Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Dent Res ; 99(4): 429-436, 2020 04.
Article in English | MEDLINE | ID: mdl-31986066

ABSTRACT

Although many variants of the parathyroid hormone 1 receptor (PTH1R) gene are known to be associated with primary failure of eruption (PFE), the mechanisms underlying the link remains poorly understood. We here performed functional analyses of PTH1R variants reported in PFE patients-namely, 356C>T (P119L), 395C>T (P132L), 439C>T (R147C), and 1148G>A (R383Q)-using HeLa cells with a lentiviral vector-mediated genetic modification. Two particular variants, P119L and P132L, had severe reduction in a level of N-linked glycosylation when compared with wild-type PTH1R, whereas the other 2 showed modest alteration. PTH1R having P119L or P132L showed marked decrease in the affinity to PTH1-34, which likely led to severely impaired cAMP accumulation upon stimulation in cells expressing these mutants, highlighting the importance of these 2 amino acid residues for ligand-mediated proper functioning of PTH1R. To further gain insights into PTH1R functions, we established the induced pluripotent stem cell (iPSC) lines from a patient with PFE and the heterozygous P132L mutation. When differentiated into osteoblastic-lineage cells, PFE-iPSCs showed no abnormality in mineralization. The mRNA expression of RUNX2, SP7, and BGLAP, the osteoblastic differentiation-related genes, and that of PTH1R were augmented in both PFE-iPSC-derived cells and control iPSC-derived cells in the presence of bone morphogenetic protein 2. Also, active vitamin D3 induced the expression of RANKL, a major key factor for osteoclastogenesis, equally in osteoblastic cells derived from control and PFE-iPSCs. In sharp contrast, exposure to PTH1-34 resulted in no induction of RANKL mRNA expression in the cells expressing P132L variant PTH1R, consistent with the idea that a type of heterozygous PTH1R gene mutation would spoil PTH-dependent response in osteoblasts. Collectively, this study demonstrates a link between PFE-associated genetic alteration and causative functional impairment of PTH1R, as well as a utility of iPSC-based disease modeling for future elucidation of pathogenesis in genetic disorders, including PFE.


Subject(s)
Receptor, Parathyroid Hormone, Type 1/genetics , Tooth Diseases , Tooth Eruption , HeLa Cells , Humans , Mutation , Parathyroid Hormone
2.
Nitric Oxide ; 72: 46-51, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29183803

ABSTRACT

Osteoclasts are multinucleated giant cells differentiated from monocyte-macrophage-lineage cells under stimulation of receptor activator of nuclear factor κ-B (RANK) ligand (RANKL) produced by osteoblasts and osteocytes. Although it has been reported that nitric oxide (NO) and reactive oxygen species (ROS) are involved in this process, the mechanism by which these labile molecules promote osteoclast differentiation are not fully understood. In this study, we investigated the formation and function of 8-nitro-cGMP, a downstream molecule of NO and ROS, in the process of osteoclast differentiation in vitro. 8-Nitro-cGMP was detected in mouse bone marrow macrophages and osteoclasts differentiated from macrophages in the presence of RANKL. Inhibition of NO synthase suppressed the formation of 8-nitro-cGMP as well as RANKL-induced osteoclast differentiation from macrophages. On the other hand, RANKL-induced osteoclast differentiation was promoted by addition of 8-nitro-cGMP to the cultures. In addition, 8-nitro-cGMP enhanced the mRNA expression of RANK, the receptor for RANKL. However, 8-bromo-cGMP, a membrane-permeable derivative of cGMP, did not have an effect on either RANKL-induced osteoclast differentiation or expression of the RANK gene. These results suggest that 8-nitro-cGMP is a novel positive regulator of osteoclast differentiation, which might help to explain the roles of NO and ROS in osteoclast differentiation.


