Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Oncotarget ; 9(30): 21396-21410, 2018 Apr 20.
Article in English | MEDLINE | ID: mdl-29765548

ABSTRACT

Peripheral T-cell lymphoma is an aggressive non-Hodgkin's lymphoma characterized by excessive proliferation of transformed mature T cells. The number and nature of genetic aberrations required and sufficient for transformation of normal T cells into lymphomas is unknown. Here, using a combinatorial in vitro-approach, we demonstrate that overexpression of MYC together with activated AKT in conditions of inhibition of intrinsic apoptosis rapidly resulted in transformation of mature mouse T cells with a frequency approaching 100%. Injection of transformed cells into mice resulted in rapid development of aggressive T cell lymphoma, characterized by spread to several organs, destruction of tissue architecture and rapid death of the animals. TcR-sequencing revealed a polyclonal repertoire of tumor cells indicating that co-expression of MYC, activated AKT and BCLXL is sufficient for tumor transformation and do not require acquisition of additional genetic events. When analyzing cells with inducible expression we found that proliferation of transformed T cells required sustained expression of both MYC and AKT. AKT exerted a dual function as it inhibited induction of, and promoted exit from, cellular quiescence and contributed to inhibion of apoptosis. Downregulation of AKT and/or MYC together with BCLXL resulted in rapid and complete elimination of cells through induction of apoptotic cell death.

2.
Cancer Sci ; 108(2): 200-207, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27960034

ABSTRACT

Fusion tyrosine kinases play a crucial role in the development of hematological malignancies. FIP1L1-PDGFRA is a leukemogenic fusion kinase that causes chronic eosinophilic leukemia. As a constitutively active kinase, FIP1L1-PDGFRA stimulates downstream signaling molecules, leading to cellular proliferation and the generation of an anti-apoptotic state. Contribution of the N-terminal FIP1L1 portion is necessary for FIP1L1-PDGFRA to exert its full transforming activity, but the underlying mechanisms have not been fully characterized. We identified PIAS1 as a FIP1L1-PDGFRA association molecule by yeast two-hybrid screening. Our analyses indicate that the FIP1L1 portion of FIP1L1-PDGFRA is required for efficient association with PIAS1. As a consequence of the association, FIP1L1-PDGFRA phosphorylates PIAS1. Moreover, the kinase activity of FIP1L1-PDGFRA stabilizes PIAS1. Therefore, PIAS1 is one of the downstream targets of FIP1L1-PDGFRA. Moreover, we found that PIAS1, as a SUMO E3 ligase, sumoylates and stabilizes FIP1L1-PDGFRA. In addition, suppression of PIAS1 activity by a knockdown experiment resulted in destabilization of FIP1L1-PDGFRA. Therefore, FIP1L1-PDGFRA and PIAS1 form a positive cross-talk through their enzymatic activities. Suppression of sumoylation by ginkgolic acid, a small molecule compound inhibiting a SUMO E1-activating enzyme, also destabilizes FIP1L1-PDGFRA, and while the tyrosine kinase inhibitor imatinib suppresses FIP1L1-PDGFRA-dependent cell growth, ginkgolic acid or siRNA of PIAS1 has a synergistic effect with imatinib. In conclusion, our results suggest that sumoylation by PIAS1 is a potential target in the treatment of FIP1L1-PDGFRA-positive chronic eosinophilic leukemia.


Subject(s)
Cell Nucleus/metabolism , Oncogene Proteins, Fusion/metabolism , Protein Inhibitors of Activated STAT/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , STAT1 Transcription Factor/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Apoptosis , HEK293 Cells , Humans , Hypereosinophilic Syndrome/drug therapy , Hypereosinophilic Syndrome/metabolism , Imatinib Mesylate/therapeutic use , Immunoblotting , Immunoprecipitation , Oncogene Proteins, Fusion/chemistry , Protein Inhibitors of Activated STAT/chemistry , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/metabolism , Receptor, Platelet-Derived Growth Factor alpha/chemistry , STAT1 Transcription Factor/chemistry , Signal Transduction , Sumoylation , Transfection/methods , mRNA Cleavage and Polyadenylation Factors/chemistry
3.
Ann Hematol ; 93(9): 1473-81, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24763514

