Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Cells ; 11(24)2022 12 16.
Article in English | MEDLINE | ID: mdl-36552865

ABSTRACT

Kallikrein-related peptidase (KLK)6 is associated with inflammatory diseases and neoplastic progression. KLK6 is aberrantly expressed in several solid tumors and regulates cancer development, metastatic progression, and drug resistance. However, the function of KLK6 in the tumor microenvironment remains unclear. This study aimed to determine the role of KLK6 in the tumor microenvironment. Here, we uncovered the mechanism underlying KLK6-mediated cross-talk between cancer cells and macrophages. Compared with wild-type mice, KLK6-/- mice showed less tumor growth and metastasis in the B16F10 melanoma and Lewis lung carcinoma (LLC) xenograft model. Mechanistically, KLK6 promoted the secretion of tumor necrosis factor-alpha (TNF-α) from macrophages via the activation of protease-activated receptor-1 (PAR1) in an autocrine manner. TNF-α secreted from macrophages induced the release of the C-X-C motif chemokine ligand 1 (CXCL1) from melanoma and lung carcinoma cells in a paracrine manner. The introduction of recombinant KLK6 protein in KLK6-/- mice rescued the production of TNF-α and CXCL1, tumor growth, and metastasis. Inhibition of PAR1 activity suppressed these malignant phenotypes rescued by rKLK6 in vitro and in vivo. Our findings suggest that KLK6 functions as an important molecular link between macrophages and cancer cells during malignant progression, thereby providing opportunities for therapeutic intervention.


Subject(s)
Kallikreins , Melanoma , Receptor, PAR-1 , Animals , Mice , Kallikreins/metabolism , Macrophages/metabolism , Receptor, PAR-1/metabolism , Tumor Microenvironment , Tumor Necrosis Factor-alpha
2.
Cancer Lett ; 417: 141-151, 2018 03 28.
Article in English | MEDLINE | ID: mdl-29307615

ABSTRACT

Rho GTPases control a wide range of cellular processes, and their deregulation is associated with promotion of an aggressive and metastatic tumor phenotype in human cancers. Rho guanine nucleotide dissociation inhibitor 1 (RhoGDI1) plays a key role in regulating the activity of Rho GTPases. However, the underlying mechanisms are still unclear. In this study, we show that protein phosphatase 1B (PPM1B) interacts with RhoGDI1 and functions as its phosphatase. Ectopic expression of PPM1B results in dephosphorylation of RhoGDI1 and, thereby, abates the activation of RhoA, Rac1 and CDC42 by epidermal growth factor (EGF). PPM1B overexpression in Hs578T and SKBR3 human breast cancer cells decreases their motility and invasiveness in vitro and cancer metastasis in vivo. In contrast, knockdown of PPM1B in MCF-7 and MDA-MB-468 human breast cancer cells that express endogenous PPM1B enhances EGF-induced RhoGDI1 phosphorylation, activation of Rho GTPases, and cancer cell migration and invasion. Knockdown of RhoA or Rac1 by siRNA reverses the enhanced cell migration seen after PP1MB depletion. Collectively, these results indicate that PPM1B negatively regulates cancer cell motility and invasiveness through dephosphorylating RhoGDI1.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement , Protein Phosphatase 2C/metabolism , rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Phosphorylation , Protein Phosphatase 2C/genetics , RNA Interference , Transplantation, Heterologous
3.
Tumour Biol ; 39(10): 1010428317733985, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29034772

ABSTRACT

Damage to mitochondria induces mitophagy, a cellular process that is gaining interest for its therapeutic relevance to a variety of human diseases. However, the mechanism underlying mitochondrial depolarization and clearance in mitophagy remains poorly understood. We previously reported that mitochondria-induced cell death was caused by knockdown of Neisseria gonorrhoeae opacity-associated-interacting protein 5 in gastric cancer. In this study, we show that Neisseria gonorrhoeae opacity-associated-interacting protein 5 loss and gain of function modulates mitophagy induced by treatment with docetaxel, a chemotherapy drug for gastric cancer. The activation of mitophagy by Neisseria gonorrhoeae opacity-associated-interacting protein 5 overexpression promoted cell survival, preventing docetaxel-induced mitochondrial clearance. Conversely, short interfering RNA-mediated knockdown of Neisseria gonorrhoeae opacity-associated-interacting protein 5 accelerated docetaxel-induced apoptosis while increasing mitochondrial depolarization, reactive oxygen species, and endoplasmic reticulum stress and decreasing adenosine triphosphate production. We also found that the mitochondrial outer membrane proteins mitofusin 2 and phosphatase and tensin homolog-induced putative kinase 1 colocalized with Neisseria gonorrhoeae opacity-associated-interacting protein 5 in mitochondria and that mitofusin 2 knockdown altered Neisseria gonorrhoeae opacity-associated-interacting protein 5 expression. These findings indicate that Neisseria gonorrhoeae opacity-associated-interacting protein 5 modulates docetaxel-induced mitophagic cell death and therefore suggest that this protein comprises a potential therapeutic target for gastric cancer treatment.


