, PD98059, a MEK inhibitor, but not SB203580 (a p38 MAPK inhibitor) and LY294002 (a PI3K inhibitor) inhibited GDNF-induced GAP43 expression, suggesting the MEK/ERK pathway has a major role in GDNF-induced GAP43 transcription. A G-protein-coupled receptor inhibitor, pertussis toxin, and S1P(1) and S1P(3) receptor antagonists (VPC23019 and CAY10444) also inhibited ERK activation. Moreover, both S1P1 and S1P3 were serine-phosphorylated by GDNF, suggesting their activated states. C/EBPß transcription factor was induced by GDNF, and DNA pull-down and chromatin immunoprecipitation assays revealed the C/EBP binding site between -131 bp and -98 bp from the first exon of GAP43. Taken together, our results showed that in TGW cells, GDNF increased SPHK1 transcription, leading to the production and secretion of S1P. Through MEK/ERK pathway, S1P stimulates GAP43 transcription with increased binding of C/EBPß to the 5'-promoter.


Subject(s)
GAP-43 Protein/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Lysophospholipids/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Sphingosine/analogs & derivatives , Transcription, Genetic , Base Sequence , Blotting, Western , Cell Line, Tumor , Chromatin Immunoprecipitation , Cloning, Molecular , DNA Primers , Humans , Polymerase Chain Reaction , Promoter Regions, Genetic , Signal Transduction , Sphingosine/metabolism
9.
Biochim Biophys Acta ; 1811(3): 119-28, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21184844

ABSTRACT

The role of sphingolipid metabolic pathway has been recognized in determining cellular fate. Although sphingolipid degradation has been extensively studied, gene expression of human sphingosine 1-phosphate lyase (SPL) catalyzing sphingosine 1-phosphate (S1P) remains to be determined. Among 5 human lung cancer cell lines examined, SPL protein levels paralleled the respective mRNA and enzyme activities. Between H1155 and H1299 cells used for further experiments, higher cellular S1P was observed in H1155 with higher SPL activity compared with H1299 with low SPL activity. GATA-4 has been reported to affect SPL transcription in Dictyostelium discoideum. GATA-4 was observed in H1155 but not in other cell lines. Overexpression of GATA-4 in H1299 increased SPL expression. However, promoter analysis of human SPL revealed that the most important region was located between -136bp and -88bp from the first exon, where 2 Sp1 sites exist but no GATA site. DNA pull-down assay of H1155 showed increased DNA binding of Sp1 and GATA-4 within this promoter region compared with H1299. Electrophoresis mobility shift assay (EMSA), chromatin immunoprecipitation (ChIP) assay, reporter assay using mutated binding motif, and mithramycin A, a specific Sp1 inhibitor, suggest the major role of Sp1 in SPL transcription and no direct binding of GATA-4 with this 5' promoter region. The collaborative role of GATA-4 was proved by showing coimmunoprecipitation of Sp1 and GATA-4 using GST-Sp1 and overexpressed GATA-4. Thus, high SPL transcription of H1155 cells was regulated by Sp1 and GATA-4/Sp1 complex formation, both of which bind to Sp1 sites of the 5'-SPL promoter.


Subject(s)
Aldehyde-Lyases/biosynthesis , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Lung Neoplasms/enzymology , Neoplasm Proteins/biosynthesis , Transcription, Genetic , Aldehyde-Lyases/genetics , Cell Line, Tumor , Dictyostelium/genetics , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Expression , Humans , Lung Neoplasms/genetics , Neoplasm Proteins/genetics , Protozoan Proteins/biosynthesis , Protozoan Proteins/genetics , Response Elements , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism
10.
J Cell Biochem ; 109(2): 375-82, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19950202

