Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Article in English | MEDLINE | ID: mdl-34299904

ABSTRACT

In April 2012, perioperative oral management (POM) was approved for inclusion in the national health insurance system of Japan to prevent the occurrence of pneumonia, a major complication in cancer patients. The subsequent decrease in the incidence of postoperative pneumonia indicated the prophylactic effect of POM. The constant increase in health expenditure necessitates a cost-effectiveness analysis. In addition, the effect of reducing healthcare costs owing to health technologies must be evaluated. In the present multi-institutional study, the cost-effectiveness analysis of POM was conducted by comparing the incidence of postoperative pneumonia and the healthcare costs between patients who received surgery for malignant tumors before (n = 11,886) and after (n = 13,668) the introduction of POM. Additionally, the effect of reducing healthcare costs was evaluated. Reductions in the number of patients who developed pneumonia, duration of hospitalization, and number of deaths were observed after the introduction of POM. The incremental cost-effectiveness ratio was 111,927 yen, hence the prevention of postoperative pneumonia needs 111,927 yen per patient in healthcare costs. Consequently, a maximum reduction of 250,368,129 yen in healthcare costs was observed between the incremental costs for pneumonia treatment and the cost of POM. These findings indicate that improvements in cost-effectiveness can be expected in the future through the development of procedure and system for POM.


Subject(s)
Neoplasms , Pneumonia , Administration, Oral , Cost-Benefit Analysis , Health Care Costs , Humans , Pneumonia/epidemiology , Pneumonia/prevention & control
2.
Clin Exp Dent Res ; 6(2): 165-173, 2020 04.
Article in English | MEDLINE | ID: mdl-32250567

ABSTRACT

AIM: We conducted a multicenter study to explore the risk factors of developing pneumonia and the effectiveness of perioperative oral management (POM) for the prevention of pneumonia in postsurgical patients. METHODS AND RESULTS: A survey covering eight regional hospitals was conducted over 4 years, from April 2010 to March 2014. Using the Diagnosis Procedure Combination database, a target group of 25,554 patients with cancer who underwent surgery was selected and assessed from a population of 346,563 patients without pneumonia on admission (sample population). The study compared the incidence of pneumonia and attempted to identify the significant predictive factors for its occurrence in these patients using multiple logistic regression analysis. Comparative assessment for the occurrence of pneumonia before and after POM implementation showed a significant incidence decrease after POM introduction in the target group, with no such change observed in the sample population. Multiple logistic regression analysis showed that the odds ratio for pneumonia occurrence after POM introduction was 0.44, indicating a reduced risk of pneumonia. CONCLUSION: POM in cancer patients was indeed effective in reducing the incidence of pneumonia in hospitals and thereby helped in preventing pneumonia during hospitalization.


Subject(s)
Dental Care/methods , Healthcare-Associated Pneumonia/epidemiology , Neoplasms/surgery , Perioperative Care/methods , Postoperative Complications/epidemiology , Adult , Aged , Aged, 80 and over , Female , Healthcare-Associated Pneumonia/etiology , Healthcare-Associated Pneumonia/prevention & control , Humans , Incidence , Japan/epidemiology , Male , Middle Aged , Oral Hygiene , Postoperative Complications/etiology , Postoperative Complications/prevention & control , Retrospective Studies , Risk Factors , Treatment Outcome
3.
Oral Oncol ; 45(9): 766-70, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19157955

ABSTRACT

It has been reported recently that histone deacetylase inhibitors (HDACIs) can block the growth of a variety of malignant tumor cells by reversing the silencing of the tumor suppressor genes; these will be the anticancer agents of the next generation. In this study, we evaluated the antitumor effects of the HDACI suberoylanilide hydroxamic acid (SAHA) on oral squamous cell carcinoma (OSCC) and investigated its molecular mechanism. SAHA suppressed the in vitro proliferation of OSCC cell lines in a dose- and time-dependent manner. Flow cytometric analyses showed that treatment with SAHA led to G1 phase cell-cycle arrest of OSCC cells, accompanying a decrease in the percentage of S-phase cells. Western blot analyses demonstrated that the expression of p21 protein was remarkably augmented and hyperacetylation of p53 was induced after SAHA treatment. These results suggest that administration of SAHA suppresses OSCC growth through G1 phase arrest. Additionally, we observed that the growth of xenograft SAS tumors in nude mice was significantly blocked by the administration of SAHA without major adverse effects.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Hydroxamic Acids/pharmacology , Mouth Neoplasms/drug therapy , Animals , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Drug Screening Assays, Antitumor , Female , G1 Phase/drug effects , Humans , Mice , Mice, Nude , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , S Phase/drug effects , Tumor Suppressor Protein p53/metabolism , Vorinostat
4.
Oral Oncol ; 44(4): 361-8, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17689285