Subject(s)
Cell Death/drug effects , Chromosomal Proteins, Non-Histone/metabolism , Mitochondria/metabolism , Mitophagy/physiology , Stomach Neoplasms/metabolism , Taxoids/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Cell Cycle Proteins , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Docetaxel , GTP Phosphohydrolases/metabolism , Humans , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Mitophagy/drug effects , Neisseria gonorrhoeae/metabolism , Phosphoric Monoester Hydrolases/metabolism , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Tensins/metabolism
5.
Cell Death Dis ; 8(3): e2682, 2017 03 16.
Article in English | MEDLINE | ID: mdl-28300829

ABSTRACT

Cystatin SN (CST1) is a specific inhibitor belonging to the cystatin superfamily that controls the proteolytic activities of cysteine proteases such as cathepsins. Our previous study showed that high CST1 expression enhances tumor metastasis and invasiveness in colorectal cancer. Recently, auranofin (AF), a gold(I)-containing thioredoxin reductase 1 (TrxR1) inhibitor, has been used clinically to treat rheumatoid arthritis. AF is a proteasome-associated deubiquitinase inhibitor and can act as an anti-tumor agent. In this study, we investigated whether CST1 expression induces autophagy and tumor cell survival. We also investigated the therapeutic effects of AF as an anti-tumor agent in colorectal cancer (CRC) cells. We found that CRC cells expressing high levels of CST1 undergo increased autophagy and exhibit chemotherapeutic resistance to AF-induced cell death, while those expressing low levels of CST1 are sensitive to AF. We also observed that knockdown of CST1 in high-CST1 CRC cells using CST1-specific small interfering RNAs attenuated autophagic activation and restored AF-induced cell mortality. Conversely, the overexpression of CST1 increased autophagy and viability in cells expressing low levels of CST1. Interestingly, high expression of CST1 attenuates AF-induced cell death by inhibiting intracellular reactive oxygen species (ROS) generation, as demonstrated by the fact that the blockage of ROS production reversed AF-induced cell death in CRC cells. In addition, upregulation of CST1 expression increased cellular glutathione reductase (GR) activity, reducing the cellular redox state and inducing autophagy in AF-treated CRC cells. These results suggest that high CST1 expression may be involved in autophagic induction and protects from AF-induced cell death by inhibition of ROS generation through the regulation of GR activity.


Subject(s)
Auranofin/pharmacology , Autophagy/drug effects , Cell Death/drug effects , Colorectal Neoplasms/metabolism , Glutathione Reductase/metabolism , Reactive Oxygen Species/metabolism , Salivary Cystatins/pharmacology , Cathepsins/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HT29 Cells , Humans , Oxidation-Reduction/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Salivary Cystatins/metabolism , Thioredoxin Reductase 1/metabolism , Up-Regulation/drug effects
6.
Oncotarget ; 7(51): 85332-85348, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27863404