ABSTRACT

A mouse cell line 3T3-L1 is differentiated into adipocytes when treated with an inducer cocktail (IDX) (insulin, dexametahsone, and a cAMP phosphodiesterase inhibitor of isobutyl-methylxanthine (IBMX)). Here, we report that PLD1, but not PLD2, mRNA and protein increased during the early differentiation process. Our analysis shows that IDX resulted in a sequential induction of C/EBPbeta, PLD1, and C/EBPalpha which is a key transcription factor of late adipocyte differentiation. Among the three inducers, IBMX + any other inducer induced mild adipocyte differentiation, whereas insulin + dexamethasone did not. IBMX increased PLD1 but not PLD2 mRNA. Forskolin, an adenylate cyclase activator, and dbcAMP also increased PLD1 mRNA, suggesting the cellular cAMP as the inducer of both adipocyte differentiation and PLD1 transcription. We focused on the regulatory mechanism of PLD1 transcription during this differentiation process. IDX or a combination of inducers including IBMX increased PLD1 promoter activity, which is consistent with mRNA analysis. Promoter analysis identified two adjacent C/EBP motifs located between -338 and -231 bp from the first exon as the IBMX responsive elements. Furthermore, overexpression of C/EBPbeta, but not C/EBPalpha, increased PLD1 mRNA and PLD1 5' promoter activity. EMSA and chromatin immunoprecipitation assay confirmed the direct binding of C/EBPbeta, but not C/EBPalpha, to these C/EBP motifs of PLD1 5' promoter. Our results show that PLD1 is a target gene of C/EBPbeta through the increased cellular cAMP during early adipocyte differentiation of 3T3-L1 cells.


Subject(s)
1-Methyl-3-isobutylxanthine/pharmacology , Adipogenesis/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cyclic AMP/metabolism , Phospholipase D/genetics , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Animals , Dexamethasone/pharmacology , Gene Expression Regulation/drug effects , Insulin/pharmacology , Mice , Phosphodiesterase Inhibitors/pharmacology , Phospholipase D/metabolism , Response Elements
11.
J Neurochem ; 112(2): 511-20, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19903244

ABSTRACT

Ceramide is the central lipid in the sphingolipid metabolism. Ceramide kinase (CERK) and its product, ceramide 1-phosphate, have been implicated in various cellular functions. However, the regulatory mechanism of CERK gene expression remains to be determined. Here, we examined CERK mRNA level during all-trans retinoic acid (ATRA)-induced differentiation of a human neuroblastoma cell line, SH-SY5Y. ATRA reduced CERK mRNA and protein levels. Over-expression and small interfering RNA (siRNA) of CERK revealed that CERK is inhibitory against ATRA-induced neuronal differentiation and cell growth arrest. ATRA inhibited the transcriptional activity of 5'-promoter of CERK. Truncation and mutation study suggests that ATRA-responsible region was mainly located in the tandem retinoic acid responsive elements (RARE) between -40 bp and the first exon. The electrophoresis mobility shift assay revealed that ATRA produced two retarded bands, which were erased by antibody against chicken ovalbumin upstream promoter transcription factor I (COUP-TFI), RARalpha, and RXRalpha, respectively. DNA pull-down assay confirmed increased binding of these transcription factors to RARE. Transient expression of RAR, RXR, and COUP-TFI and siRNA transfection of these genes revealed that COUP-TFI inhibited CERK mRNA. Furthermore, chromatin immunoprecipitation assay showed the recruitment of co-repressors as well as three transcription factors. These results suggest that COUP-TFI was the ATRA-responsive suppressive transcription factor of CERK gene transcription.


Subject(s)
COUP Transcription Factor I/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Transcriptional Activation/drug effects , Tretinoin/pharmacology , Binding Sites/drug effects , COUP Transcription Factor I/genetics , Cell Death/drug effects , Cell Proliferation/drug effects , Electrophoretic Mobility Shift Assay/methods , Exons/drug effects , Exons/physiology , Humans , Immunoprecipitation/methods , Molecular Sequence Data , Neurites/drug effects , Neuroblastoma , Phosphotransferases (Alcohol Group Acceptor)/genetics , Promoter Regions, Genetic/drug effects , RNA, Small Interfering/pharmacology , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Retinoid X Receptors/metabolism , Transfection/methods
12.
Leuk Res ; 34(1): 50-4, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19616848