Subject(s)
Cell Differentiation , Cyclic GMP/analogs & derivatives , Osteoclasts/physiology , RANK Ligand/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Cyclic GMP/metabolism , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinases/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Macrophages/cytology , Male , Mice, Inbred Strains , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Osteoclasts/cytology , Osteoclasts/drug effects , RANK Ligand/pharmacology , Receptor Activator of Nuclear Factor-kappa B/genetics
3.
Eur Cell Mater ; 29: 290-302, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25990932

ABSTRACT

Anodically oxidized titanium surfaces, prepared by spark discharge, have micro-submicron surface topography and nano-scale surface chemistry, such as hydrophilic functional groups or hydroxyl radicals in parallel. The complexity of the surface characteristics makes it difficult to draw a clear conclusion as to which surface characteristic, of anodically oxidized titanium, is critical in each biological event. This study examined the in vitro biological changes, induced by various surface characteristics of anodically oxidized titanium with, or without, release of hydroxyl radicals onto the surface. Anodically oxidized titanium enhanced the expression of genes associated with differentiating osteoblasts and increased the degree of matrix mineralization by these cells in vitro. The phenotypes of cells on the anodically oxidized titanium were the same with, or without, release of hydroxyl radicals. However, the nanomechanical properties of this in vitro mineralized tissue were significantly enhanced on surfaces, with release of hydroxyl radicals by oxidation effects. In addition, the mineralized tissue, produced in the presence of bone morphogenetic protein-2 on bare titanium, had significantly weaker nanomechanical properties, despite there being higher osteogenic gene expression levels. We show that enhanced osteogenic cell differentiation on modified titanium is not a sufficient indicator of enhanced in vitro mineralization. This is based on the inferior mechanical properties of mineralized tissues, without either being cultured on a titanium surface with release of hydroxyl radicals, or being supplemented with lysyl oxidase family members.


Subject(s)
Osteoblasts/drug effects , Titanium/chemistry , Titanium/pharmacology , Transcriptome/drug effects , Alkaline Phosphatase/genetics , Amino Acid Oxidoreductases/genetics , Animals , Animals, Newborn , Bone Morphogenetic Protein 2/pharmacology , Cells, Cultured , Electrochemistry , Electrodes , Male , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Osteopontin/genetics , Oxidation-Reduction , Photoelectron Spectroscopy , Reactive Oxygen Species/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Sp7 Transcription Factor , Surface Properties , Transcription Factors/genetics , X-Ray Diffraction
4.
Biomaterials ; 31(33): 8546-55, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20810162

ABSTRACT

Photo-functionalized radical reactions on TiO(2) have been correlated with adsorption of organic impurities and decreasing hydrophilicity of titanium-based biomaterials. Such reactive oxygen species (ROS) spontaneously generated on oxidized titanium surfaces may also have important roles against time-dependent degradation of biological ability and adherent micro-organisms. This study examined in vitro biological ability as a function of time and antimicrobial activity on oxidized titanium surfaces without photo-functionalization. Mechanically polished titanium and thermally oxidized titanium surfaces that had been stored for 4 wks showed adsorbed organic impurities with decreased surface hydrophilicity. Even after the storage period, anodically oxidized titanium surfaces enabled super-hydrophilicity without adsorption of organic impurities, because of the ROS and the hydrophilic functional groups generated on the surfaces. The osteogenic gene expressions of osteoblasts cultured on anodically oxidized titanium surfaces with or without storage were significantly higher than those on thermally oxidized titanium and polished titanium surfaces. Titanium surfaces anodically oxidized in a solution with chloride achieved antimicrobial activity against an oral microorganism due to the amount of ROS generated on the surface. Thus, titanium anodically oxidized in solution with chloride may have potential use for titanium-based internal fixation devices.


Subject(s)
Chlorides/pharmacology , Materials Testing , Titanium/pharmacology , Adsorption/drug effects , Animals , Anti-Infective Agents/pharmacology , Cell Count , Electrodes , Humans , Mice , Microbial Sensitivity Tests , Osteoblasts/cytology , Osteoblasts/drug effects , Oxidation-Reduction/drug effects , Oxygen/analysis , Phenotype , Photoelectron Spectroscopy , Solutions , Surface Properties/drug effects , Time Factors , X-Ray Diffraction
5.
J Periodontal Res ; 42(4): 367-76, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17559635

