Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Cell Signal ; 52: 83-94, 2018 12.
Article in English | MEDLINE | ID: mdl-30189244

ABSTRACT

Tetraspanin membrane proteins form physical complexes with signaling molecules and have been suggested to influence the signaling events of associated molecules. Of the tetraspanin proteins, CD82 has been shown to promote homotypic cell-cell adhesion, which partially accounts for its role in suppressing cancer invasion and metastasis. We found here that CD82-induced cell-cell adhesion is attributed to increased E-cadherin expression through CD82-mediated downregulation of the E-cadherin repressor Snail. The Snail repression by CD82 resulted from the reduced binding of the Sp1 transcription factor to the Snail gene promoter. Notably, high CD82 expression did not allow the fibronectin matrix to induce Sp1 phosphorylation, implicating CD82 inhibition of the fibronectin-integrin signaling-dependent Sp1 activation. Meanwhile, E-cadherin upregulated by CD82 pulled ß-catenin up to the membrane region, and consequently reduced the amount of cytoplasmic ß-catenin that was able to move into to the nucleus. The Wnt signal-induced nuclear translocation of ß-catenin was also inhibited by the CD82 function of upregulating E-cadherin. Overall, high CD82 expression was likely to suppress fibronectin adhesion-induced Sp1 activation signaling for Snail expression, resulting in continuous E-cadherin expression, which contributed not only to the maintenance of strong cell-cell adhesion but also to the blockage of nuclear ß-catenin signaling.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Cell Adhesion , Kangai-1 Protein/physiology , Prostatic Neoplasms/metabolism , Snail Family Transcription Factors/metabolism , Sp1 Transcription Factor/metabolism , beta Catenin/metabolism , Cell Line, Tumor , Fibronectins/metabolism , Humans , Integrins/metabolism , Male , Wnt Signaling Pathway
2.
Prostate ; 77(4): 350-360, 2017 03.
Article in English | MEDLINE | ID: mdl-27813113

ABSTRACT

BACKGROUND: Repression of the KAI1 metastasis suppressor gene is closely associated with malignancy and poor prognosis in many human cancer types including prostate cancer. Since gene repression in human cancers frequently results from epigenetic alterations by DNA methylation and histone modifications, we examined whether the KAI1 gene becomes silenced through these epigenetic mechanisms in prostate cancer. METHODS: KAI1 mRNA and protein levels were determined by RT-PCR and immunoblotting analyses, respectively. Methylation status of the KAI1 promoter DNA in prostate cancer cell lines and tissues was evaluated by methylation-specific PCR analysis of bisulfite-modified genomic DNAs. Methylated CpG sites in the KAI1 promoter were identified by sequencing the PCR clones of the bisulfite-modified KAI1 promoter DNA. KAI1 protein levels in human prostate cancer tissue samples were examined by immunofluorescence staining of the tissues with an anti-KAI1 antibody. RESULTS: Among the three human prostate cancer cell lines examined, PC3 and DU145 cells exhibited markedly decreased levels of KAI1 mRNA and protein as compared to LNCaP cells, even though the exogenous KAI1 promoter not being methylated was normally functional in all these cell lines. Treatment of the low KAI1-expressing cell lines with a demethylating agent, 5'-aza-2'-deoxycytidine, significantly elevated KAI1 expression levels, implicating the involvement of DNA methylation in KAI1 downregulation. Methylation of CpG islands within the KAI1 promoter region was observed in the low KAI1-expressing cells, but not in the high KAI1-expressing cells. Also, methyl CpG-binding proteins such as MBD2 and MeCP2 were complexed to the KAI1 promoter in the low KAI1-expressing cells. Bisulfite sequencing analysis identified the intensively methylated CpG residues in the KAI1 promoter clones derived from prostate cancer cells and tissues with no or low KAI1 expression. As in prostate cancer cell lines, prostate cancer tissues from patients also displayed a negative association between KAI1 expression levels and methylation status of the KAI1 promoter. CONCLUSIONS: The present data suggest that the KAI1 gene might be repressed by epigenetic alterations through the promoter CpG-site methylation during prostate cancer progression. This epigenetic mechanism could provide a clue for understanding how the KAI1 gene was silenced in metastatic prostate cancers. Prostate 77: 350-360, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
CpG Islands/physiology , Epigenetic Repression/physiology , Genes, Tumor Suppressor/physiology , Kangai-1 Protein/metabolism , Promoter Regions, Genetic/physiology , Prostatic Neoplasms/metabolism , Base Sequence , Cell Line, Tumor , Humans , Kangai-1 Protein/genetics , Male , Prostatic Neoplasms/genetics
3.
J Biol Chem ; 289(22): 15691-704, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24733393