ABSTRACT

Kallikrein-related peptidase 6 (KLK6) is a biomarker of gastric cancer associated with poor prognosis. Mechanisms by which KLK6 could be exploited for chemotherapeutic use are unclear. We evaluated auranofin (AF), a compound with cytotoxic effects, in KLK6-deficient cells, and we investigated whether KLK6 expression induces autophagy and acquisition of drug resistance in gastric cancer. Using cultured human cells and a mouse xenograft model, we investigated how KLK6 affects antitumor-reagent-induced cell death and autophagy. Expression levels of KLK6, p53, and autophagy marker LC3B were determined in gastric cancer tissues. We analyzed the effects of knockdown/overexpression of KLK6, LC3B, and p53 on AF-induced cell death in cancer cells. Increased KLK6 expression in human gastric cancer tissues and cells inhibited AF-induced cell motility due to increased autophagy and p53 levels. p53 dependent induction of KLK6 expression increased autophagy and drug resistance, whereas KLK6 silencing decreased the autophagy level and increased drug sensitivity. During AF-induced cell death, KLK6 and LC3B colocalized to autophagosomes, associated with p53, and were then trafficked to the cytosol. In the xenograft model of gastric cancer, KLK6 expression decreased AF-induced cell death and KLK6-induced autophagy increased AF resistance. Taken together, the data suggest that the induction of autophagic processes through KLK6 expression may increase acquisition of resistance to AF. Our findings may contribute to a new paradigm for tumor therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Auranofin/pharmacology , Autophagy/drug effects , Drug Resistance, Neoplasm , Kallikreins/metabolism , Stomach Neoplasms/drug therapy , Animals , Autophagosomes/drug effects , Autophagosomes/enzymology , Cell Line, Tumor , Cell Movement/drug effects , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Kallikreins/genetics , Mice, Nude , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Protein Transport , RNA Interference , Signal Transduction/drug effects , Stomach Neoplasms/enzymology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Time Factors , Transfection , Tumor Burden/drug effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
7.
Oncotarget ; 7(4): 4195-209, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26675260

ABSTRACT

Toll-like receptor 4 (TLR4) is important in promoting the immune response in various cancers. Recently, TLR4 is highly expressed in a stage-dependent manner in gastric cancer, but the regulatory mechanism of TLR4 expression has been not elucidated it. Here, we investigated the mechanism underlying regulation of TLR4 expression through promoter methylation and histone modification between transcriptional regulation and silencing of the TLR4 gene in gastric cancer cells. Chromatin immunoprecipitation was carried out to screen for factors related to TLR4 methylation such as MeCP2, HDAC1, and Sp1 on the TLR4 promoter. Moreover, DNA methyltransferase inhibitor 5-aza-deoxycytidine (5-aza-dC) induced demethylation of the TLR4 promoter and increased H3K4 trimethylation and Sp1 binding to reactivate silenced TLR4. In contrast, although the silence of TLR4 activated H3K9 trimethylation and MeCP2 complex, combined treatment with TLR4 agonist and 5-aza-dC upregulated H3K4 trimethylation and activated with transcription factors as Sp1 and NF-κB. This study demonstrates that recruitment of the MeCP2/HDAC1 repressor complex increases the low levels of TLR4 expression through epigenetic modification of DNA and histones on the TLR4 promoter, but Sp1 activates TLR4 high expression by hypomethylation and NF-κB signaling in gastric cancer cells.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Methyl-CpG-Binding Protein 2/metabolism , NF-kappa B/metabolism , Sp1 Transcription Factor/metabolism , Stomach Neoplasms/genetics , Toll-Like Receptor 4/genetics , Apoptosis , Cell Proliferation , Chromatin Immunoprecipitation , DNA Methylation , Gene Silencing , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histones/metabolism , Humans , Immunoenzyme Techniques , Methyl-CpG-Binding Protein 2/genetics , NF-kappa B/genetics , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sp1 Transcription Factor/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Tissue Array Analysis , Toll-Like Receptor 4/metabolism , Tumor Cells, Cultured
8.
Hum Vaccin Immunother ; 11(10): 2440-5, 2015.
Article in English | MEDLINE | ID: mdl-26091286

ABSTRACT

BR-TD-1001 was developed as a booster for the immunity maintenance of diphtheria and tetanus. The aim of this study was to evaluate the safety and immunogenicity of BR-TD-1001 (test vaccine) in comparison with placebo and an active comparator in healthy Korean adults. A randomized, double-blind, placebo-controlled, active comparator, phase I clinical trial was conducted. Fifty subjects were randomly assigned to one of 3 treatment groups in a ratio of 2:2:1, and were administered a single intramuscular dose of test vaccine, active comparator, or placebo, respectively. All subjects were monitored for 4 weeks after injection. The antibody titers of the patients 2 and 4 weeks after vaccination were compared with the baseline. The frequencies of all adverse events including adverse drug reactions in the test group were not statistically different from those of the other treatment groups (P = 0.4974, 0.3061). No serious adverse event occurred, and no subject was withdrawn from the study for safety. The seroprotection rates against both tetanus and diphtheria at 4 weeks after vaccination were over 0.95. For anti-tetanus antibody, the geometric mean titer in the test group was significantly higher than those of the other groups (P = 0.0364, 0.0033). The geometric mean titer of anti-diphtheria antibody in the test group was significantly higher than the value of the placebo (P = 0.0347) while it was not for the value of the active comparator (P = 0.8484). In conclusion, BR-TD-1001 was safe, well-tolerated, and showed sufficient immunogenicity as a booster for diphtheria and tetanus.