ABSTRACT

Molecular inhibition of epidermal growth factor receptor (EGFR) signaling is a promising cancer treatment strategy. We examined whether inhibition of EGFR signaling would affect the susceptibility of oral squamous cell carcinoma (OSCC) cells to Fas-mediated apoptosis. Treatment of OSCC cells with an anti-EGFR monoclonal antibody, C225, and an EGFR tyrosine kinase inhibitor, AG1478, which target the extracellular and intracellular domains of the receptor, respectively, inhibited phosphorylation of EGFR and its downstream effector molecule Akt and amplified the induction of Fas-mediated apoptosis. In OSCC cells treated with EGFR inhibitors, Fas-mediated apoptosis was accompanied by caspase-8 activation but not Bid cleavage. Caspase-3 and -8 inhibitors reduced the effect of EGFR inhibitors on Fas-mediated apoptosis in OSCC cells, but a caspase-9 inhibitor did not. These results indicate that the pro-apoptotic activity of EGFR inhibitors in OSCC cells depends on the extrinsic pathway of the caspase cascade. Although EGFR inhibitors did not affect the expression of Fas, the Fas-associated death domain protein, or procaspase-8 in OSCC cells, the inhibition downregulated cellular FLICE-inhibitory protein (c-FLIP). Moreover, knockdown of c-FLIP in HSC-2 cells with a small interfering RNA strongly enhanced Fas-mediated apoptosis. These results suggest that the EGFR signaling pathway may, in part, regulate Fas-mediated apoptosis in OSCC cells through c-FLIP expression.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Squamous Cell/pathology , ErbB Receptors/antagonists & inhibitors , Mouth Neoplasms/pathology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Carcinoma, Squamous Cell/metabolism , Caspase 8/metabolism , Cetuximab , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Fas-Associated Death Domain Protein/metabolism , Humans , Mouth Neoplasms/metabolism , Neoplasm Proteins/metabolism , Phosphorylation/drug effects , Protein Tyrosine Phosphatases/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines , Tumor Cells, Cultured , Tyrphostins/pharmacology , fas Receptor/metabolism , fas Receptor/physiology
5.
Int J Oncol ; 30(6): 1469-76, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17487368

ABSTRACT

Epidermal growth factor (EGF) is known to be involved in the proliferation and metastasis of squamous cell carcinoma (SCC), suggesting that the EGF receptor (EGFR) must also contribute to SCC development. In combination with conventional anti-cancer drugs, agents that block EGFR may represent an efficient means of inhibiting proliferation and inducing apoptosis in SCC cells. We investigated the effects of combining an anti-EGFR monoclonal antibody (C225) or an EGFR-selective tyrosine kinase inhibitor (AG1478) with the conventional anti-cancer drug cisplatin on the oral SCC (OSCC) cell lines NA and Ca9-22. We detected constitutive expression of EGFR on the cell membranes of both cell lines. OSCC cell proliferation was inhibited by C225, AG1478 and cisplatin in a dose-dependent manner. The combination of C225 or AG1478 with cisplatin at concentrations

Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cisplatin/pharmacology , Mouth Neoplasms/drug therapy , Tyrphostins/pharmacology , Antibodies, Monoclonal, Humanized , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cetuximab , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , Humans , Inhibitor of Apoptosis Proteins/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-bcl-2/drug effects , Quinazolines
6.
Int J Oncol ; 30(5): 1163-71, 2007 May.
Article in English | MEDLINE | ID: mdl-17390018