ABSTRACT

FIP1-like 1 (FIP1L1) is associated with two leukemogenic fusion genes: FIP1L1-retinoic acid receptor alpha (RARA) and FIP1L1-platelet-derived growth factor receptor alpha (PDGFRA). Analyses of a series of deletion mutants revealed that the FIP1 motif in FIP1L1-RARA plays a pivotal role in its homodimerization and transcriptional repressor activity. However, in FIP1L1-PDGFRA, the C-terminal PDGFRA portion possesses the ability of forming a homodimer by itself, making FIP1L1 dispensable for constitutive activation of this kinase. Both the full-length and the C-terminal PDGFRA portion of FIP1L1-PDGFRA could transform the IL-3-dependent hematopoietic cell line, BAF-B03. Moreover, when either the full-length or the C-terminal PDGFRA portion of FIP1L1-PDGFRA was introduced in these cells, they grew in the absence of IL-3. The cells having the C-terminal PDGFRA portion of FIP1L1-PDGFRA, however, were partially IL-3 dependent, whereas the cells having the full-length FIP1L1-PDGFRA became completely IL-3 independent for their growth. Taken together, these results show that FIP1L1 differentially contributes to the pathogenesis of distinct types of leukemia.


Subject(s)
Leukemia/classification , Leukemia/genetics , Oncogene Proteins, Fusion/physiology , Receptor, Platelet-Derived Growth Factor alpha/physiology , mRNA Cleavage and Polyadenylation Factors/physiology , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , HEK293 Cells , HeLa Cells , Humans , Interleukin-3/pharmacology , Oncogene Proteins, Fusion/chemistry , Protein Interaction Domains and Motifs/genetics , Protein Multimerization/genetics , Receptor, Platelet-Derived Growth Factor alpha/chemistry , mRNA Cleavage and Polyadenylation Factors/chemistry
4.
Asian Pac J Cancer Prev ; 13(9): 4827-34, 2012.
Article in English | MEDLINE | ID: mdl-23167427

ABSTRACT

Tumor suppressor genes have received much attention for their roles in the development of human malignancies. Gelsolin has been found to be down-regulated in several types of human cancers, including leukemias. It is, however, expressed in macrophages, which are the final differentiation derivatives for the monocytic myeloid lineage, implicating this protein in the differentiation process of such cells. In order to investigate the role of gelsolin in leukaemic cell differentiation, stable clones over-expressing ectopic gelsolin, and a control clone were established from U937 leukaemia cells. Unlike the control cells, both gelsolin-overexpressing clones displayed retarded growth, improved monocytic morphology, increased NADPH and NSE activities, and enhanced surface expression of the ß-integrin receptor, CD11b, when compared with the parental U937 cells. Interestingly, RT- PCR and western blot analysis also revealed that gelsolin enhanced p21CIP1 mRNA and protein expression in the overexpressing clones. Moreover, transient transfection with siRNA silencing P21CIP1, but not the control siRNA, resulted in a reduction in monocytic differentiation, accompanied by an increase in proliferation. In conclusion, our work demonstrates that gelsolin, by itself, is capable of inducing monocytic differentiation in U937 leukaemia cells, most probably through p21CIP1 activation.


Subject(s)
Cell Differentiation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gelsolin/metabolism , Gene Expression Regulation, Neoplastic , Monocytes/metabolism , CD11b Antigen/metabolism , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Esterases/metabolism , Gelsolin/genetics , Genetic Vectors , Humans , Monocytes/pathology , NADP/metabolism , Phenotype , RNA Interference , RNA, Messenger/metabolism , Receptors, Cell Surface/metabolism , Transcription Factors/metabolism , Transfection , U937 Cells , Up-Regulation
5.
Biochem Biophys Res Commun ; 427(4): 790-5, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-23044421

ABSTRACT

AMP-activated protein kinase (AMPK) is a hetero-trimeric Ser/Thr kinase composed of a catalytic α subunit and regulatory ß and γ subunits; it functions as an energy sensor that controls cellular energy homeostasis. In response to an increased cellular AMP/ATP ratio, AMPK is activated by phosphorylation at Thr172 in the α-subunit by upstream AMPK kinases (AMPKKs), including tumor suppressor liver kinase B1 (LKB1). To elucidate more precise molecular mechanisms of AMPK activation, we performed yeast two-hybrid screening and isolated the complementary DNA (cDNA) encoding the nuclear protein Artemis/DNA cross-link repair 1C (DCLRE1C) as an AMPKα2-binding protein. Artemis was found to co-immunoprecipitate with AMPKα2, and the co-localization of Artemis with AMPKα2 in the nucleus was confirmed by immunofluorescence staining in U2OS cells. Moreover, over-expression of Artemis enhanced the phosphorylation of AMPKα2 and the AMPK substrate acetyl-CoA carboxylase (ACC). Conversely, RNAi-mediated knockdown of Artemis reduced AMPK and ACC phosphorylation. In addition, Artemis markedly increased the physical association between AMPKα2 and LKB1. Taken together, these results suggest that Artemis functions as a positive regulator of AMPK signaling by stabilizing the LKB1-AMPK complex.