Subject(s)
Diphtheria Toxoid/adverse effects , Diphtheria Toxoid/immunology , Diphtheria-Tetanus Vaccine/adverse effects , Diphtheria-Tetanus Vaccine/immunology , Immunization, Secondary/methods , Tetanus Toxoid/adverse effects , Tetanus Toxoid/immunology , Adult , Antibodies, Bacterial/blood , Antitoxins/blood , Asian People , Diphtheria Toxoid/administration & dosage , Diphtheria-Tetanus Vaccine/administration & dosage , Double-Blind Method , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Healthy Volunteers , Humans , Injections, Intramuscular , Male , Placebos/administration & dosage , Tetanus Toxoid/administration & dosage , Treatment Outcome , Young Adult
9.
Oncotarget ; 5(2): 519-29, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24504172

ABSTRACT

Collagen triple helix repeat-containing 1 (CTHRC1) is known to be aberrantly upregulated in most human solid tumors, although the functional roles of CTHRC1 in colorectal cancer remain unclear. In this study, we investigated the occurrence of CTHRC1 upregulation and its role in vivo and in vitro. The expression profile and clinical importance of CTHRC1 were examined by reverse transcription-polymerase chain reaction and immunohistochemical analyses in normal and tumor patient samples. CTHRC1 was detectable in normal tissues, but also was highly expressed in tumor specimens. CTHRC1 upregulation was significantly associated with demethylation of the CTHRC1 promoter in colon cancer cell lines and tumor tissues. Clinicopathologic analyses showed that nodal status and expression of CTHRC1 (95% CI 0.999-3.984, p=0.05) were significant prognostic factors for disease-free survival. Promoter CpG methylation and hypermethylation status were measured by bisulfite sequencing and pyrosequencing analysis. Furthermore, we showed that overexpression of CTHRC1 in the SW480 and HT-29 cell lines increased invasiveness, an effect mediated by extracellular signal-regulated kinase (ERK)-dependent upregulation of matrix metalloproteinase 9 (MMP9). Consistent with this, we found that knockdown of CTHRC1 attenuated ERK activation and cancer cell invasivity. These results demonstrate that CTHRC1 expression is elevated in human colon cancer cell lines and clinical specimens, and promotes cancer cell invasivity through ERK-dependent induction of MMP9 expression. Our results further suggest that high levels of CTHRC1 expression are associated with poor clinical outcomes.


Subject(s)
Adenocarcinoma/metabolism , Colorectal Neoplasms/metabolism , Extracellular Matrix Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Matrix Metalloproteinase 9/biosynthesis , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Cell Movement/physiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease-Free Survival , Enzyme Induction , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Female , HT29 Cells , Humans , Male , Middle Aged , Transfection
10.
FEBS Lett ; 587(22): 3668-74, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24080033

ABSTRACT

Eukaryotic translation initiation factor 3 is composed of 13 subunits (eIF3a through eIF3m) and plays an essential role in translation. During apoptosis, several caspases rapidly down-regulate protein synthesis by cleaving eIF4G, -4B, -3j, and -2α. In this study, we found that the activation of caspases by cisplatin in T24 cells induces the cleavage of subunit G of the eIF3 complex (eIF3g). The cleavage site (SLRD(220)G) was identified, and we found that the cleaved N-terminus was translocated to the nucleus, activating caspase-3, and that it also showed a strong DNase activity. These data demonstrate the important roles of eIF3g in the translation initiation machinery and in DNA degradation during apoptosis.


Subject(s)
Caspase 3/metabolism , Caspase 7/metabolism , Deoxyribonucleases/metabolism , Eukaryotic Initiation Factor-3/metabolism , Active Transport, Cell Nucleus , Amino Acid Motifs , Cell Line, Tumor , Cell Nucleus/metabolism , DNA Cleavage , Enzyme Activation , Eukaryotic Initiation Factor-3/chemistry , Humans , Peptide Fragments/metabolism , Proteolysis
11.
Biochem Biophys Res Commun ; 396(3): 684-90, 2010 Jun 04.
Article in English | MEDLINE | ID: mdl-20438703