ABSTRACT

The detection of minimal residual disease (MRD) in bone marrow is very important in the clinical management of malignant lymphoma. So far, the assessment of MRD in cases of diffuse large B cell lymphoma (DLBCL) has had some technical limitations, such as requiring patient-specific primers and complicated experimental steps. To resolve these problems, we applied a tumor-specific epigenetic alteration of the p57KIP2 gene as a biomarker for detecting MRD in DLBCL. The methylation of the p57KIP2 gene was analyzed in 63 cases of DLBCL by methylation-specific real-time quantitative PCR. Methylation of the p57KIP2 gene was detected in 53 (84.1%) of these 63 cases of DLBCL. We could detect one p57KIP2 gene-methylated cell among 10,000 unmethylated cells by the serial dilution experiment. This sensitivity is proved to be equivalent to that of detection of bcl2/IgH rearrangement by real-time quantitative PCR. This sensitivity could be converted to the detection of two methylated genomes per reaction. Using clinical material, the same results were confirmed. In this study, we established a convenient and universal method for detecting MRD in DLBCL. This technique is applicable for over 80% of patients with DLBCL. This could promote systemic MRD studies in the area of DLBCL.


Subject(s)
Biomarkers, Tumor/genetics , Cyclin-Dependent Kinase Inhibitor p57/genetics , DNA Methylation , Lymphoma, Large B-Cell, Diffuse/genetics , Neoplasm, Residual , Base Sequence , DNA Primers , Humans , Lymphoma, Large B-Cell, Diffuse/pathology , Polymerase Chain Reaction , Promoter Regions, Genetic
13.
Nagoya J Med Sci ; 71(3-4): 127-36, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19994725

ABSTRACT

The underlying mechanisms of oncogene-induced phospholipase D (PLD) activation have not been fully elucidated. The effect of the mutated-ras on PLD mRNA was examined using colon cancer cell lines as well as mock- and mutated ras-transfected NIH3T3 cells. Ras-mutation and activation were correlated, and cells with enhanced ras-activation showed increased PLD1 mRNA and protein. Analysis of the 5' PLD1 promoter using a representative cell line, DLD-1 and also mutated ras-NIH3T3, showed one Sp1-site as the important ras-responsible motif. Spl inhibition with mithramycin A and Spl siRNA inhibited PLD1 protein expression and its promoter activity. Sp1 but not Sp3 protein level and increased Sp1-motif binding activity were correlated with ras activation. Furthermore, overexpression of Sp1 in drosophila SL2 cells lacking Sp family proteins increased PLD1 promoter activity. EMSA and chromatin immunoprecipitation assay confirmed the importance of Sp1 protein binding to the Sp1-motif in ras-induced PLD1 mRNA expression.


Subject(s)
Genes, ras , Mutation , Phospholipase D/genetics , Sp1 Transcription Factor/physiology , Animals , Caco-2 Cells , Gene Expression Regulation , Humans , Mice , Promoter Regions, Genetic , RNA, Messenger/analysis
14.
Biochim Biophys Acta ; 1789(11-12): 681-90, 2009.
Article in English | MEDLINE | ID: mdl-19698806

ABSTRACT

Mg(2+)-dependent neutral SMases (NSMases) have emerged as prime candidates for stress-induced ceramide production. Among isoforms identified, previous reports have suggested the importance of NSMase2. However, its activation mechanism has not been precisely reported. Here, we analyzed the mechanism of NSMase2 gene expression by the anti-cancer drug, daunorubicin (DA). DA increased cellular ceramides (C16, C18 and C24) and NSMase activity of a human breast cancer cell line, MCF-7. DA remarkably increased the NSMase2 message and protein, whereas little change in NSMase1 and NSMase3 mRNAs and only a mild increase in acid SMase mRNA were observed. Overexpression and a knock down of NSMase2 indicated that NSMase2 played a role in DA-induced cell death. NSMase2 promoter analysis revealed that three Sp1 motifs located between -148 and -42bp upstream of the first exon were important in basic as well as in DA-induced promoter activity. Consistently, luciferase vectors containing three consensus Sp1-motifs but not its mutated form showed DA-induced transcriptional activation. DA-treated MCF-7 showed increased Sp3 protein. In SL2 cells lacking Sp family proteins, both Sp1 and Sp3 overexpression increased NSMase promoter activity. Increased binding of Sp family proteins by DA to three Sp1 motifs was shown by electrophoresis mobility shift and ChIP assays.