ABSTRACT

In general, oral squamous cell carcinoma (OSCC) cells are relatively resistant to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis during culture in vitro. Here, we studied the role of phosphatidylinositol 3-kinase (PI 3-K)/Akt in survival and apoptosis of these cells. The PI 3-K inhibitors wortmannin and LY294002 markedly suppressed phosphorylation of Akt and accelerated TRAIL-mediated apoptosis in OSCC cells. Addition of TRAIL to PI 3-K inhibitor-treated cells resulted in caspase-8 activation and loss of mitochondrial membrane potential. Furthermore, inhibitors of caspase-3, -8 and -9 reduced the accelerative effect of PI 3-K inhibitors on TRAIL-mediated apoptosis. These results suggest that the pro-apoptotic effect of PI 3-K inhibitors on TRAIL-mediated apoptosis may contribute to both the extrinsic and intrinsic pathways. Although PI 3-K inhibitors did not affect expression of the TRAIL receptors DR4 and DR5, we observed a marked reduction in expression of cellular FLICE-inhibitory protein (c-FLIP), Bcl-2, cellular inhibitor of apoptosis protein-1 (cIAP-1) and X-linked IAP (XIAP), whereas Bax was up-regulated and no significant difference was observed in expression of Bcl-xL, Bak or cIAP-2. Therefore, the PI 3-K/Akt signaling pathway provides partial regulation of TRAIL-mediated apoptosis in OSCC cells via modulation of c-FLIP, Bcl-2, Bax, cIAP-1 and XIAP expression. These results suggest that PI 3-K inhibitors may represent a novel strategy for overcoming resistance to TRAIL-mediated apoptosis in OSCC cells.


Subject(s)
Apoptosis , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Phosphoinositide-3 Kinase Inhibitors , TNF-Related Apoptosis-Inducing Ligand/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , DNA Fragmentation , Humans , Ligands , Membrane Potentials , Mitochondria/pathology , Mouth Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism
7.
Oral Oncol ; 42(7): 745-52, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16527514

ABSTRACT

In general, oral squamous cell carcinoma (OSCC) cells are relatively resistant to Fas-mediated apoptosis during in vitro culture. Here, we studied the role of survival/apoptosis associated phosphatidylinositol 3-kinase (PI 3-K)/Akt in this process. We found that both PI 3-K inhibitors, wortmannin and LY294002, markedly suppressed the phosphorylation of Akt and accelerated Fas-mediated apoptosis in OSCC cells. It was found that caspase-3 and -8 inhibitors reduced the accelerative effect of PI 3-K inhibitor on Fas-mediated apoptosis in OSCC cells, but not caspase-9 inhibitor. Although PI 3-K inhibitors did not affect the Fas expression of OSCC cells, cellular FLICE-inhibitory protein (c-FLIP) levels were markedly reduced by PI 3-K inhibitor treatment. Moreover, antisense oligonucleotide to c-FLIP confirmed that the down-regulation of c-FLIP enhanced the sensitization to Fas-mediated apoptosis in OSCC cells. These results suggest that PI 3-K/Akt signaling pathway may, in part, regulate Fas-mediated apoptosis in OSCC cells through c-FLIP expression.


Subject(s)
Apoptosis/drug effects , Carcinoma, Squamous Cell/pathology , Enzyme Inhibitors/pharmacology , Mouth Neoplasms/pathology , Phosphoinositide-3 Kinase Inhibitors , fas Receptor/metabolism , Androstadienes/pharmacology , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Carcinoma, Squamous Cell/metabolism , Chromones/pharmacology , Down-Regulation , Flow Cytometry , Humans , Morpholines/pharmacology , Mouth Neoplasms/metabolism , Oncogene Protein v-akt/metabolism , Phosphorylation , Signal Transduction/drug effects , Tumor Cells, Cultured , Wortmannin
8.
Int Immunopharmacol ; 6(3): 334-41, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16428068

ABSTRACT

Inflammatory stimuli, such as cytokines, can induce cyclooxygenase-2 (COX-2) expression in neutrophils. Selective, anti-inflammatory COX-2 inhibitors have been developed for patients with acute inflammatory diseases. Recent work has shown that selective COX-2 inhibitors interfere with tumor cell growth. The purpose of this study was to examine the capability of selective COX-2 inhibitors on Fas-mediated apoptosis in cytokine-stimulated neutrophils. Tumor necrosis factor-alpha (TNF-alpha) and granulocyte-macrophage colony-stimulating factor (GM-CSF) enhanced prostaglandin E2 (PGE2) release through the induction of COX-2 in neutrophils. This effect was not seen with either interleukin (IL)-1beta or IL-8. TNF-alpha-and GM-CSF-induced PGE2 release was blocked by the addition of the selective COX-2 inhibitor, N-(2-cyclohexyloxy-4-nitrophenyl)-methanesulfonamide (NS-398; 1 microM). GM-CSF, IL-1beta and IL-8 suppressed Fas-mediated apoptosis in neutrophils; however, this effect was not seen with TNF-alpha. The anti-apoptotic effect of cytokines on Fas-mediated neutrophil apoptosis was attenuated by the addition of NS-398 (100 microM). These results suggest that NS-398 operates via two distinct mechanisms for regulating apoptosis and COX-2 activation in neutrophils. This distinction is indicated by the difference in concentration of NS-398 required for acceleration of Fas-mediated neutrophil apoptosis, and the inhibition of PGE2 synthesis. Moreover, NS-398 suppressed the anti-apoptotic activity of IL-8 and IL-1beta, but did not induce COX-2; therefore, the pro-apoptotic mechanism of the selective COX-2 inhibitor may be unrelated to COX-2 activity. Thus, a selective COX-2 inhibitor may contribute to the reduction of acute inflammation through the enhancement of neutrophil apoptosis.