ABSTRACT

N-myc downstream-regulated gene 2 (NDRG2) implicated in cellular growth and differentiation was previously reported as it is specifically expressed in primary and in vitro-differentiated dendritic cells (DCs) from monocytes and CD34(+) progenitor cells. However, its function has yet to be investigated in DCs. Here, the novel NDRG2 function about modulation of cytokines in DC was observed in this study. The secretion of IL-10 was not found in the monocyte-derived DC cells with high level of NDRG2 expression, but IL-10 was abundantly secreted up to 1ng/ml in the monocyte-derived macrophages with low level of NDRG2 expression, and further confirmed that the expression of IL-10 was dramatically increased in NDRG2-silenced DCs under presence of LPS, and significantly reduced in the NDRG2-overexpressed U937 cells under stimulation of PMA. The secretion of IL-12p70 was significantly reduced in the siNDRG2 introduced DC cells. The intracellular signaling of IL-10 secretion was markedly inhibited by SB203580, inhibitor of p38 MAPK, in the LPS-activated DCs and phosphorylation of p38 MAPK was decreased in the NDRG2 introduced U937 cells under PMA-stimulation. Taken together, NDRG2 might have a pivotal role as one of intrinsic factors for the modulation of p38 MAPK phosphorylation, and subsequently involve in controlling of IL-10 production.


Subject(s)
Interleukin-10/biosynthesis , Myeloid Cells/metabolism , Tumor Suppressor Proteins/metabolism , Cells, Cultured , Dendritic Cells/metabolism , Humans , Imidazoles/pharmacology , Interleukin-10/genetics , Lipopolysaccharides , Macrophages/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , RNA, Small Interfering/genetics , Tumor Suppressor Proteins/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Cancer Lett ; 237(1): 56-66, 2006 Jun 08.
Article in English | MEDLINE | ID: mdl-16009487

ABSTRACT

The efficacy of cisplatin in cancer chemotherapy is limited by the development of resistance. To elucidate the molecular basis of resistance to cisplatin, we compared cisplatin-induced apoptotic responses of the parental human bladder cancer cell line, T24 and its resistant subclone, T24R2. In T24 cells, cisplatin induce apoptosis and the activation of caspase-8, -9 and -3 and poly(ADP-ribose) polymerase cleavage. The expression levels of Fas, FasL, and FADD were not changed by the treatment with cisplatin. Furthermore, neither Fas-neutralizing antibody nor dominant negative mutant of FADD affected cisplatin-induced apoptosis. Western blot analysis of subcellular fractions showed that cisplatin induced redistribution of Bax and cytochrome c. Thus, cisplatin causes apoptosis in a death receptor-independent and mitochondria-dependent fashion in T24 cells. In contrast, overexpressed Bcl-2 protein inhibited cisplatin-induced Bax translocation and its downstream events in T24R2. Downregulation of Bcl-2 by RNAi potentiated the redistribution of Bax and cytochrome c and reversed cisplatin-resistance. Our results indicate that upregulation of Bcl-2 contributes to the development of cisplatin-resistance and usage of siRNA which targets the Bcl-2 gene may offer a potential tool to reverse the resistance to cisplatin in bladder cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Caspases/metabolism , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism , Apoptosis , Caspase 3 , Caspase 8 , Caspase 9 , Cell Line, Tumor , Cytochromes c/metabolism , Enzyme Activation/drug effects , Humans , Mitochondria/drug effects , Mitochondria/enzymology , Protein Transport/drug effects , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , RNA Interference , Up-Regulation , Urinary Bladder Neoplasms
13.
Biochem Biophys Res Commun ; 338(2): 1126-36, 2005 Dec 16.
Article in English | MEDLINE | ID: mdl-16259956

ABSTRACT

Apolipoprotein A-I (apoA-I), the major protein component of serum high-density lipoprotein, exhibits anti-inflammatory activity in atherosclerosis. In this study, we demonstrate that apoA-I inhibits DC differentiation and maturation. DC differentiated from monocytes in the presence of apoA-I showed a decreased expression of surface molecules such as CD1a, CD80, CD86, and HLA-DR. In addition, these DC exhibited decreased endocytic activity and weakened allogeneic T-cell activation. During DC differentiation in the presence of apoA-I, PGE(2) and IL-10, which are known to be DC differentiation inhibitors and/or modulators of DC function, were produced at remarkable rates, whereas IL-12 production in the cells after stimulation with CD40 mAb and IFN-gamma was significantly decreased in comparison with the control DC. T cells stimulated by apoA-I-pretreated DC produced significantly low levels of IFN-gamma, and apoA-I inhibited cross-talk between DC and NK cells, in terms of IL-12 and IFN-gamma production. Therefore, apoA-I appears to play an important role in modulating both innate immune response and inflammatory response. The novel inhibitory function of apoA-I on DC differentiation and function may facilitate the development of new therapeutic interventions in inflammatory diseases.