ABSTRACT

BACKGROUND AND OBJECTIVE: Interleukin-1beta-stimulated receptor activator of nuclear factor-kappaB ligand (RANKL) expression in human periodontal ligament cells is partially mediated by endogenous prostaglandin E2, whereas mitogen-activated protein kinases (MAPKs) are implicated in regulating various interleukin-1-responsive genes. We investigated herein the involvement of MAPKs in interleukin-1beta-stimulated RANKL expression in human periodontal ligament cells. MATERIAL AND METHODS: Human periodontal ligament cells were pretreated separately with specific inhibitors of MAPKs, including extracellular signal-regulated kinase, p38 MAPK and c-Jun N-terminal kinase, and subsequently treated with interleukin-1beta. Following each treatment, the phosphorylation of each MAPK, the expression of RANKL, and the production of prostaglandin E2 were determined. RANKL activity was evaluated using an assay to determine the survival of prefusion osteoclasts. RESULTS: Interleukin-1beta induced RANKL expression at the mRNA and protein levels, as well as RANKL activity in human periodontal ligament cells. Interleukin-1beta also activated extracellular signal-regulated kinase, p38 MAPK, and c-Jun N-terminal kinase. Pretreatment with each MAPK inhibitor partially, but significantly, suppressed interleukin-1beta-induced RANKL expression and its activity, as well as prostaglandin E2 production. CONCLUSION: In human periodontal ligament cells, three types of MAPK inhibitor may abrogate RANKL expression and activity induced by interleukin-1beta, directly or indirectly through partial suppression of prostaglandin E2 synthesis. In addition, extracellular signal-regulated kinase, p38 MAPK, and c-Jun N-terminal kinase signals may co-operatively mediate interleukin-1beta-stimulated RANKL expression and its activity in those cells.


Subject(s)
Interleukin-1beta/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Periodontal Ligament/enzymology , RANK Ligand/metabolism , Adolescent , Adult , Animals , Cattle , Dinoprostone/analysis , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Female , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Male , Mice , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Osteoblasts/enzymology , Osteoclasts/enzymology , Periodontal Ligament/cytology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
6.
J Dent Res ; 86(2): 163-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17251517

ABSTRACT

Dental preparation sometimes causes transient congestion, edema, and necrosis of the pulp. We hypothesized that nitric oxide (NO) is involved in the pathophysiological changes in pulp after preparation. The mRNA and protein expression of the inducible isoform of NO synthase (iNOS) was examined in murine pulp after dental preparation. The effects of NO on the proliferation, mineralization, and apoptosis of pulp cells were also studied in vitro. We found that not only iNOS, but also mRNAs for alkaline phosphatase and plasma membrane glycoprotein-1, were expressed in the pulp after preparation. NOC-18, an NO donor, suppressed the proliferation of pulp cells without inducing cell death, whereas it promoted the mineralization of cells cultured in the presence of beta-glycerophosphate, ascorbic acid, dexamethasone, and KH(2)PO(4). Under these conditions, NOC-18 induced the apoptosis of pulp cells. These results suggest that NO regulates the growth, apoptosis, and mineralization of pulp cells.


Subject(s)
Dental Cavity Preparation/adverse effects , Dental Pulp/metabolism , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide/physiology , Alkaline Phosphatase/biosynthesis , Animals , Apoptosis , Cell Differentiation , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cells, Cultured , Dental Pulp/cytology , Dental Pulp/physiopathology , Enzyme Induction , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Donors/pharmacology , Nitroso Compounds/pharmacology , Phosphoric Diester Hydrolases/biosynthesis , Pulpitis/etiology , Pyrophosphatases/biosynthesis , Tooth Calcification
7.
Br J Cancer ; 86(7): 1150-6, 2002 Apr 08.
Article in English | MEDLINE | ID: mdl-11953864

ABSTRACT

Cyclooxygenase-2 is the rate-limiting enzyme in synthesis of prostaglandins and other eicosanoids. Prior reports have shown that inhibition of cyclooxygenase-2 activity, either by selective inhibitors or by antisense oligonucleotide, results in suppression of growth of squamous cell carcinoma cell lines which express high cyclooxygenase-2 levels, such as NA, a cell line established from a squamous cell carcinoma of the tongue. To investigate the mechanisms by which cyclooxygenase-2 inhibitors suppressed growth of these cells, the effects of NS-398, the selective cyclooxygenase-2 inhibitor, on cell-cycle distribution were examined. NS-398 induced G0/G1 cell-cycle arrest in NA cells which expressed cyclooxygenase-2. G0/G1 arrest induced by NS-398 was accompanied by up-regulation of cyclin-dependent kinase inhibitor p21, but not by up-regulation of the other cyclin-dependent kinase inhibitors. Transfection with p21 antisense oligonucleotide inhibited cell-cycle arrest induced by NS-398. Accumulation in G0/G1 was also observed in NA cells transfected with cyclooxygenase-2 antisense oligonucleotide. On the other hand, NS-398-treated NA cells showed a loss of plasma membrane asymmetry, a marker of early events in apoptosis. However, NS-398 did not induce other morphological and biochemical changes related to apoptotic cell death. These results suggest that cyclooxygenase-2 inhibitor induces G0/G1 cell-cycle arrest in NA cells by up-regulation of p21. Our results also suggest that NS-398 is not sufficient to complete the whole process of apoptosis in NA cells, although it induces an early event in apoptosis.