Subject(s)
Daunorubicin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Sphingomyelin Phosphodiesterase/genetics , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Binding Sites/genetics , Blotting, Western , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Ceramides/metabolism , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Humans , Promoter Regions, Genetic/genetics , Protein Binding , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/genetics , Sp3 Transcription Factor/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Transfection
15.
J Clin Exp Hematop ; 47(1): 15-8, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17510533

ABSTRACT

ID4 is a member of the inhibitor of DNA binding (ID) family, and inhibits the binding of basic helix-loop-helix transcription factors to DNA. In some malignant tumors, ID4 is inactivated by promoter hypermethylation, and is thought to be a candidate tumor suppressor gene (TSG). We have found that the promoter region of the ID4 gene is frequently methylated in lymphoid cell lines (100%, 9/9), primary diffuse large B-cell lymphoma (95%, 19/20), and follicular lymphoma (100%, 10/10). Somatic mutation of the ID4 gene was also examined, and no mutations were found. These findings suggest that the ID4 gene might be inactivated by DNA hypermethylation, and may function as a TSG in malignant lymphoma.


Subject(s)
DNA Methylation , Inhibitor of Differentiation Proteins/genetics , Lymphoma, B-Cell/genetics , Lymphoma, Follicular/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Promoter Regions, Genetic , Genes, Tumor Suppressor , Humans , Lymph Nodes
16.
Biochim Biophys Acta ; 1635(2-3): 104-16, 2003 Dec 30.
Article in English | MEDLINE | ID: mdl-14729073

ABSTRACT

The prolonged treatment with phorbol 12-myristate 13-acetate (PMA) of a human megakaryoblastic leukemia cell line, MEG-O1, induced increase of sphingosine kinase (SPHK) enzyme activity and SPHK1 protein expression as well as SPHK1 message. Protein kinase C (PKC) inhibitor prevented the PMA-induced SPHK1 gene expression. To elucidate the regulatory mechanism of this gene expression, we examined the promoter area (distal to the first exon) and its binding proteins. Luciferase analyses showed that the area of 300 bp from the first exon was sufficient for PMA-responsiveness, and that specificity protein 1 (Sp1)- and two activator protein 2 (AP-2)-binding motifs within this area were necessary for responsiveness. Inhibitors for PKC and MEK1 decreased this PMA-induced promoter activity. Electrophoresis mobility shift assay (EMSA) showed that Sp1 protein was originally bound to the Sp1 site and that two additional bands bound to the two AP-2 motifs were observed only when stimulated with PMA in MEG-O1 cells. The appearance of these bands resulted from binding to an unknown protein rather than AP-2. These results indicated that PMA up-regulates SPHK1 gene expression through PMA-responsive elements of the 5' promoter area of the gene, and suggested that PMA-mediated SPHK1 gene expression would be mediated via PKC- and ERK-dependent signal transduction pathway by binding the transcription factor to AP-2 motifs.


Subject(s)
Gene Expression Regulation, Enzymologic , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinase C/physiology , Binding Sites , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Enzymologic/drug effects , Humans , Leukemia , Mitogen-Activated Protein Kinases/physiology , Oligonucleotide Probes , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/genetics , Promoter Regions, Genetic/physiology , Signal Transduction , Sp1 Transcription Factor/metabolism , Tetradecanoylphorbol Acetate , Transcription Factor AP-2 , Transcription Factors/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...