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/metabolism , Cell Line, Tumor , DNA-Binding Proteins , Endonucleases , Enzyme Stability , Humans , Immunoprecipitation , Nuclear Proteins/genetics
6.
J Innate Immun ; 3(3): 216-26, 2011.
Article in English | MEDLINE | ID: mdl-21454962

ABSTRACT

With an array of activating and inhibitory receptors, natural killer (NK) cells can specifically eradicate infected and transformed cells. Target cell killing is achieved through directed release of lytic granules. Recognition of target cells also induces production of chemokines and cytokines that can coordinate immune responses. Upon contact with susceptible cells, a multiplicity of activating receptors can induce signals for adhesion. Engagement of the integrin leukocyte functional antigen-1 mediates firm adhesion, provides signals for granule polarization and orchestrates the structure of an immunological synapse that facilitates efficient target cell killing. Other activating receptors apart from leukocyte functional antigen-1 signal for lytic granule exocytosis, a process that requires overcoming a threshold for activation of phospholipase C-γ, which in turn induces STIM1- and ORAI1-dependent store-operated Ca²+ entry as well as exocytosis mediated by the SNARE-containing protein syntaxin-11 and regulators thereof. Cytokine and chemokine release follows a different secretory pathway which also requires phospholipase C-γ activation and store-operated Ca²+ entry. Recent studies of human NK cells have provided insights into a hierarchy of effector functions that result in graded responses by NK cell populations. Responses display cellular heterogeneity and are influenced by environmental cues. This review highlights recent knowledge gained on the molecular pathways for and regulation of NK cell activation.


Subject(s)
Killer Cells, Natural/immunology , Lymphocyte Activation , Receptors, Natural Killer Cell/immunology , Signal Transduction , Animals , Cell Communication , Cytokines/immunology , Humans , Immunity, Innate
7.
Clin Cancer Res ; 16(15): 3964-75, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20670950

ABSTRACT

PURPOSE: To develop a novel diagnostic method for the assessment of drug efficacy in chronic myeloid leukemia (CML) patients individually, we generated a biosensor that enables the evaluation of BCR-ABL kinase activity in living cells using the principle of fluorescence resonance energy transfer (FRET). EXPERIMENTAL DESIGN: To develop FRET-based biosensors, we used CrkL, the most characteristic substrate of BCR-ABL, and designed a protein in which CrkL is sandwiched between Venus, a variant of YFP, and enhanced cyan fluorescent protein, so that CrkL intramolecular binding of the SH2 domain to phosphorylated tyrosine (Y207) increases FRET efficiency. After evaluation of the properties of this biosensor by comparison with established methods including Western blotting and flow cytometry, BCR-ABL activity and its response to drugs were examined in CML patient cells. RESULTS: After optimization, we obtained a biosensor that possesses higher sensitivity than that of established techniques with respect to measuring BCR-ABL activity and its suppression by imatinib. Thanks to its high sensitivity, this biosensor accurately gauges BCR-ABL activity in relatively small cell numbers and can also detect <1% minor drug-resistant populations within heterogeneous ones. We also noticed that this method enabled us to predict future onset of drug resistance as well as to monitor the disease status during imatinib therapy, using patient cells. CONCLUSION: In consideration of its quick and practical nature, this method is potentially a promising tool for the prediction of both current and future therapeutic responses in individual CML patients, which will be surely beneficial for both patients and clinicians.