ABSTRACT

Despite the importance of multiple tetraspanin proteins in cancer invasion and metastasis, little is known about the role and significance of tetraspanin CD81 in these processes. In the present study, we examined CD81 effects on melanoma cell invasiveness and metastasis. Transfection of CD81 into melanoma cells lacking endogenous CD81 expression significantly enhanced the migrating, invasive, and metastatic abilities of melanoma cells. Interestingly, membrane type 1 matrix metalloproteinase (MT1-MMP) expression was found in CD81-expressing melanoma cells but not in CD81-deficient cells. siRNA knockdown of CD81 in melanoma cells with endogenous CD81 demonstrated decreased MT1-MMP levels and cell motility. Notably, CD81-induced cell migration was abrogated by antibody blocking and siRNA knockdown of MT1-MMP, indicating that MT1-MMP is responsible for CD81-stimulated melanoma cell migration. Promoter analysis revealed an essential role of the Sp1 transcription factor in CD81-induced MT1-MMP transcription. We also demonstrate that the Sp1-activating Akt pathway is involved in adhesion-dependent CD81 signaling to induce MT1-MMP expression and cell motility. Importantly, human skin cancer tissue specimens displayed a positive correlation of CD81 with MT1-MMP expression levels and a close association of CD81 with malignant melanomas. Taken together, these results strongly suggest that CD81 stimulates melanoma cell motility by inducing MT1-MMP expression through the Akt-dependent Sp1 activation signaling pathway, leading to increased melanoma invasion and metastasis.


Subject(s)
Matrix Metalloproteinase 14/genetics , Melanoma/enzymology , Signal Transduction/physiology , Skin Neoplasms/enzymology , Tetraspanin 28/metabolism , Carcinoma, Basal Cell/enzymology , Carcinoma, Basal Cell/pathology , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/physiology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Matrix Metalloproteinase 14/metabolism , Melanoma/secondary , Neoplasm Invasiveness , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Skin Neoplasms/pathology , Sp1 Transcription Factor/metabolism , Tetraspanin 28/genetics , Up-Regulation/physiology
4.
Mol Immunol ; 55(3-4): 418-23, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23583086

ABSTRACT

Evidence for the immunoregulatory function of lipid molecules in addition to proteins is accumulating. Based on our previous reports on the production of prostaglandin E2 (PGE2) and prostacyclin by human follicular dendritic cells (FDCs), we hypothesized that these prostaglandins (PGs) have a regulatory effect on the survival, proliferation, and differentiation of germinal center B (GC) cells. We observed that PGE2 and a prostacyclin analog (beraprost) specifically enhanced the viable recovery of GC B cells in dose-dependent manners. FDC-like cells also mimicked the effect of PGE2, which was significantly inhibited in the presence of indomethacin. The viable recovery was due to the prevention of cell death by PGE2 and beraprost but did not result from augmented proliferation of GC B cells. The effect of PGE2 and beraprost was manifest when they were added at early times of the culture. Interestingly, we found that the combined addition of a pan-caspase inhibitor and a necroptosis inhibitor gave rise to a similar result to PGE2. Finally, PGE2 and beraprost enhanced the generation of both CD20⁻CD38⁺ plasma cells and CD20⁺CD38⁻ memory B cells from GC B cells. Our current data suggest that FDCs play an important function of sustaining GC B cell survival by providing PGs. Our data also implies that selective cyclooxygenase inhibitors administered during infection or vaccination may have an adverse effect on the course of humoral immune response.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Dendritic Cells, Follicular/immunology , Dendritic Cells, Follicular/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Prostaglandins/metabolism , B-Lymphocyte Subsets/cytology , Cell Differentiation/immunology , Cell Survival/drug effects , Cell Survival/immunology , Dinoprostone/metabolism , Dinoprostone/physiology , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Germinal Center/cytology , Humans , Immunity, Humoral
5.
Int Immunopharmacol ; 15(4): 735-42, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23499642

ABSTRACT

Immune regulating functions of lipid mediators are being recognized. Prostaglandins (PGs) are derived from phospholipid by cyclooxygenase-2 (COX-2) in inflammatory tissues. Based upon our previous data that imply an immune modulating activity of prostacyclin, we hypothesized that PGs promote the humoral immune responses in vivo. To test this hypothesis, we examined the effect of a prostacyclin analog beraprost and PGE2 on the antibody responses that were induced by immunizing mice with keyhole limpet hemocyanin (KLH). Beraprost was used due to the extremely unstable property of prostacyclin. Our results showed that beraprost indeed promoted the production of anti-KLH antibodies, which was dose-dependent and specific to beraprost because PGE2 did not modulate the antibody response compared with controls. The enhancing effect of beraprost was reproduced in the secondary responses, suggesting that memory B cell generation during the primary response was significantly affected by beraprost. Analysis of the isotypes of anti-KLH antibodies revealed that beraprost stimulated the production of IgA and IgG subisotypes but not IgM. However, germinal center B cell generation and the affinity of anti-KLH antibodies were not affected by beraprost administration. To confirm these results we immunized COX-2 knockout mice with KLH and analyzed whether the results with wild-type mice were reflected in the absence of PGs. The primary and secondary antibody responses were significantly impaired in the KO animals. The levels of anti-KLH IgG subisotypes and IgA were severely reduced in KO mice whereas those of IgM were comparable to controls. These results reveal an unrecognized function of PG in the humoral immune responses.