Subject(s)
Apoptosis/drug effects , Carcinoma, Squamous Cell/pathology , Cell Cycle/drug effects , Cyclins/biosynthesis , Cyclooxygenase Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Nitrobenzenes/pharmacology , Sulfonamides/pharmacology , Tongue Neoplasms/pathology , Blotting, Western , Cell Membrane/pathology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , DNA Primers , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/pharmacology , Membrane Proteins , Oligonucleotides, Antisense , Polymerase Chain Reaction , Prostaglandin-Endoperoxide Synthases/pharmacology , Prostaglandins/pharmacology , Transfection , Tumor Cells, Cultured , Up-Regulation
8.
Eur J Cancer ; 37(12): 1570-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11506967

ABSTRACT

Prostaglandins (PG) are known to play important roles in the proliferation and differentiation of leukaemia cells. The effect of the inhibitors of cyclooxygenase-2 (COX-2), a rate-limiting enzyme for the synthesis of PG, on the proliferation and differentiation of leukaemia cell lines was investigated. COX-2 inhibitors, NS-398 and nabumetone, suppressed the proliferation of U-937 and ML-1 cells by inducing a G0/G1 cell-cycle arrest. Cell-cycle arrest induced by these COX-2 inhibitors was not associated with an upregulation of the cyclin-dependent kinase inhibitors. COX-2 inhibitors also inhibited the differentiation of these cells induced by interferon-gamma (IFN-gamma), tumour necrosis factor-alpha (TNF-alpha) and retinoic acid (RA). Treatment with NS-398 did not suppress the levels of PGs produced by these cells. Although COX-2 antisense oligonucleotide showed a similar inhibitory effect on these cells, its inhibitory effect was smaller than that of NS-398. These results suggest that COX-2 inhibitors may suppress the proliferation and differentiation of leukaemia cells both via COX-2-dependent and -independent pathways.


Subject(s)
Antineoplastic Agents/therapeutic use , Butanones/therapeutic use , Cyclooxygenase Inhibitors/therapeutic use , Isoenzymes/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Nitrobenzenes/therapeutic use , Sulfonamides/therapeutic use , Blotting, Western , Cell Division/drug effects , Cell Transformation, Neoplastic/drug effects , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Leukemia, Myeloid, Acute/pathology , Membrane Proteins , Nabumetone , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured/drug effects , U937 Cells/drug effects
9.
J Oral Pathol Med ; 30(1): 41-7, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11140899

ABSTRACT

Prostaglandins (PGs) are known to play important roles in the proliferation of various types of cancer cells. PGs are produced by the action of cyclooxygenase (COX) enzymes, and two forms of COX, COX-1 and COX-2, have been described. Previous studies have demonstrated that overexpression of COX-2 is associated with colon carcinogenesis, tumor invasion and metastatic potential of colon cancer. In this study, the role of COX-2 on proliferation of squamous cell carcinoma cell lines was investigated. NS-398, a selective COX-2 inhibitor, inhibited proliferation of NA cells, a squamous cell caricinoma cell line that constitutively expresses COX-2 mRNA. NS-398 suppressed the spontaneous production of PGE2 by NA cells, and the antiproliferative effect of NS-398 was abolished by addition of PGE2. Similar results were obtained from experiments using COX-2 antisense oligonucleotide. These results suggest that specific inhibition of COX-2 inhibits proliferation of cancer cells expressing COX-2 mRNA via suppression of PGE2 production.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/biosynthesis , Isoenzymes/antagonists & inhibitors , Tongue Neoplasms/enzymology , Cell Division/drug effects , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Humans , Isoenzymes/metabolism , Membrane Proteins , Nitrobenzenes/pharmacology , Oligonucleotides, Antisense/pharmacology , Prostaglandin-Endoperoxide Synthases/metabolism , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/enzymology
10.
Anticancer Res ; 20(5A): 3117-21, 2000.
Article in English | MEDLINE | ID: mdl-11062731