Subject(s)
Biosensing Techniques/methods , Fluorescence Resonance Energy Transfer/methods , Fusion Proteins, bcr-abl/analysis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/therapeutic use , Benzamides , Blotting, Western , Cell Separation , Drug Resistance, Neoplasm/drug effects , Flow Cytometry , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Luminescent Agents , Nuclear Proteins/metabolism , Phosphorylation , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Sensitivity and Specificity
8.
Am J Pathol ; 175(1): 400-11, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19528349

ABSTRACT

Tumor hypoxia directly promotes genomic instability and facilitates cell survival, resulting in tumors with a more aggressive phenotype. The proto-oncogene pim-1 regulates apoptosis and the cell cycle by phosphorylating target proteins. Overexpression of Pim-1 can cause genomic instability and contribute to lymphomagenesis. It is not clear whether Pim-1 is involved in hypoxia-mediated tumor survival in solid tumors. Here, we show that hypoxia can stabilize Pim-1 by preventing its ubiquitin-mediated proteasomal degradation and can cause Pim-1 translocation from the cytoplasm to the nucleus. Importantly, overexpression of Pim-1 increases NIH3T3 cell transformation exclusively under hypoxic conditions, suggesting that Pim-1 expression under hypoxia may be implicated in the transformation process of solid tumors. Also, blocking Pim-1 function by introduction of dominant negative Pim-1 resensitizes pancreatic cancer cells to apoptosis induced by glucose-deprivation under hypoxia. Introduction of short interfering RNAs for Pim-1 also resensitizes cancer cells to glucose deprivation under hypoxic conditions, while forced overexpression of Pim-1 causes solid tumor cells to become resistant to glucose deprivation. Moreover, dominant negative Pim-1 reduces tumorigenicity in pancreatic cancer cells and HeLa xenograft mouse models. Together, our studies indicate that Pim-1 plays a distinct role in solid tumor formation in vivo, implying that Pim-1 may be a novel target for cancer therapy.


Subject(s)
Cell Hypoxia/physiology , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Animals , Blotting, Western , Cell Transformation, Neoplastic/genetics , Flow Cytometry , Fluorescent Antibody Technique , HeLa Cells , Humans , Immunohistochemistry , Mice , Mice, SCID , NIH 3T3 Cells , Neoplasms/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins c-pim-1/genetics , Transplantation, Heterologous , Ubiquitination , Up-Regulation
9.
Nat Prod Res ; 23(6): 561-71, 2009.
Article in English | MEDLINE | ID: mdl-19384733

ABSTRACT

Plants are an important source of several clinically useful anti-cancer agents. A volatile extract was obtained from Ricinus communis L. (Euphorbiaceae) leaves by standard hydrodistillation and subsequent extraction of the cohobated water in chloroform. GC-MS identified three monoterpenoids: 1,8-cineole, camphor and alpha-pinene, and a sesquiterpenoid: beta-caryophyllene, as the main constituents. The leaf extract is cytotoxic to several human tumour cell lines in a dose-dependent fashion, with IC(50) values ranging between 10-40 microg mL(-1). Apoptosis was shown to be induced in SK-MEL-28 human melanoma cells at a concentration of 20 microg mL(-1), as identified by means of morphological examination, nuclear staining and flow cytometric analysis of DNA content. Translocation of phosphatidyl serine to the cell membrane's external surface and loss of mitochondrial membrane potential were also detected. This study provides further insight into the potential use of mixtures of terpenoids as they occur in nature, as inducers of apoptosis in cancer cells.


Subject(s)
Cytotoxins/isolation & purification , Monoterpenes/isolation & purification , Plant Extracts/isolation & purification , Plant Leaves/chemistry , Ricinus/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cytotoxins/pharmacology , Dose-Response Relationship, Drug , Flow Cytometry , Gas Chromatography-Mass Spectrometry , Humans , Inhibitory Concentration 50 , Microscopy, Phase-Contrast , Monoterpenes/pharmacology , Plant Extracts/pharmacology
10.
Biol Blood Marrow Transplant ; 14(11): 1226-30, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18940676

ABSTRACT

Hepatitis B virus (HBV)-reverse seroconversion (RS) following allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a frequent late-onset complication in recipients with previous HBV infection. We followed 38 allo-HSCT recipients with previous HBV infection, and conducted posttransplant HB vaccine intervention in 13 recipients. First, we followed the recipients without any intervention (historic control) until 2003; hence, we commenced HB vaccination. Out of the patients who underwent transplantation after 2003, 13 recipients were immunized by a standard three-dose regimen after immunosuppressant cessation (vaccine group), whereas 12 recipients were observed without any intervention (nonvaccine group). Eight of the 13 historic control group recipients and 3 of the 12 nonvaccine group recipients, but none of the 13 vaccine group recipients, suffered HBV-RS. Cumulative risks of HBV-RS at 3 years post-HSCT in the historic control, nonvaccine and vaccine groups were 41%, 39%, and 0% respectively (P=.022). We therefore conclude that intervention with HB vaccines is significantly effective in preventing post-HSCT HBV-RS.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hepatitis B/prevention & control , Vaccination , Virus Activation , Adult , Aged , Female , Hematologic Neoplasms/complications , Hematologic Neoplasms/therapy , Hepatitis B/complications , Hepatitis B Vaccines , Hepatitis B virus , Humans , Male , Middle Aged , Retrospective Studies , Risk Factors , Time Factors , Transplantation, Homologous
12.
J Immunol ; 179(7): 4616-25, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17878359