Subject(s)
Antibody Affinity/drug effects , B-Lymphocytes/drug effects , Epoprostenol/analogs & derivatives , Germinal Center/drug effects , Immunity, Humoral/drug effects , Immunoglobulin G/immunology , Animals , Antibody Affinity/immunology , Antigens/immunology , B-Lymphocytes/immunology , Cyclooxygenase 2/genetics , Dinoprostone/biosynthesis , Dinoprostone/immunology , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Epoprostenol/pharmacology , Flow Cytometry , Germinal Center/immunology , Hemocyanins/immunology , Immunity, Humoral/immunology , Immunoglobulin A/biosynthesis , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin M/biosynthesis , Immunoglobulin M/blood , Immunoglobulin M/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Isoforms
6.
J Biol Chem ; 287(38): 32027-39, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22843693

ABSTRACT

Tetraspanin CD151 associates with laminin-binding α(3)ß(1)/α(6)ß(1) integrins in epithelial cells and regulates adhesion-dependent signaling events. We found here that CD151 plays a role in recruiting Ras, Rac1, and Cdc42, but not Rho, to the cell membrane region, leading to the formation of α(3)ß(1)/α(6)ß(1) integrin-CD151-GTPases complexes. Furthermore, cell adhesion to laminin enhanced CD151 association with ß(1) integrin and, thereby, increased complex formation between the ß(1) family of integrins and small GTPases, Ras, Rac1, and Cdc42. Adhesion receptor complex-associated small GTPases were activated by CD151-ß(1) integrin complex-stimulating adhesion events, such as α(3)ß(1)/α(6)ß(1) integrin-activating cell-to-laminin adhesion and homophilic CD151 interaction-generating cell-to-cell adhesion. Additionally, FAK and Src appeared to participate in this adhesion-dependent activation of small GTPases. However, engagement of laminin-binding integrins in CD151-deficient cells or CD151-specific siRNA-transfected cells did not activate these GTPases to the level of cells expressing CD151. Small GTPases activated by engagement of CD151-ß(1) integrin complexes contributed to CD151-induced cell motility and MMP-9 expression in human melanoma cells. Importantly, among the four tetraspanin proteins that associate with ß(1) integrin, only CD151 exhibited the ability to facilitate complex formation between the ß(1) family of integrins and small GTPases and stimulate ß(1) integrin-dependent activation of small GTPases. These results suggest that CD151 links α(3)ß(1)/α(6)ß(1) integrins to Ras, Rac1, and Cdc42 by promoting the formation of multimolecular complexes in the membrane, which leads to the up-regulation of adhesion-dependent small GTPase activation.


Subject(s)
Gene Expression Regulation, Neoplastic , Integrin beta1/metabolism , Monomeric GTP-Binding Proteins/metabolism , Tetraspanin 24/physiology , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Cell Adhesion , Cell Line, Tumor , Humans , Laminin/chemistry , Matrix Metalloproteinase 9/metabolism , Melanoma/metabolism , Microscopy, Fluorescence/methods , Models, Biological , RNA, Small Interfering/metabolism , Signal Transduction , Subcellular Fractions
7.
Int Immunopharmacol ; 12(4): 635-42, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22406175

ABSTRACT

Prostaglandins (PGs) are emerging as important immune mediators. Since our first report on the expression of prostacyclin synthase in the germinal centers, we have investigated production mechanisms and biological functions of PG using human follicular dendritic cell (FDC)-like cells. In the previous report, we observed that TGF-ß enhances PG production, and IL-4 prevents this upregulation. To elucidate the inhibitory mechanism of IL-4, its effects on the key enzyme leading to PG production were analyzed in this study. IL-4 but not IL-10 inhibited TGF-ß-induced COX-2 expression at both mRNA and protein levels. Next the early signaling molecules of IL-4 were identified by siRNA technology. IL-4 induced tyrosine phosphorylation of STAT1, 3, and 6, but only JAK1-STAT6 pathway was responsible for the prevention of COX-2 augmentation and PG production. Phosphorylated STAT6 accumulated in the nucleus rapidly upon IL-4 addition, and the complete inhibition of COX-2 upregulation required 24 h of pretreatment with IL-4, implying that newly transcribed molecules mediate the inhibitory signals downstream of STAT6. Interestingly, unphosphorylated STAT6 proteins were constitutively expressed in the nucleus, and depletion of STAT6 impaired background level expression of COX-2 and PGs. Our results highlight the crucial roles of TGF-ß and IL-4 in the regulation of PG production, which lead us to suggest that T cells play an important role in FDC production of PGs.


Subject(s)
Dendritic Cells, Follicular/metabolism , Interleukin-4/metabolism , Janus Kinase 1/metabolism , Prostaglandins/metabolism , STAT6 Transcription Factor/metabolism , Transforming Growth Factor beta/metabolism , Cell Line , Cyclooxygenase 2/metabolism , Humans , Phosphorylation
8.
Cell Immunol ; 273(2): 109-14, 2012.
Article in English | MEDLINE | ID: mdl-22321156

ABSTRACT

Follicular dendritic cells (FDCs) are an essential cellular component of the germinal center (GC) and are believed to exert regulatory effects on the various stages of GC reactions. According to our previous reports, human FDCs express prostacyclin synthase, and prostacyclin analogues augment adhesion and co-stimulatory molecules on the surface of activated B cells. These findings prompted us to investigate whether FDCs would contribute to the antigen-presenting capability of B cells by using the well-established FDC-like cells, HK cells, and tonsillar B cells. Our results show that HK cells significantly enhance the expression levels of CD54, CD80, and CD86 on the surface of activated B cells. The enhancing effect of HK cells on CD86 is impeded by indomethacin and an EP4 antagonist, implying that a certain prostaglandin is mediating the up-regulation. Prostacyclin indeed recapitulates the enhancing effect on CD86, which is inhibited by EP4 as well as IP antagonists. B cells co-cultured with HK cells exhibit an augmented APC activity, which is inhibited by CD86 neutralization. These results reveal another unrecognized function of human FDC.