Subject(s)
Apolipoprotein A-I/administration & dosage , Dendritic Cells/cytology , Dendritic Cells/metabolism , Dinoprostone/metabolism , Interleukin-10/metabolism , Monocytes/cytology , Monocytes/metabolism , Cell Differentiation/drug effects , Dendritic Cells/drug effects , Dose-Response Relationship, Drug , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Monocytes/drug effects
14.
J Cell Biochem ; 96(4): 869-78, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16155875

ABSTRACT

Melanocortins, besides their central roles, have also recently been reported to regulate adipocyte metabolism. In this study, we attempted to characterize the mechanism underlying alpha-melanocyte-stimulating hormone (MSH)-induced lipolysis, and compared it with that of the adrenocorticotrophin hormone (ACTH) in 3T3-L1 adipocytes. Similar to ACTH, MSH treatment resulted in the release of glycerol into the cell supernatant. The activity of hormone-sensitive lipase, a rate-limiting enzyme, which is involved in lipolysis, was significantly increased by MSH treatment. In addition, a variety of kinases, including protein kinase A (PKA) and extracellular signal-regulated kinase (ERK) were also phosphorylated as the result of MSH treatment, and their specific inhibitors caused a reduction in MSH-induced glycerol release and HSL activity, indicating that MSH-induced lipolysis was mediated by these kinases. These results suggest that PKA and ERK constitute the principal signaling pathways implicated in the MSH-induced lipolytic process via the regulation of HSL in 3T3-L1 adipocytes.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Lipolysis/drug effects , Signal Transduction/drug effects , alpha-MSH/pharmacology , 1-Methyl-3-isobutylxanthine/pharmacology , 3T3-L1 Cells , Adipocytes/cytology , Adrenocorticotropic Hormone/pharmacology , Animals , Cells, Cultured , Hydrolysis/drug effects , Mice , Nitrobenzoates/metabolism , Phenotype , Protein Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Melanocortin, Type 2/genetics , Receptors, Corticotropin/genetics , Receptors, Melanocortin
15.
Proteomics ; 5(8): 2112-22, 2005 May.
Article in English | MEDLINE | ID: mdl-15852342

ABSTRACT

Cervical carcinoma is one of the most prevalent cancers in women worldwide, and human papillomavirus (HPV) type 16 is the most common agent linked to human cervical carcinoma. In order to identify various relevant factors affected by the E7 oncogene, we established a stable cell line, which constitutively expressed E7 using the HaCaT human keratinocyte cell line. The increased expression and activity of catalase in the E7-expressing HaCaT cells (HaCaT/E7) were verified via matrix-assisted laser desorption/ionization-time of flight, Western blot, and reverse transcription-polymerase chain reaction analyses. The regulation of catalase by E7 was investigated by the detection of catalase promoter activity. E7 enhanced the activities of both the catalase promoter and nuclear factor-kappaB, one of the major transcription factors regulating the expression of the catalase gene. HaCaT/E7 cells produced lower quantities of intracellular reactive oxygen species (ROS), and appeared to be more resistant to H(2)O(2)-induced cell death. Moreover, in order to test the specific effects of E7 on catalase induction, the HaCaT/E7 cells were transiently transfected with E7 antisense vector, resulting in reductions in both the expression and activity of catalase, and a recovery of intracellular ROS levels, thus resulting in recovered sensitivity to H(2)O(2)-induced cell death. These results suggest that the HPV 16 E7 oncogene induces higher resistance to ROS-induced cell injury in the E7-infected cells, probably via the modulation of several anti-oxidant enzymes, including catalase.