ABSTRACT

Although many anticancer drugs have been reported to induce apoptosis in cancer cells, the underlying mechanism remains unclear (1-3). Recent studies have revealed that the caspase family of cysteine proteases have been shown to play an important role in the regulation of several apoptotic processes. Thus, the present study investigated whether apoptosis induced by anticancer drugs is mediated by the activation of caspase cascade. NA cells, a squamous cell carcinoma cell line, were exposed to cisplatin (CDDP) or 5-fluorouracil (5-FU) with or without inhibitors of caspase 1, 3 and 8. Analysis of DNA fragmentation revealed that caspase inhibitors consistently inhibited DNA fragmentation induced by 5-FU. During the early stages of apoptosis, phosphatidylserine (PS) is translocated from the inner side of the plasma membrane to the cell surface. This PS externalization was markedly inhibited by treatment with caspase-8 inhibitor. These findings suggested that 5-FU induced apoptosis was mediated by the activation of a caspase cascade involving caspase 1, 3 and 8.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Fluorouracil/pharmacology , Antineoplastic Agents/pharmacology , Bisbenzimidazole , Carcinoma, Squamous Cell , Caspase 1/metabolism , Caspase 3 , Caspase 8 , Caspase 9 , Caspase Inhibitors , Cell Nucleus , Cisplatin/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , DNA Fragmentation/drug effects , Flow Cytometry/methods , Fluorescent Dyes , Humans , Staining and Labeling/methods , Tongue Neoplasms , Tumor Cells, Cultured
12.
J Oral Pathol Med ; 28(8): 371-6, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10478963

ABSTRACT

P53 has important regulatory functions in cell growth, differentiation and apoptosis. Here we analyzed the effects of p53 on the growth response of oral mucosal keratinocytes (OMKCs) using p53-deficient (p53-/-) mice. No morphological difference was found between p53-/- and wild-type (p53+/+) oral mucosa. In a long-term culture, p53-/- OMKCs continued to proliferate past the point at which p53+/+ became senescent. The percentage of p53-/- OMKCs in the G0/G1 phase was lower than that of p53+/+ OMKCs. Proliferation of cultured OMKCs induced by epidermal growth factor (EGF) and interleukin-(IL)-1alpha was more strongly enhanced in p53-/- than in p53+/+ mice. Such an enhanced response was not due to increased mRNA expression of growth factor receptors. These data suggest that p53 acts as a modulator of G1 arrest in OMKCs and is also involved in the regulation of responses to EGF and IL-1alpha without affecting the expression of their receptors.


Subject(s)
Keratinocytes/cytology , Mouth Mucosa/cytology , Tumor Suppressor Protein p53/physiology , Animals , Blotting, Northern , Blotting, Western , Cell Cycle , Cell Division/drug effects , Cells, Cultured , Epidermal Growth Factor/pharmacology , ErbB Receptors/metabolism , Keratinocytes/metabolism , Mice , Mice, Knockout , Mouth Mucosa/abnormalities , Mouth Mucosa/metabolism , RNA, Messenger/metabolism , Receptors, Interleukin-1/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
13.
Br J Cancer ; 80(7): 954-63, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10362102

ABSTRACT

Cisplatin (CDDP) and 5-fluorouracil (5-FU) are common anti-tumour agents, and the anti-tumour effect of CDDP and 5-FU are synergistically enhanced by combined treatment. To clarify the mechanisms of this synergism, we examined the effect of CDDP and 5-FU on the expression of cell adhesion molecules involved in recognition of cancer cells by T lymphocytes. When NA cells, a squamous cell carcinoma cell line, were exposed to CDDP and 5-FU for 18 h, the expression of intercellular adhesion molecule-1 (ICAM-1) was synergistically induced, whereas CDDP or 5-FU alone did not induce the expression of ICAM-1, as determined by flow cytometry. Expression of ICAM-2 and ICAM-3, which are recognized by the same counter receptor on T-cells, were not up-regulated by CDDP and 5-FU. RT-PCR analysis showed that the induction of ICAM-1 on NA cells might be due to transcriptional induction of ICAM-1 mRNA. Treatment with genistein, a protein tyrosine kinase (PTK) inhibitor, inhibited the induction of ICAM-1 on NA cells by CDDP and 5-FU, whereas staurosporin, a protein kinase C inhibitor, did not. Although CDDP and 5-FU induced binding at the nuclear factor kappa B (NF-kappaB) site in the ICAM-1 promoter, pretreatment with genistein did not prevent CDDP and 5-FU-induced binding at the NF-kappaB site. Moreover, a NF-kappaB nuclear translocation inhibitor did not inhibit the induction of ICAM-1 expression by treatment with CDDP and 5-FU. The synergistic effect of CDDP and 5-FU was not specific to NA cells, since ICAM-1 was synergistically induced by CDDP and 5-FU on HSC-4 cells, a squamous cell carcinoma cell line. These findings indicate that treatment with CDDP and 5-FU induces ICAM-1 expression by a NF-kappaB independent regulatory mechanism involving PTK.