Subject(s)
B-Lymphocytes/immunology , B7-2 Antigen/immunology , Dendritic Cells, Follicular/immunology , Immunity, Innate , Antigen Presentation , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B7-2 Antigen/antagonists & inhibitors , B7-2 Antigen/genetics , B7-2 Antigen/metabolism , Cell Adhesion/immunology , Cell Communication/genetics , Cell Communication/immunology , Coculture Techniques , Cytochrome P-450 Enzyme System/immunology , Cytochrome P-450 Enzyme System/metabolism , Dendritic Cells, Follicular/cytology , Dendritic Cells, Follicular/drug effects , Epoprostenol/immunology , Epoprostenol/metabolism , Flow Cytometry , Gene Expression , Humans , Indomethacin/pharmacology , Intramolecular Oxidoreductases/immunology , Intramolecular Oxidoreductases/metabolism , Lymphocyte Activation , Palatine Tonsil/cytology , Palatine Tonsil/immunology , Primary Cell Culture , Prostaglandin Antagonists/pharmacology , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/immunology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
9.
J Immunol ; 186(7): 3866-73, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21339360

ABSTRACT

Lipid mediators are emerging as important regulators of the immune system. Based on our previous result that shows strong expression of prostacyclin synthase in the germinal center, we investigated whether prostacyclin would regulate the APC function of B cells. Owing to the very short half-life of prostacyclin in experimental conditions, we used a more stable analog, beraprost. Beraprost increased the amounts of the costimulatory molecule CD86 but not CD80 on the surface of activated B cells in time- and dose-dependent manners. However, the enhancing effect of beraprost was not observed on memory B cells, centroblasts, and centrocytes. Beraprost required BCR and CD40 signals to upregulate CD86 expression levels. Other prostanoids such as PGE(2), 6-keto-PGF(1α), and PGF(2α) failed to alter CD86 expression levels, whereas other prostacyclin analogs were as potent as beraprost. Results carried out with receptor antagonists revealed that beraprost enhanced CD86 levels by binding to prostacyclin receptor IP and by increasing intracellular cAMP concentrations. Beraprost-treated B cells potently stimulated allogeneic T cells, which was significantly abolished by CD86 neutralization. Our data imply an unrecognized cellular and molecular mechanism about the germinal center reactions.


Subject(s)
Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , B7-2 Antigen/biosynthesis , Epoprostenol/analogs & derivatives , Gene Expression Regulation/immunology , Up-Regulation/immunology , B7-2 Antigen/genetics , Cell Differentiation/immunology , Cells, Cultured , Coculture Techniques , Dendritic Cells, Follicular/immunology , Dendritic Cells, Follicular/metabolism , Epoprostenol/metabolism , Epoprostenol/physiology , Germinal Center/cytology , Germinal Center/immunology , Germinal Center/metabolism , Humans , Lymphocyte Activation/immunology , Palatine Tonsil/cytology , Palatine Tonsil/immunology , Palatine Tonsil/metabolism , Protein Binding/immunology , Receptors, Epoprostenol/metabolism , Receptors, Epoprostenol/physiology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
10.
Mol Immunol ; 48(6-7): 966-72, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21277633

ABSTRACT

Originally discovered as a B cell growth and differentiation factor, IL-4 displays a variety of functions in many different cell types. Germinal center T cells are abundant producers of IL-4. In a recent report, we demonstrated that IL-4 inhibits prostaglandins (PGs) production in follicular dendritic cell (FDC)-like cells, HK. To understand the inhibitory mechanisms of IL-4, its effects on the biosynthesis of enzymes in charge of PG production were assessed in this study. Although IL-4 did not affect COX-1 expression, it specifically inhibited LPS-induced COX-2 biosynthesis at mRNA and protein levels. Protein expression of mPGES-1, a downstream enzyme of COX-2, was also markedly diminished by IL-4 but not by IL-10, maximizing the inhibitory activity. Next, we attempted to identify the early signaling molecules that led to this inhibition of COX-2 expression. Although IL-4 induced tyrosine phosphorylation of JAK1 and TYK2, RNA interference experiments revealed that only JAK1 was responsible for the IL-4-stimulated STAT6 phosphorylation. Knocking down JAK1 and STAT6 ablated the inhibitory effect of IL-4 on COX-2 expression and significantly reduced production of PGE(2) and prostacyclin. Similar results were obtained with IL-13. Pharmacologic inhibitors of ERK and p38 mitogen-activated protein kinases inhibited the COX-2 upregulation. However, IL-4 did not affect LPS-induced phosphorylation of ERK and p38. These results stress the essential roles of JAK1 and STAT6 in the early signaling pathway of IL-4 and IL-13 leading to suppression of COX-2 expression and repression of PG production by HK cells. Our results suggest that T cells via IL-4 play a regulatory role in PG generation in FDC. IL-4 therapeutics may be applied to immune disorders where normal and ectopic expression of germinal center reactions needs to be regulated.