Subject(s)
Catalase/biosynthesis , Cell Death , Oncogene Proteins, Viral/metabolism , Oxidative Stress/genetics , Blotting, Western , Catalase/genetics , Catalase/metabolism , Cell Line , Electrophoresis, Gel, Two-Dimensional , Electrophoretic Mobility Shift Assay , Enzyme Induction , Female , Gene Expression Regulation, Viral , Humans , Keratinocytes/cytology , Mass Spectrometry , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transfection , Up-Regulation
16.
J Ethnopharmacol ; 98(3): 339-43, 2005 Apr 26.
Article in English | MEDLINE | ID: mdl-15814270

ABSTRACT

Jaceosidin (4',5,7-trihydroxy-3',6-dimethoxyflavone) was isolated from Artemisia argyi as a putative oncogene inhibitor. Jaceosidin inhibited binding between oncoprotein E6 of the human papillomavirus and the p53 tumor suppressor protein. In addition, jaceosidin inhibited binding between the E7 oncoprotein and the Rb tumor suppressor protein, and also inhibited the function of HPV-16 harboring cervical cancer cells, including SiHa and CaSki. Collectively, jaceosidin inhibited the functions of the E6 and E7 oncoproteins of the human papillomavirus, suggesting that this compound might be used as a potential drug for the treatment of cervical cancers associated with the human papillomavirus.


Subject(s)
Artemisia , Flavonoids/pharmacology , Oncogene Proteins, Viral/drug effects , Repressor Proteins/drug effects , Tumor Cells, Cultured/drug effects , Uterine Cervical Neoplasms , Cell Survival/drug effects , Female , Flavonoids/isolation & purification , Humans , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins , Plant Leaves , Repressor Proteins/metabolism
17.
J Immunol Methods ; 296(1-2): 125-34, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15680157

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are transcription factors that directly modulate gene expression by binding to specific ligands. It has been established that PPARgamma ligands play an essential role in obesity, diabetes, and inflammation. Recently, a great deal of research has focused on the screening of PPARgamma ligands. In this study, both a human peroxisome proliferator-activated receptors gamma2 (PPARgamma2) recombinant protein and a specific monoclonal antibody against PPARgamma2 were produced in order to screen PPARgamma ligands. Analysis of deletion mutants revealed that monoclonal anti-PPARgamma antibody Pgamma48.34A possesses an antigenic determinant in the N-terminal region (31-84 a.a) of human PPARgamma2. The results of Western blot testing revealed that Pgamma48.34A recognized both glutathione S-transferase (GST)- and his-tagged human and mouse PPARgamma recombinant proteins and also identified PPARgamma in adipocytes and mouse tissues. Compared to some commercially available antibodies, this antibody does not bind with skimmed milk or BSA and exhibits a higher degree of specificity. An in vitro binding assay revealed that PPARgamma2 was bound to steroid receptor coactivator-1 (SRC-1) in a dose-responsive manner in the presence of indomethacin, and Pr48.34A was able to detect PPARgamma in a complex consisting of PPARgamma and SRC-1. Using Pgamma48.34A antibody, an enzyme-linked immunosorbent assay (ELISA) system based on the binding between fPPARgamma2 and SRC-1 has been optimized to screen new PPARgamma ligands. This new antibody, Pgamma48.34A, exhibits higher degrees of both specificity and sensitivity against PPARgamma than do other commercial anti-PPARgamma antibody, and may constitute a profound contribution to the screening of PPARgamma ligands as well as the functional study of PPARgamma.


Subject(s)
Antibodies, Monoclonal/immunology , Enzyme-Linked Immunosorbent Assay , PPAR gamma/immunology , Animals , Antibody Specificity/immunology , Antigens/immunology , Epitope Mapping , Epitopes/chemistry , Epitopes/immunology , Histone Acetyltransferases , Humans , Indomethacin/pharmacology , Ligands , Mice , Nuclear Receptor Coactivator 1 , PPAR gamma/chemistry , PPAR gamma/metabolism , Transcription Factors/drug effects , Transcription Factors/immunology
18.
Exp Mol Med ; 36(5): 428-43, 2004 Oct 31.
Article in English | MEDLINE | ID: mdl-15557815

ABSTRACT

Immunization with dendritic cells (DCs) pulsed with tumor antigen can activate tumor-specific cytotoxic T lymphocytes (CTL), which is responsible for tumor protection and regression. In this study, we examined whether DCs pulsed with necrotic tumor lysates can efficiently prevent malignant melanoma tumor cell metastasis to the lung. DCs derived from mouse bone marrow were found to produce remarkably elevated levels of IL-12 after being pulsed with the tumor lysates. Moreover, immunization with these DCs induced CTL activation and protected mice from metastasis development by intravenously inoculated tumor cells. In addition, these DCs activated NK cells in vitro in a contact-dependent manner, and induced NK activities in vivo. Furthermore, NK cell depletion before DC vaccination significantly reduced the tumor-specific CTL activity, IFN-gamma production, and IFN-gamma- inducible gene expression, and eventually interfered with the antitumor effect of tumor-pulsed DCs. Finally, similar findings with respect to NK cell dependency were obtained in the C57BL/ 6J-bg/bg mice, which have severe deficiency in cytolytic activity of NK cells. These data suggest that the antitumor effect elicited by DC vaccination, at least in a B16 melanoma model, requires the participation of both cytolytic NK and CD8(+) T cells. The findings of this study would provide important data for the effective design of DC vaccines for cancer immunotherapy.