Subject(s)
Antigens, Differentiation , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cisplatin/pharmacology , Fluorouracil/pharmacology , Intercellular Adhesion Molecule-1/biosynthesis , NF-kappa B/metabolism , Antigens, CD/biosynthesis , Cell Adhesion Molecules/biosynthesis , Cytokines/metabolism , DNA/metabolism , Drug Synergism , E-Selectin/biosynthesis , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , NF-kappa B/antagonists & inhibitors , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Vascular Cell Adhesion Molecule-1/biosynthesis
14.
Biochem Biophys Res Commun ; 257(2): 395-9, 1999 Apr 13.
Article in English | MEDLINE | ID: mdl-10198224

ABSTRACT

Nitric oxide (NO) is an unstable free radical that functions as a cytotoxic agent secreted by macrophages to kill cancer cells. Here we report the effect of NO on the expression of intercellular adhesion molecule-1 (ICAM-1) on cancer cells. NO donors such as SNP, SNAP and SIN-1 up-regulated the expression of ICAM-1 on NA cells, a squamous cell carcinoma cell line. Northern blot analysis showed that the induction of ICAM-1 might be due to transcriptional induction of ICAM-1 mRNA. Up-regulation of ICAM-1 mRNA by NO donors was inhibited by carboxy-PTIO, a NO scavenger. Although NF-kappaB activity was induced by NO donors, AP-1 was not induced by them. Staurosporin, a protein kinase C (PKC) inhibitor, inhibited the induction of ICAM-1 on NA cells by NO, whereas genistein, a protein tyrosine kinase inhibitor, did not. These findings indicate that NO up-regulates ICAM-1 expression on cancer cells by a regulatory mechanism involving PKC and suggest that NF-kappaB, but not AP-1, might be involved in induction of ICAM-1 by NO in cancer cells.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Intercellular Adhesion Molecule-1/metabolism , Nitric Oxide/pharmacology , Up-Regulation/drug effects , Benzoates/pharmacology , Carcinoma, Squamous Cell/pathology , Cell Adhesion Molecules/metabolism , Free Radical Scavengers/pharmacology , Genistein/pharmacology , Humans , Imidazoles/pharmacology , Intercellular Adhesion Molecule-1/genetics , Molsidomine/analogs & derivatives , Molsidomine/pharmacology , NF-kappa B/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , RNA, Messenger/metabolism , S-Nitroso-N-Acetylpenicillamine , Staurosporine/pharmacology , Tongue Neoplasms , Transcription Factor AP-1/metabolism , Tumor Cells, Cultured
15.
Anticancer Res ; 17(2A): 865-71, 1997.
Article in English | MEDLINE | ID: mdl-9137419