Subject(s)
Cyclooxygenase 2/metabolism , Dendritic Cells, Follicular/enzymology , Interleukins/pharmacology , Intramolecular Oxidoreductases/metabolism , Janus Kinase 1/metabolism , Prostaglandins/biosynthesis , STAT6 Transcription Factor/metabolism , Dendritic Cells, Follicular/drug effects , Humans , Interleukin-13/pharmacology , Interleukin-4/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Monocytes/drug effects , Monocytes/enzymology , Phosphorylation/drug effects , Prostaglandin-E Synthases , Signal Transduction/drug effects
11.
J Biol Chem ; 285(19): 14475-85, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20220142

ABSTRACT

A cancer/testis antigen, CAGE, is widely expressed in various cancer tissues and cancer cell lines but not in normal tissues except the testis. In the present study, ectopic expression of CAGE in fibroblast cells resulted in foci formation, suggesting its cell-transforming ability. Using stable HeLa transfectant clones with the tetracycline-inducible CAGE gene, we found that CAGE overexpression stimulated both anchorage-dependent and -independent cell growth in vitro and promoted tumor growth in a xenograft mouse model. Cell cycle analysis showed that CAGE augments the levels of cyclin D1 and E, thereby activating cyclin-associated cyclin-dependent kinases and subsequently accelerating the G(1) to S progression. Moreover, increased cyclin D1 and E levels in CAGE-overexpressing cells were observed even in a growth arrested state, indicating a direct effect of CAGE on G(1) cyclin expression. CAGE-induced expression of cyclins D1 and E was found to be mediated by AP-1 and E2F-1 transcription factors, and among the AP-1 members, c-Jun and JunD appeared to participate in CAGE-mediated up-regulation of cyclin D1. CAGE overexpression also enhanced retinoblastoma phosphorylation and subsequent E2F-1 nuclear translocation. In contrast, small interfering RNA-mediated knockdown of CAGE suppressed the expression of G(1) cyclins, activation of AP-1 and E2F-1, and cell proliferation in both HeLa cervical cancer cells and Malme-3M melanoma cells. These results suggest that the cancer/testis antigen CAGE possesses oncogenic potential and promotes cell cycle progression by inducing AP-1- and E2F-dependent expression of cyclins D1 and E.


Subject(s)
Cell Proliferation , Cyclin D1/metabolism , Cyclin E/metabolism , DEAD-box RNA Helicases/physiology , E2F1 Transcription Factor/metabolism , Transcription Factor AP-1/metabolism , Animals , Blotting, Western , Cell Transformation, Neoplastic , Chromatin Immunoprecipitation , Cyclin D1/genetics , Cyclin E/genetics , E2F1 Transcription Factor/antagonists & inhibitors , E2F1 Transcription Factor/genetics , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Female , G1 Phase , HeLa Cells , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Promoter Regions, Genetic , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor AP-1/genetics , Up-Regulation , Xenograft Model Antitumor Assays
12.
BMB Rep ; 41(12): 863-7, 2008 Dec 31.
Article in English | MEDLINE | ID: mdl-19123977

ABSTRACT

CD320 has been recently discovered and reported as a follicular dendritic cell (FDC) protein. Although CD320 is known to enhance proliferation of germinal center (GC) B cells, little other information is available. In this study, we investigated its cellular distribution in the GC. Confocal microscopy of human tonsil sections revealed co-localization of CD320 with CD19 and CD38 but not with CD3 indicating that GC B cells expressed CD320 in addition to FDC. In purified GC B cells, CD320 expression was inhibited in the nucleus, membrane and cytoplasm. Reverse transcriptase-polymerase chain reaction confirmed CD320 mRNA expression in B cells. These finding indicate that CD320 is expressed in B cells in addition to FDC, and that its GC activity may be more complicated than previously thought.


Subject(s)
Antigens, CD/metabolism , B-Lymphocytes/immunology , Dendritic Cells, Follicular/metabolism , ADP-ribosyl Cyclase 1/metabolism , Antigens, CD/genetics , Antigens, CD19/metabolism , Base Sequence , CD3 Complex/metabolism , DNA Primers/genetics , Gene Expression , Germinal Center/cytology , Germinal Center/immunology , Humans , Immunohistochemistry , Membrane Glycoproteins/metabolism , Microscopy, Confocal , Palatine Tonsil/cytology , Palatine Tonsil/immunology , Receptors, Cell Surface , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution
13.
Mol Immunol ; 44(12): 3168-72, 2007 May.
Article in English | MEDLINE | ID: mdl-17337058

ABSTRACT

We have recently demonstrated that human follicular dendritic cells (FDCs) strongly express prostacyclin synthase. The purpose of this study is to investigate the production mechanism of prostacyclin using the established human FDC line, HK. The levels of PGIS protein expression did not vary during the different stages of the cell cycle. We stimulated HK cells with various inflammatory cytokines but, none of the tested stimuli modulated PGIS expression significantly. However, incubation of HK cells with tumor necrosis factor (TNF)-alpha gave rise to a significant increase in the protein level of cyclooxygenase (COX)-2. Furthermore, elevated levels of prostacyclin secretion stimulated by TNF-alpha were markedly down-regulated by indomethacin and a selective COX-2 inhibitor. These results suggest that the production of prostacyclin in FDC is controlled by the regulation of upstream COX-2 but not by terminal PGIS protein production. This study has important implications for the development of new anti-inflammatory drugs.