ABSTRACT

Cancers escape immune surveillance through the manipulation of the host's immune system. Sequestration of dendritic cells (DCs) within tumor tissues and the subsequent inhibition of their migration is one of the several mechanisms by which tumors induce immunosuppression. In view of recent findings depicting the improvement of tumor immune responses in cancer patients following all-trans retinoic acid (ATRA) treatment, we sought to identify the effects of ATRA on DC mobility in the context of tumor immunotherapy. Our results demonstrate that ATRA, added to differentiating murine bone marrow progenitor cells, enhances the invasive capacity of the resulting DCs. Immature DCs injected intratumorally in mice show increased accumulation in draining lymph nodes, but not in nondraining lymph nodes and spleens, when differentiated in the presence of ATRA. The in vitro migration of mature DCs through the basement membrane matrix toward the lymphoid chemokines CCL19 and CCL21 is enhanced in these cells, albeit not in the presence of a matrix metalloproteinase (MMP) inhibitor. An increase in MMP production with a simultaneous decrease in the production of their inhibitors (tissue inhibitors of matrix metalloproteinase or TIMPs) is provoked by ATRA. This affects the MMP/TIMP balance in DCs, in particular that of MMP-9 and TIMP-1, favoring protease activity and thus allowing for enhanced DC mobilization. In conclusion, this study demonstrates that ATRA is capable of improving DC trafficking in a tumor milieu and, in view of the encouraging results obtained in the clinic, further supports the notion that ATRA might be a valuable chemical adjuvant to current immunotherapeutic strategies for cancer.


Subject(s)
Dendritic Cells/drug effects , Dendritic Cells/enzymology , Lymph Nodes/drug effects , Lymph Nodes/enzymology , Matrix Metalloproteinases/metabolism , Tissue Inhibitor of Metalloproteinases/metabolism , Tretinoin/pharmacology , Animals , Cell Differentiation , Cell Movement/drug effects , Cells, Cultured , Dendritic Cells/cytology , Female , Gene Expression Regulation , Lymph Nodes/cytology , Matrix Metalloproteinases/genetics , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Receptors, CCR7 , Receptors, Chemokine/genetics , Tissue Inhibitor of Metalloproteinases/genetics
13.
Exp Cell Res ; 313(15): 3337-48, 2007 Sep 10.
Article in English | MEDLINE | ID: mdl-17651733

ABSTRACT

There is accumulating evidence demonstrating that HIF-1 functions as a key regulator of the adaptation responses to hypoxia in cancer tissues. To this evidence, we add that adaptation responses to glucose deprivation plus hypoxia are also necessary for the survival of tumor cells in the tumor microenvironment as cancer tissues are exposed to glucose deprivation as well as hypoxia. We found that adrenomedullin (AM), VEGF, Glut-1, Glut-3, and Hexokinase-2 among 45 hypoxia-inducible genes investigated were expressed at higher levels under glucose-deprived hypoxic conditions than under hypoxic conditions. Glucose deprivation activated the AMPK under normoxia and hypoxia. Compound C, an inhibitor of AMPK, suppressed the expressions of AM and VEGF which had already been enhanced under glucose-deprived hypoxic conditions. siRNAs for both AMPKalpha1 and AMPKalpha2 suppressed the expressions of AM and VEGF. HIF-1alpha protein level and the transcriptional activity of HIF-1 under glucose-deprived hypoxic conditions were thus found to be similar to those under hypoxic conditions. Furthermore, tumor cells in 15 out of 20 human pancreatic cancer tissue specimens were stained by anti-phospho-AMPKalpha antibody. Our results thus suggest that the enhanced expressions of those genes mediated by the activation of AMPK and HIF-1 therefore play a pivotal role in the tumor formation of pancreatic cancers.