Subject(s)
Apoptosis/drug effects , Cyclooxygenase 2 Inhibitors/pharmacology , Neutrophils/drug effects , Nitrobenzenes/pharmacology , Sulfonamides/pharmacology , fas Receptor/physiology , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/genetics , Cytokines/physiology , Dinoprostone/metabolism , Enzyme Induction/drug effects , Humans , Neutrophils/enzymology , RNA, Messenger/metabolism
9.
J Surg Res ; 134(1): 114-23, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16376940

ABSTRACT

BACKGROUND: Neutrophils undergo rapid Fas-mediated apoptosis during in vitro culture. The purpose of this study was to investigate the effects of surgical stress upon the Fas-mediated apoptotic response in circulating neutrophils. MATERIALS AND METHODS: Blood samples were drawn from eight patients with a mandibular prognathism, and who had undergone a bilateral sagittal split ramus osteotomy, at 2 days before, and at 1 and 5 days after surgery. The circulating neutrophils in each blood sample were then evaluated for their susceptibility to Fas-mediated apoptosis in either the presence or the absence of autogenous plasma. RESULTS: Fas-induced apoptosis in the neutrophils of these surgically treated patients was found to be slightly accelerated at 1 day postoperatively in the presence of FBS, compared with 2 days preoperatively and 5 days postoperatively. However, we obtained different results for these experiments in the presence of autogenous plasma. The Fas-induced apoptotic response levels in the neutrophils at day 1 postsurgery following exposure to autogenous plasma were significantly suppressed compared with the levels at both 2 days preoperatively and 5 days postoperatively. The Fas expression levels on the cell surface of the neutrophils were not altered, but the levels of soluble Fas (sFas) in the plasma were reduced to almost inverse levels during the postoperative periods. The levels of granulocyte-macrophage colony-stimulating factor, interleukin-6, and interleukin-8 levels in the plasma were also markedly raised in the plasma from each of these patients at 1 day postoperatively. However, the anti-apoptotic effects of the plasma on the Fas-mediated neutrophil apoptosis were not influenced by the addition of their neutralizing antibodies for these cytokines. The suppressive effects of postoperative plasma on Fas-mediated neutrophil apoptosis were blocked by the phosphatidylinositol 3-kinase (PI 3-K) inhibitors, LY294002, and wortmannin. Additionally, these effects were also abrogated by the extracellular signal-regulated kinase (ERK) inhibitor, PD98059, but not by the p38 mitogen-activated protein kinase inhibitor, SB203580. CONCLUSIONS: The increase in sFas levels in the plasma of patients with acute inflammation may lead to the inhibition of Fas-mediated neutrophil apoptosis. Moreover, the activation of the PI 3-K and ERK signaling-dependent pathways may, in part, also contribute to the down-regulation of the Fas-mediated apoptotic response in neutrophils.


Subject(s)
Apoptosis , Inflammation/blood , Neutrophils/physiology , Surgical Procedures, Operative , fas Receptor/physiology , Acute Disease , Adult , C-Reactive Protein/analysis , Cytokines/blood , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Leukocyte Count , Orthognathic Surgical Procedures , Osteotomy , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology
10.
J Surg Res ; 130(1): 110-8, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16289597