Subject(s)
Cyclooxygenase 2/metabolism , Dendritic Cells/metabolism , Epoprostenol/biosynthesis , Cell Cycle , Cell Line , Cyclooxygenase 2/genetics , Cytochrome P-450 Enzyme System/metabolism , Dendritic Cells/cytology , Gene Expression Regulation/drug effects , Humans , Intramolecular Oxidoreductases/metabolism , Tumor Necrosis Factor-alpha/pharmacology
14.
J Biol Chem ; 281(46): 34833-47, 2006 Nov 17.
Article in English | MEDLINE | ID: mdl-16984917

ABSTRACT

The CD99 gene encodes two distinct transmembrane proteins by alternative splicing of its transcript. To examine the effects of two CD99 isoforms on the invasive phenotypes of breast cancer cells, MDA-MB-231 and MCF-7 human breast cancer cell lines were stably transfected with CD99 cDNAs encoding the major wild-type form (type I) or a minor splice variant (type II). As a result, expression of CD99 type II, but not type I, markedly elevated the motility, binding to fibronectin, MMP-9 expression, and invasiveness of MDA-MB-231 and MCF-7 breast cancer cells. In MDA-MB-435 breast cancer cells expressing both CD99 type I and type II, invasion-related cellular activities were inhibited by the transfection of small interfering RNA (siRNA) targeted to CD99 type II. Meanwhile, CD99 type II-induced MMP-9 expression in MDA-MB-231 cells was shown to be mediated by the binding of AP-1 factors to the MMP-9 gene promoter. Gel shift assay revealed that ligation of CD99 type II with antibody resulted in the binding of JunD to the AP-1 site of the MMP-9 promoter region. Initiation of CD99 type II signaling by antibody ligation increased expression of JunD and FosB AP-1 factors, along with phosphorylation of Src, Akt, p38 MAPK, ERK, and JNK. Knockdown of JunD and FosB by siRNA transfection abolished the positive effects of CD99 type II on the motility and MMP-9 expression of MDA-MB-231 cells. Increased expression of JunD and FosB as well as elevated cell motility and MMP-9 expression by CD99 type II ligation were also abrogated by inhibitors, dominant-negative forms, and siRNAs for Akt1, ERK1/2, and JNK1 but not for p38 MAPK. These results suggest that expression of a splice variant of CD99 contributes to the invasive ability of human breast cancer cells by up-regulating AP-1-mediated gene expression through the Akt-dependent ERK and JNK signaling pathways.


Subject(s)
Antigens, CD/metabolism , Breast Neoplasms/metabolism , Cell Adhesion Molecules/metabolism , Cell Movement , Extracellular Signal-Regulated MAP Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Matrix Metalloproteinase 9/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factor AP-1/metabolism , 12E7 Antigen , Alternative Splicing , Antigens, CD/genetics , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Interference , RNA, Small Interfering , Signal Transduction
15.
J Biol Chem ; 281(34): 24279-92, 2006 Aug 25.
Article in English | MEDLINE | ID: mdl-16798740

ABSTRACT

The tetraspanin membrane protein CD151 has been suggested to regulate cancer invasion and metastasis by initiating signaling events. The CD151-mediated signaling pathways involved in this regulation remain to be revealed. In this study, we found that stable transfection of CD151 into MelJuSo human melanoma cells lacking CD151 expression significantly increased cell motility, matrix metalloproteinase-9 (MMP-9) expression, and invasiveness. The enhancement of cell motility and MMP-9 expression by CD151 overexpression was abrogated by inhibitors and small interfering RNAs targeted to focal adhesion kinase (FAK), Src, p38 MAPK, and JNK, suggesting an essential role of these signaling components in CD151 signaling pathways. Also, CD151-induced MMP-9 expression was shown to be mediated by c-Jun binding to AP-1 sites in the MMP-9 gene promoter, indicating AP-1 activation by CD151 signaling pathways. Meanwhile, CD151 was found to be associated with alpha(3)beta(1) and alpha(6)beta(1) integrins in MelJuSo cells, and activation of associated integrins was a prerequisite for CD151-stimulated MMP-9 expression and activation of FAK, Src, p38 MAPK, JNK, and c-Jun. Furthermore, CD151 on one cell was shown to bind to neighboring cells expressing CD151, suggesting that CD151 is a homophilic interacting protein. The homophilic interactions of CD151 increased motility and MMP-9 expression of CD151-transfected MelJuSo cells, along with FAK-, Src-, p38 MAPK-, and JNK-mediated activation of c-Jun in an adhesion-dependent manner. Furthermore, C8161 melanoma cells with endogenous CD151 were also shown to respond to homophilic CD151 interactions for the induction of adhesion-dependent activation of FAK, Src, and c-Jun. These results suggest that homophilic interactions of CD151 stimulate integrin-dependent signaling to c-Jun through FAK-Src-MAPKs pathways in human melanoma cells, leading to enhanced cell motility and MMP-9 expression.