Subject(s)
Glucose Transport Proteins, Facilitative/biosynthesis , Glucose/physiology , Hexokinase/biosynthesis , Hypoxia-Inducible Factor 1/biosynthesis , Neovascularization, Pathologic/metabolism , Pancreatic Neoplasms/metabolism , Protein Kinases/biosynthesis , AMP-Activated Protein Kinase Kinases , Animals , Biomarkers, Tumor/biosynthesis , Cell Hypoxia , Cell Line, Tumor , Gene Expression Regulation , Humans , Hypoxia , Mice , Mice, SCID , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/blood supply , Phosphorylation , Protein Kinases/genetics , RNA, Small Interfering/genetics , Up-Regulation
14.
Cancer Res ; 67(7): 3345-55, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17409444

ABSTRACT

Although hypovasculature is an outstanding characteristic of pancreatic cancers, the tumor cells survive and proliferate under severe hypoxic, glucose-deprived conditions caused by low blood supply. It is well known that the hypoxia-inducible factor-1 pathway is essential for the survival of pancreatic cancer cells under hypoxic conditions. To discover how pancreatic cancer cells adapt to glucose deprivation as well as hypoxia, we sought glucose deprivation-inducible genes by means of a DNA microarray system. We identified 63 genes whose expression was enhanced under glucose-deprived conditions at >2-fold higher levels than under normal glucose conditions. Among these genes, asparagine synthetase (ASNS) was studied in detail. Although it is known to be associated with drug resistance in leukemia and oncogenesis triggered by mutated p53, its function is yet to be determined. In this study, we found that glucose deprivation induced the overexpression of ASNS through an AMP-activated protein kinase-independent and activating transcription factor-4-dependent manner and that ASNS protects pancreatic cancer cells from apoptosis induced by glucose deprivation itself. ASNS overexpression also induced resistance to apoptosis triggered by cisplatin [cis-diammine-dichloroplatinum (CDDP)] and carboplatin, but not by 5-fluorouracil, paclitaxel, etoposide, or gemcitabine. We show that glucose deprivation induces the activation of c-jun NH(2)-terminal kinase (JNK)/stress-activated protein kinase (SAPK) in a mock transfectant but not in an ASNS transfectant. Consequently, an inhibitor of JNK/SAPK decreased the sensitivity of pancreatic cancer cells to apoptosis by glucose deprivation and CDDP. These results strongly suggest that ASNS is induced by glucose deprivation and may play a pivotal role in the survival of pancreatic cancer cells under glucose-deprived conditions.


Subject(s)
Apoptosis/physiology , Aspartate-Ammonia Ligase/biosynthesis , Cisplatin/pharmacology , Glucose/deficiency , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Apoptosis/drug effects , Asparagine/metabolism , Asparagine/pharmacology , Aspartate-Ammonia Ligase/genetics , Aspartate-Ammonia Ligase/metabolism , Cell Line, Tumor , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Phosphorylation , RNA, Small Interfering/genetics , Transfection
15.
Eur J Immunol ; 35(12): 3468-77, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16259004

ABSTRACT

As most solid tumors are hypoxic, dendritic cells (DC) in solid tumors are also exposed to hypoxia. While many adaptation responses of tumor cells to hypoxia are known, it is yet to be determined how hypoxia affects the functions of DC. To explore the effects of hypoxia on the functions of DC, we compared the expression of surface markers, cytokines, chemokine receptors and matrix metalloproteinases (MMP) of human monocyte-derived DC (hmDC) differentiated under hypoxia to those differentiated under normoxia. Both groups of hmDC expressed similar levels of surface markers and cytokines. However, expression of MMP-9 and membrane type-1-MMP, as well as migrating activity, was significantly suppressed in hmDC differentiated under hypoxia compared with their normoxia counterparts. We also demonstrated that trichostatin A restored the production of MMP-9 in hmDC, under hypoxia. Collectively, our findings show that a hypoxic microenvironment suppresses the production of MMP in hmDC, most probably through the deacetylation of promoter regions of MMP, thus suppressing the migrating activity of hmDC. Our results suggest that the hypoxic microenvironment in solid tumor tissues may suppress the function of DC.


Subject(s)
Cell Movement/immunology , Dendritic Cells/cytology , Dendritic Cells/enzymology , Hypoxia/enzymology , Matrix Metalloproteinase Inhibitors , Biomarkers , Cells, Cultured , Dendritic Cells/metabolism , Humans , Matrix Metalloproteinase 1/biosynthesis , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Monocytes/cytology , Monocytes/enzymology , Monocytes/metabolism , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Tissue Inhibitor of Metalloproteinase-1/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...