ABSTRACT

Nitric oxide (NO) is an unstable free radical gas known as an effector molecule of macrophage cytotoxicity against cancer cells. Although several mechanisms of NO-mediated cytotoxicity have been proposed, this phenomenon remains to be characterized in detail. To explore the mechanisms by which NO kills cancer cells, we made use of sodium nitroprusside (SNP), which releases NO in the culture medium. SNP showed a dose-dependent cytotoxic effect on NA cells, an epithelial cancer cell line. When NA cells were killed by SNP, high levels of NO2- (stable end product of NO) were detected in the culture medium. The cell death induced by SNP was mediated by apoptosis, as demonstrated by the presence of nuclear condensation and blebbing of the nuclear membrane, and internucleosomal DNA fragmentation quantified by a specific ELISA. Northern blot analysis revealed that c-myc mRNA expression of NA cells was rapidly reduced by treatment with SNP. RT-PCR analysis showed that c-myb mRNA was expressed in untreated NA cells, and c-myb mRNA level of NA cells was dose-dependently reduced by treatment with SNP. These results indicate that SNP exerts its cytotoxic effect on NA cells through spontaneous release of NO. Cytotoxicity induced by SNP is at least partially mediated via the process known as apoptosis. Our results also suggest that down-regulation of c-myc and c-myb proto-oncogenes might be involved in SNP-induced cytotoxicity.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Genes, myc , Nitroprusside/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Trans-Activators/genetics , DNA Fragmentation , Down-Regulation , Humans , Nitric Oxide/physiology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myb , RNA, Messenger/analysis , Tumor Cells, Cultured
16.
J Inflamm ; 46(1): 23-31, 1995.
Article in English | MEDLINE | ID: mdl-8832969

ABSTRACT

Mice with a targeted deletion of either the interferon (IFN)-gamma gene or the IFN-gamma receptor gene (IFN-gamma R(0/0) mice) fail to survive infection with the Bacillus Calmette-Guerin (BCG) strain of Mycobacterium bovis. Here we show that resident peritoneal macrophages isolated 2 weeks after BCG infection from IFN-gamma R(0/0) mice produced significantly less nitric oxide (NO) than wild-type macrophages. However, the response to lipopolysaccharide (LPS) was not completely abrogated in the IFN-gamma R(0/0) macrophages. BCG infection of wild-type mice led to a marked increase in their urinary nitrite/nitrate levels, as previously described. This increase in urinary nitrite/nitrate was not detected in BCG- infected IFN-gamma R(0/0) mice, indicating that no other cytokine can replace IFN-gamma as a mediator of increased NO synthesis after BCG infection in the intact organism. A comparison of circulating levels of IFN-gamma in BCG-infected animals revealed that sera from IFN-gamma R(0/0) mice contained up to 66-fold more IFN-gamma than sera from identically treated wild-type mice. To determine if the higher levels of circulating IFN-gamma were due to increased IFN-gamma synthesis, we compared the amounts of IFN-gamma mRNA present in the spleens of BCG-infected wild-type and IFN-gamma R(0/0) mice. No increase in IFN-gamma mRNA levels was detected in the spleens from IFN-gamma R(0/0) mice. Since the generation of IFN-gamma protein in cultured spleen cells was also not increased in IFN-gamma R(0/0) mice, we conclude that clearance of IFN-gamma from the circulation is impaired in IFN-gamma R(0/0) mice, thus revealing a heretofore unrecognized important role for the IFN-gamma receptor in the regulation of IFN-gamma levels in the intact organism.


Subject(s)
Interferon-gamma/metabolism , Mycobacterium bovis , Nitric Oxide/metabolism , Receptors, Interferon/deficiency , Tuberculosis/metabolism , Animals , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Nitrates/urine , Nitrites/urine , Receptors, Interferon/genetics , Spleen/metabolism , Interferon gamma Receptor
17.
Hokkaido Igaku Zasshi ; 69(6): 1332-8, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7705746

ABSTRACT

Mice with a targeted disruption in the IFN-gamma receptor gene (IFN-gamma R0/0) provided a useful model to ask to what extent other cytokines could replace IFN-gamma in macrophage activation. In thioglycollate-elicited peritoneal macrophages from wild-typy (WT) mice, TNF enhanced nitric oxide (NO) release in the presence of IFN-gamma, though TNF alone was not effective. In macrophages from IFN-gamma R0/0 mice, which are not responsive to IFN-gamma, TNF completely failed to stimulate NO release. The NO inducing effects of IFN-alpha/beta were indistinguishable in IFN-gamma R0/0 and WT macrophages. The important role of IFN-gamma in the regulation of the induced expression of MHC class II antigen (Ia) was confirmed by showing that after systemic infection with the BCG strain of Mycobacterium bovis, peritoneal macrophages from IFN-gamma R0/0 mice had a lower level of Ia expression than macrophages from WT mice. BCG infection was not lethal for WT mice whereas all IFN-gamma R0/0 mice died 7-9 weeks after infection. It is well known that BCG infection greatly sensitizes mice to lethal action of LPS. Injection of LPS 2 weeks after BCG inoculation was significantly less lethal for IFN-gamma R0/0 mice than for WT mice. Reduced lethality of LPS correlated with a drastically reduced TNF-alpha production in the IFN-gamma R0/0 mice after BCG infection and LPS challenge. The greatly reduced ability of BCG-infected IFN-gamma R0/0 mice to produce TNF-alpha may be an important factor in their inability to resist BCG infection.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Interferon-alpha/physiology , Interferon-beta/physiology , Interferon-gamma/physiology , Mice, Knockout/genetics , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Gene Deletion , Gene Targeting , Histocompatibility Antigens Class II/metabolism , Interferon Regulatory Factor-1 , Macrophage Activation , Mice , Mycobacterium bovis , Nitric Oxide/metabolism , Phosphoproteins/genetics , Phosphoproteins/physiology , Receptors, Interferon/genetics , Interferon gamma Receptor
19.
Science ; 264(5167): 1921-4, 1994 Jun 24.
Article in English | MEDLINE | ID: mdl-8009222