Subject(s)
Antigens, CD/metabolism , Matrix Metalloproteinase 9/biosynthesis , Melanoma/metabolism , Cell Adhesion , Cell Communication , Cell Line, Tumor , Cell Movement , Humans , Integrins/metabolism , MAP Kinase Signaling System , Melanoma/pathology , Protein Binding , Proto-Oncogene Proteins c-jun/metabolism , RNA, Small Interfering , Signal Transduction , Tetraspanin 24 , Up-Regulation
16.
Radiat Res ; 165(5): 562-9, 2006 May.
Article in English | MEDLINE | ID: mdl-16669711

ABSTRACT

The possibility that radiation-induced alterations in gene expression are tissue specific and are related to apoptosis was examined using samples from brain, heart, lung, spleen and intestine from female C57BL6 mice after exposure to 0.2 Gy radiation. Apoptosis was the highest in spleen and intestine, moderate in lung, and absent in brain and heart. However, the mRNA expression of Trp53 and Cdkn1a (p21) after irradiation was not different among the organ types, and immunohistochemistry revealed that all the organs expressed these two proteins after irradiation. When expression patterns of 23 genes in the organs were examined by RT-PCR, neogenine, Apo1, nuclease sensitive element binding protein 1, syntaxin, cyclin G1, hNOP56, paraoxonase and glutathione peroxidase were overexpressed after irradiation in all the organs sampled, suggesting them as universal exposure markers for low-dose radiation. Sialyltransferase may be a candidate for radiation detection in spleen and intestine, which are radiosensitive organs. Because Sod1 (Cu/ZnSOD) and alphaB crystalline were expressed only in spleen, and protein tyrosine kinase and platelet membrane glycoprotein lib were expressed in both spleen and lung, these genes may also be potential markers for detection of radiation exposure, especially low-dose radiation, in these tissues. These data suggested possible tissue-specific markers of low-dose radiation exposure and suggested potential novel genetic modifiers of radiation response.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/radiation effects , Gene Expression/radiation effects , Organ Specificity , Whole-Body Irradiation , Animals , Dose-Response Relationship, Radiation , Female , Mice , Mice, Inbred C57BL , Radiation Dosage
17.
J Immunol ; 175(3): 1658-64, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-16034106

ABSTRACT

Stromal cells in the lymphoid organs provide a microenvironment where lymphocytes undergo various biological processes such as development, homing, clonal expansion, and differentiation. Follicular dendritic cells (FDCs) in the primary and secondary follicles of the peripheral lymphoid tissues interact with lymphocytes by contacting directly or producing diffusible molecules. To understand the biological role of human FDC at the molecular level, we developed a mAb, 3C8, that recognizes FDC but not bone marrow-derived cells. Through expression cloning and proteome analysis, we identified the protein that is recognized by 3C8 mAb, which revealed that FDC expresses prostacyclin synthase. The 3C8 protein purified from FDC-like cells indeed displayed the enzymatic activity of prostacyclin synthase and converted PGH2 into prostacyclin. In addition, prostacyclin significantly inhibited proliferation of T cells but delayed their spontaneous apoptosis. These findings may help explain why T cells constitute only a minor population compared with B cells in the germinal center.


Subject(s)
Cell Proliferation , Cytochrome P-450 Enzyme System/biosynthesis , Dendritic Cells, Follicular/enzymology , Dendritic Cells, Follicular/immunology , Intramolecular Oxidoreductases/biosynthesis , Lymphocyte Count , T-Lymphocyte Subsets/cytology , Amino Acid Sequence , Apoptosis/drug effects , Apoptosis/immunology , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Cloning, Molecular , Cytochrome P-450 Enzyme System/isolation & purification , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Germinal Center/cytology , Germinal Center/enzymology , Germinal Center/immunology , Growth Inhibitors/pharmacology , Humans , Intramolecular Oxidoreductases/isolation & purification , Molecular Sequence Data , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/immunology
18.
Exp Mol Med ; 37(3): 230-9, 2005 Jun 30.
Article in English | MEDLINE | ID: mdl-16000878

ABSTRACT

Expression of matrix metalloproteinase-2 and -9 (MMP-2 and MMP-9), which correlates with tumor invasion and metastasis, has been known to be regulated by several intracellular signaling pathways. Since the CD9 membrane protein has been implicated in signal transduction and malignant progression of cancer cells, we examined the functional involvement of CD9 in the regulation of MMP-2 and MMP-9 expression by using stable CD9 transfectant clones of MelJuso human melanoma cells. The CD9 cDNA-transfected cells with elevated CD9 expression displayed increased MMP-2 and decreased MMP-9 expression when compared with the mock transfectant cells. Among several signal pathway inhibitors tested, SB203580 and SP600125, which inhibit p38 MAPK and JNK respectively, completely blocked the CD9-stimulated MMP-2 expression. Phosphorylation levels of p38 MAPK and c-Jun in MelJuso cells were also significantly increased by CD9 transfection. In addition, the down-regulation of p38 MAPK and JNK by siRNA transfection resulted in a decrease in MMP-2 expression by MelJuso cells. Promoter analysis and gel shift assay showed that the CD9-induced MMP-2 expression is mediated by a functional AP-1 site through interactions with AP-1 transcription factors including c-Jun. These results suggest that CD9 induces MMP-2 expression by activating c- Jun through p38 MAPK and JNK signaling pathways in human melanoma cells.