ABSTRACT

BACKGROUND: Human neutrophils undergo rapid apoptosis during in vitro culture. The aim of this study was to investigate the role of interleukin-8 (IL-8) on neutrophil apoptosis in surgery-induced inflammation. MATERIALS AND METHODS: Blood samples were drawn from 21 patients with mandibular prognathism 2 days before, and 1 and 5 days after orthognathic surgery. The IL-8 levels in the separated plasma were measured using an ELISA kit. The expression of two receptors for IL-8, CXCR1, and CXCR2, and their role in neutrophil apoptosis was evaluated using a flow cytometer. RESULTS: The IL-8 levels in the plasma were correlated with acute inflammatory markers, such as peripheral blood neutrophil counts and C-reactive protein levels. Both IL-8 receptors were markedly raised in patient-derived neutrophils 1 day post-operatively. Recombinant IL-8 (0-100 ng/ml) suppressed apoptosis in fresh-isolated neutrophils from healthy donors dose-dependently. Neutrophil apoptosis 1 day post-operatively was slightly accelerated in the presence of fetal bovine serum compared to the value 2 days pre-operatively and 5 days post-operatively. In contrast, in the presence of autogenous plasma, neutrophil apoptosis was significantly suppressed 1 day post-operatively compared to the value 2 days pre-operatively and 5 days post-operatively. Moreover, the anti-apoptotic effect of plasma on neutrophil apoptosis was partially decreased by the addition of anti-IL-8 neutralizing antibody. CONCLUSIONS: These results suggest that circulating neutrophils are susceptible to augmentation by IL-8 through the reinforcement of IL-8 receptors in acute inflammatory conditions. Furthermore, IL-8 may, in part, contribute to the regulation of neutrophil survival during the inflammatory response.


Subject(s)
Apoptosis/immunology , Interleukin-8/immunology , Neutrophils/cytology , Oral Surgical Procedures , Adult , Antibodies/immunology , Apoptosis/drug effects , C-Reactive Protein/metabolism , Flow Cytometry , Humans , Interleukin-8/blood , Interleukin-8/pharmacology , Leukocyte Count , Neutrophils/drug effects , Neutrophils/metabolism , Postoperative Period , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/metabolism , Stress, Physiological/immunology
11.
Oncol Rep ; 14(5): 1215-22, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16211288

ABSTRACT

Promyelocytic leukemia HL-60 cells are resistant to Fas-mediated apoptosis. The signaling pathway for Fas-mediated apoptosis in various cells, including HL-60 cells, is currently unknown. Here, we studied the role of survival/apoptosis associated phosphatidylinositol 3-kinase (PI 3-K)/Akt in this process. We found that both PI 3-K inhibitors, wortmannin and LY294002, markedly suppressed phosphorylation of Akt and Bad in HL-60 cells. PI 3-K inhibitors significantly accelerated not only spontaneous apoptosis, but also Fas-induced apoptosis in HL-60 cells. The pro-apoptotic effect of PI 3-K inhibitors favored Fas-mediated apoptosis rather than spontaneous apoptosis in HL-60 cells. The caspase-3 or -8 inhibitor reduced the pro-apoptotic effect of the PI 3-K inhibitors for Fas-mediated apoptosis in HL-60 cells, but the caspase-9 inhibitor did not. Although PI 3-K inhibitors did not affect Fas expression in HL-60 cells, cellular FLICE-inhibitory protein (c-FLIP) levels were markedly reduced by PI 3-K inhibitor treatment. Furthermore, antisense oligonucleotide of c-FLIP confirmed that down-regulation of c-FLIP enhanced sensitization to Fas-mediated apoptosis in HL-60 cells. These results suggest that the PI 3-K/Akt signaling pathway may, in part, regulate Fas-mediated apoptosis in HL-60 cells through c-FLIP expression.


Subject(s)
Apoptosis/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Phosphatidylinositol 3-Kinases/physiology , Apoptosis/physiology , CASP8 and FADD-Like Apoptosis Regulating Protein , Caspase Inhibitors , Cell Survival , Down-Regulation , Enzyme Inhibitors/pharmacology , HL-60 Cells , Humans , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , Signal Transduction , fas Receptor/physiology
12.
Int Immunopharmacol ; 3(10-11): 1519-28, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12946449