Subject(s)
Cancer Vaccines/therapeutic use , Dendritic Cells/transplantation , Killer Cells, Natural/immunology , Melanoma, Experimental/therapy , Animals , Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Chemokine CCL7 , Cytokines/biosynthesis , Cytokines/immunology , Dendritic Cells/immunology , Female , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-12/biosynthesis , Interleukin-12/immunology , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Lymphocyte Activation/immunology , Lymphocyte Depletion , Melanoma, Experimental/immunology , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Monocyte Chemoattractant Proteins/biosynthesis , Monocyte Chemoattractant Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology
19.
Cancer Lett ; 212(1): 61-70, 2004 Aug 20.
Article in English | MEDLINE | ID: mdl-15246562

ABSTRACT

The potent anti-cancer agent cis-diamminedichloroplatinum (II) (cisplatin) is currently used for treating bladder cancer. However, clinical use of this drug for long periods is often limited because of the appearance of cisplatin-resistant bladder tumor cells. We employed the method of a differential display reverse transcriptase polymerase chain reaction to identify the differentially expressed genes in the parental human bladder cancer cell line, T24 and three cisplatin-resistant cell lines. We report here that cisplatin-resistant cell lines overexpress Bcl-2 family protein Bcl-2-related gene expressed in fetal liver (Bfl-1)/A1 as compared with their parental cell. Cisplatin and gamma-irradiation induced expression of Bfl-1/A1 in T24R2 cells but not in T24 cells. Among Bcl-2 family members, Bfl-1/A1 showed the most significant alteration of the expression level in resistant cells. The nuclear translocation of nuclear factor-kappaB (NF-kappaB) by cisplatin and gamma-irradiation selectively occurred in T24R2 cells. Mitochondrial depolarization and cell death by cisplatin were also prevented in T24R2 cells. Moreover, Bfl-1/A1 inhibited cisplatin- and TNF-alpha-induced apoptosis in BOSC23 cells. Our findings suggest that the induction of Bfl-1/A1 by NF-kappaB may be important in controlling resistance to cisplatin responses in bladder tumor cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , NF-kappa B/pharmacology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Apoptosis , Humans , Minor Histocompatibility Antigens , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Up-Regulation
20.
Proteomics ; 4(3): 839-48, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14997504

ABSTRACT

Human papillomaviruses (HPVs) have been recognized as the primary cause of cervical cancer. HPV 16 E7 binds to tumor suppressor retinoblastoma protein, and interferes with its function, causing release of the transcription factor E2F, which influences expression of cell cycle-related genes. This study was performed to identify the genes and proteins modulated by the HPV E7 oncogene. An HPV-negative cervical cancer cell line (C33A) was prepared to establish a stable cell line expressing E7. In order to analyze the target molecules modulated by E7 expression, we used two approaches: matrix-assisted laser desorption/ionization-time of flight mass spectrometry (MALDI-TOF MS) and DNA microarrays. Forty-seven spots were identified in C33A/E7 by two-dimensional electrophoresis and MALDI/TOF MS. Protein disulfide isomerase A3, integrase interactor 1 protein, growth inhibitory protein, glutathione S-transferase P, and vav proto-oncogene were down-regulated, whereas heat shock 60 kDa protein 1, Ku70 binding protein, alpha enolase, 26S proteasome subunit were up-regulated. A genomic approach using a microarray kit showed that IL-12R beta 1, cytochrome c, and tumor necrosis factor receptor II were induced by the E7 oncogene. These results suggest that E7 can evade immune surveillance by suppressing or inducing these cell signaling factors, cell cycle regulators, and chaperones.


Subject(s)
Oncogene Proteins, Viral/chemistry , Proteins/chemistry , Proteomics/methods , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cytokines/metabolism , DNA, Complementary/metabolism , DNA-Binding Proteins/metabolism , E2F Transcription Factors , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Mass Spectrometry , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins , Protein Array Analysis , Proteome , Proto-Oncogene Mas , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Silver Staining , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transcription Factors/metabolism , Transfection , Uterine Cervical Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...