Subject(s)
Antigens, CD/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Matrix Metalloproteinase 2/metabolism , Melanoma/metabolism , Membrane Glycoproteins/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , Electrophoretic Mobility Shift Assay , Enzyme Activation , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/genetics , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/metabolism , Melanoma/pathology , Promoter Regions, Genetic , RNA, Small Interfering/pharmacology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tetraspanin 29 , Transcription Factor AP-1/metabolism , Transfection , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
19.
Oncogene ; 24(23): 3715-25, 2005 May 26.
Article in English | MEDLINE | ID: mdl-15806174

ABSTRACT

Since radiation-induced caspase-dependent apoptosis and ROS generation were partially prevented by HSP25 overexpression, similar to the treatment of control cells with antioxidant agents such as DPI and tiron, questions arise whether radiation-mediated ROS generation contributes to the apoptotic cell death, and also whether HSP25 overexpression can reduce ROS mediated apoptotic cell death. In the present study, radiation-induced cytochrome c release from mitochondria and activation of caspases accompanied by a decrease of mitochondrial membrane potential in Jurkat T cells were shown to be inhibited by mitochondrial complex I inhibitor rotenone, suggesting that mitochondrial ROS might be important in radiation-induced caspase-dependent apoptosis. When HSP25 was overexpressed, effects similar to the treatment of cells with the antioxidants were obtained, indicating that HSP25 suppressed radiation-induced mitochondrial alteration that resulted in apoptosis. Furthermore, activation of p38 MAP kinase by radiation was associated with radiation-induced cell death and ROS production and PKCdelta was an upstream molecule for p38 MAP kinase activation, ROS generation and subsequent caspase-dependent apoptotic events. However, in the HSP25 overexpressed cells, the above-described effects were blocked. In fact, radiation-induced membrane translocation of PKCdelta and tyrosine phosphorylation were inhibited by HSP25. Based on the above data, we suggest that HSP25 downregulates PKCdelta, which is a key molecule for radiation-induced ROS generation and mitochondrial-mediated caspase-dependent apoptotic events.


Subject(s)
Apoptosis/radiation effects , Heat-Shock Proteins/physiology , Neoplasm Proteins/physiology , Protein Kinase C/physiology , Reactive Oxygen Species/metabolism , Enzyme Activation , HSP27 Heat-Shock Proteins , Humans , Jurkat Cells , Mitochondria/physiology , Molecular Chaperones , Phosphorylation , Protein Kinase C-delta , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Free Radic Biol Med ; 36(4): 429-44, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14975446

ABSTRACT

HSP25 has been shown to induce resistance to radiation and oxidative stress; however, its exact mechanisms remain unclear. In the present study, a high concentration of H2O2 was found to induce DNA fragmentation in L929 mouse fibroblast cells, and HSP25 overexpression attenuated this phenomenon. To elucidate the mechanisms of H2O2-mediated cell death, ERK1/2, p38 MAPK, and JNK1/2 phosphorylation in the cells after treatment with H2O2 were examined. ERK1/2 and JNK1/2 were activated by H2O2; ERK1/2 activation was inhibited in HSP25-overexpressed cells, while JNK1/2 was indifferent. Inhibition of ERK1/2 activation by treatment of the cells with PD98059 or dominant-negative ERK2 transfection blocked H2O2-induced cell death; similarly treated HSP25-overexpressed cells were not at all affected. Moreover, inhibition of JNK1/2 by dominant-negative JNK1 or JNK2 transfection did not affect H2O2-mediated cell death in control cells. Dominant-negative Ras or Raf transfection inhibited H2O2-mediated ERK1/2 activation and cell death in control cells. On the contrary, HSP25-overexpressed cells did not show any differences. Upstream pathways of H2O2-mediated ERK1/2 activation and cell death involved both tyrosine kinase (PDGFbeta receptor and Src) and PKCdelta, while in HSP25-overexpressed cells these kinases did not respond to H2O2 treatment. Since HSP25 overexpression reduced reactive oxygen species (ROS), increased manganese superoxide dismutase (MnSOD) gene expression, and increased enzyme activity, involvement of MnSOD in HSP25-mediated attenuation of H2O2-mediated ERK1/2 activation and cell death was examined. Blockage of MnSOD with antisense oligonucleotides prevented DNA fragmentation and returned the ERK1/2 activation to the control level. Indeed, when MnSOD was overexpressed in L929 cells, similar to in HSP25-overexpressed cells, DNA fragmentation and ERK1/2 activation were reduced. From the above results, we suggest for the first time that reduced oxidative damage by HSP25 was due to MnSOD-mediated downregulation of ERK1/2.


Subject(s)
Apoptosis , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , MAP Kinase Signaling System/physiology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Oxidative Stress , Superoxide Dismutase/metabolism , Animals , Apoptosis/drug effects , Cell Line , Enzyme Activation/drug effects , Flavonoids/pharmacology , Gene Expression , Hydrogen Peroxide/pharmacology , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinase 9 , Mitogen-Activated Protein Kinases/metabolism , Molecular Chaperones , Oxidative Stress/drug effects , Phosphorylation/drug effects , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Transfection , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...