ABSTRACT

To clarify whether a selective cyclooxygenase-2 (COX-2) inhibitor can affect various functions in human peripheral blood neutrophils. For this purpose, the effects of selective COX-2 inhibitors, NS-398 and nimesulide, on the expression of COX-2, PGE2 release and respiratory burst, degranulation and cytokine release in activated neutrophils were examined. Peripheral blood neutrophils were stimulated with formyl-methionyl-leucyl-phenylalanine (FMLP; 100 nM) or opsonized zymosan (OZ; 200 microg/ml). Then, the expression of COX-2 at protein and mRNA levels was detected by Western blot analysis and RT-PCR. The concentration of prostaglandin E2 (PGE2) and cytokines in the culture supernatant of neutrophils was determined using ELISA. Superoxide generation was measured by the cytochrome c reduction method. Elastase activity was measured using a chromogenic substrate assay specific for human neutrophil elastase. FMLP and OZ enhanced PGE2 release through induction of COX-2 protein and mRNA expression. FMLP- or OZ-induced PGE2 release was abolished by the addition of NS-398 or nimesulide; nevertheless, even a high concentration of COX-2 inhibitor did not change FMLP- or OZ-induced expression of COX-2 at message and protein levels. Although FMLP- or OZ-induced superoxide generation and elastase release were not affected by the addition of COX-2 inhibitor, cytokine release such as interleukin (IL)-1beta, IL-6 and IL-8 was significantly inhibited by high concentration of COX-2 inhibitor, but tumor necrosis factor-alpha (TNF-alpha) was partially attenuated. These studies showed that selective COX-2 inhibitors, NS-398 and nimesulide, suppressed PGE2 and proinflammatory cytokine release in activated neutrophils. These results suggest that selective COX-2 inhibitors may contribute to resolution of acute inflammation through the reduction of inflammatory cytokine release in activated neutrophils.


Subject(s)
Cyclooxygenase Inhibitors/pharmacology , Cytokines/antagonists & inhibitors , Isoenzymes/antagonists & inhibitors , Neutrophils , Adult , Cells, Cultured , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Dinoprostone/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Isoenzymes/biosynthesis , Membrane Proteins , Neutrophils/enzymology , Neutrophils/metabolism , Nitrobenzenes/pharmacology , Prostaglandin-Endoperoxide Synthases/biosynthesis , Sulfonamides/pharmacology , Superoxides/antagonists & inhibitors
13.
Int J Cancer ; 106(4): 619-625, 2003 Sep 10.
Article in English | MEDLINE | ID: mdl-12845662

ABSTRACT

Our study was conducted to investigate whether anticancer drugs, cisplatin (CDDP) and/or 5-fluorouracil (5-FU), can modulate Fas-mediated apoptosis in oral squamous cell carcinoma (OSCC) cell lines. When OSCC cell lines, NA and HSC-4, were treated with CDDP and/or 5-FU, Fas and its mRNA expression on the plasma membrane were enhanced. An increase in caspase-3 and -8 activities was then observed by the addition of agonistic anti-Fas antibody, CH-11. Apoptosis of OSCC cells treated with anticancer drugs were significantly enhanced by CH-11, whereas untreated cells were nearly resistant to apoptosis. Moreover, the combination of CDDP and 5-FU resulted in an increasing susceptibility to apoptosis. Caspase-3 and -8 inhibitors, but not caspase-9 inhibitor, reduced Fas-mediated apoptosis enhanced by the anticancer drugs. Furthermore, OSCC cells treated with anticancer drugs exhibited decreased cellular FADD-like interleukin 1-converting enzyme-inhibitory protein (c-FLIP) levels, whereas neither the Fas-associated death domain-containing protein (FADD) nor procaspase-8 changed the expression. Moreover, antisense oligonucleotide to c-FLIP confirmed that down-regulation of c-FLIP induced sensitization to Fas-mediated apoptosis. These results suggest that CDDP and 5-FU may enhance the susceptibility to Fas-mediated apoptosis through down-regulation of c-FLIP. From these findings, a new potential strategy may be developed to improve the efficacy of anticancer drugs.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Arabidopsis Proteins , Carcinoma, Squamous Cell/pathology , Cisplatin/pharmacology , Fluorouracil/pharmacology , Intracellular Signaling Peptides and Proteins , Mouth Neoplasms/pathology , fas Receptor/metabolism , Annexin A5/metabolism , Blotting, Western , CASP8 and FADD-Like Apoptosis Regulating Protein , Carcinoma, Squamous Cell/metabolism , Carrier Proteins/metabolism , Caspase Inhibitors , Caspases/metabolism , DNA Primers/chemistry , Down-Regulation , Enzyme Inhibitors/metabolism , Fatty Acid Desaturases/metabolism , Flow Cytometry , Humans , Mouth Neoplasms/metabolism , Oligonucleotides, Antisense/pharmacology , Polymerase Chain Reaction , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...