ABSTRACT

The mechanisms underlying interferon (IFN)-induced antiviral states are not well understood. Interferon regulatory factor-1 (IRF-1) is an IFN-inducible transcriptional activator, whereas IRF-2 suppresses IRF-1 action. The inhibition of encephalomyocarditis virus (EMCV) replication by IFN-alpha and especially by IFN-gamma was impaired in cells from mice with a null mutation in the IRF-1 gene (IRF-1-/- mice). The IRF-1-/- mice were less resistant than normal mice to EMCV infection, as revealed by accelerated mortality and a larger virus titer in target organs. The absence of IRF-1 did not clearly affect replication of two other types of viruses. Thus, IRF-1 is necessary for the antiviral action of IFNs against some viruses, but IFNs activate multiple activation pathways through diverse target genes to induce the antiviral state.


Subject(s)
Cardiovirus Infections/immunology , DNA-Binding Proteins/physiology , Interferon-alpha/pharmacology , Interferon-gamma/pharmacology , Phosphoproteins/physiology , Transcription Factors/physiology , Virus Replication , Animals , Cardiovirus Infections/microbiology , Cells, Cultured , DNA-Binding Proteins/genetics , Encephalomyocarditis virus/physiology , Gene Expression Regulation , Interferon Regulatory Factor-1 , Mice , Mutation , Phosphoproteins/genetics , Simplexvirus/physiology , Transcription Factors/genetics , Vesicular stomatitis Indiana virus/physiology
20.
J Interferon Res ; 14(2): 49-55, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8077765

ABSTRACT

The modulation of cell growth by tumor necrosis factor-alpha (TNF-alpha), or TNF-alpha in combination with transforming growth factor-beta (TGF-beta) or interferon-gamma (IFN-gamma) was investigated. TNF-alpha inhibited the proliferation of U937 cells, a monocytic leukemic cell line, and of NA cells that were established from oral squamous cell carcinoma. TNF-alpha showed a cytolytic effect on NA cells in the presence of actinomycin D. TNF-alpha in combination with TGF-beta and TNF-alpha combined with INF-gamma synergistically inhibited the cell proliferation of U937 and NA cells, respectively. TNF-alpha dose-dependently reduced c-myc mRNA expression of U937 and NA cells within 1 h. The combination of TNF-alpha and TGF-beta in U937 cells and that of TNF-alpha and IFN-gamma in NA cells cooperatively reduced the expression of c-myc mRNA. TNF-alpha had little or no effect on the half-life of c-myc mRNA, indicating that c-myc mRNA expression was reduced at transcriptional level. Cycloheximide did not mediate the inhibition of c-myc gene expression, suggesting that the TNF-alpha action was independent of de novo protein synthesis. These data suggest that the reduction of c-myc gene at transcriptional level by TNF-alpha or TNF-alpha in combination with TGF-beta or IFN-gamma plays a primary role in the inhibition of cell growth at an early stage.


Subject(s)
Interferon-gamma/immunology , Monocytes/immunology , Proto-Oncogene Proteins c-myc/biosynthesis , Transforming Growth Factor beta/immunology , Tumor Necrosis Factor-alpha/immunology , Cell Division/immunology , Cycloheximide/pharmacology , Dose-Response Relationship, Immunologic , Down-Regulation/drug effects , Down-Regulation/immunology , Humans , RNA, Messenger , Recombinant Proteins , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...