101
|
Park YC, Jeong JH, Park KJ, Choi HJ, Park YM, Jeong BK, Higuchi Y, Yoo YH. Sulindac activates nuclear translocation of AIF, DFF40 and endonuclease G but not induces oligonucleosomal DNA fragmentation in HT-29 cells. Life Sci 2005; 77:2059-70. [PMID: 15946692 DOI: 10.1016/j.lfs.2005.04.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2004] [Accepted: 04/25/2005] [Indexed: 11/29/2022]
Abstract
Sulindac is one of the most widely studied nonsteroidal anti-inflammatory drugs in the prevention of colon cancer. Thus, from the viewpoint of colon cancer chemotherapy it is important to reveal the mechanism of sulindac-induced cell death. This study was undertaken to dissect the molecular mechanism underlying sulindac-induced apoptosis in human colon cancer cell line HT-29 (mutant p53), focusing on nuclear translocation of AIF, DFF and endonuclease G. On induction of apoptosis by sulindac, it was associated with decreased mitochondrial membrane potential, nuclear expression of active caspase-3, cleavage of poly(ADP-ribose) polymerase, translocation of mitochondrial proteins to the nucleus, and morphological evidence of nuclear condensation. However, sulindac led to only disintegration of nuclear DNA into high molecular weight DNA fragments of about 100-300 kbp as determined by a pulse-field gel electrophoresis, suggesting a predominantly AIF-mediated cell death process. In summary, our findings indicate that sulindac induces large-scale DNA fragmentation without oligonucleosomal DNA fragmentation. This result suggests that nuclear translocation of DFF and endonuclease G are not sufficient for the induction of oligonucleosomal DNA fragmentation in HT-29 cells.
Collapse
Affiliation(s)
- Young Chul Park
- Department of Anatomy and Cell Biology, Dong-A University College of Medicine (BK21 Program), Busan 602-714, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
102
|
Abir F, Alva S, Kaminski DL, Longo WE. The role of arachidonic acid regulatory enzymes in colorectal disease. Dis Colon Rectum 2005; 48:1471-83. [PMID: 15868226 DOI: 10.1007/s10350-005-0015-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE Nonsteroidal anti-inflammatory drugs have a wide ranging effect on diseases of the colon and rectum. Interestingly, nonsteroidal anti-inflammatory drugs seem to play a beneficial role in colorectal cancer chemoprevention and adenoma regression, but may have a deleterious effect in inflammatory bowel disease. Prostaglandin inhibition is central to both the beneficial and toxic effects of this class of drugs. Arachidonic acid metabolism is essential to prostaglandin synthesis. METHODS A Medline search using "nonsteroidal anti-inflammatory drugs," "colon cancer," "inflammatory bowel disease," "colitis," "COX inhibitors," "arachidonic acid," and "chemoprevention" as key words was performed for English-language articles. Further references were obtained through cross-referencing the bibliography cited in each work. RESULTS Based on numerous studies, nonsteroidal anti-inflammatory drugs have a beneficial role in colon cancer and colonic adenomas. However, they have been reported to have a deleterious effect on the colon in inflammatory bowel disease and have been shown to cause colitis. Nonsteroidal anti-inflammatory drugs work via multiple pathways, some well defined, and others unknown. CONCLUSIONS In the new millennium, nonsteroidal anti-inflammatory drugs may be used for chemoprevention of colorectal and other cancers. In addition, they may be used in combination with surgery and chemotherapy to primarily treat colorectal carcinoma. Undoubtedly, the use of novel cyclooxygenase inhibitors with less of a toxicity profile will allow more widespread use of nonsteroidal anti-inflammatory drugs for a variety of diseases. The future of this class of drugs is promising.
Collapse
Affiliation(s)
- Farshad Abir
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut 06520-8062, USA
| | | | | | | |
Collapse
|
103
|
The role of inflammation in Alzheimer's disease neuropathology and clinical dementia. From epidemiology to treatment. NEURODEGENER DIS 2005. [DOI: 10.1017/cbo9780511544873.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
104
|
Ashktorab H, Dawkins FW, Mohamed R, Larbi D, Smoot DT. Apoptosis induced by aspirin and 5-fluorouracil in human colonic adenocarcinoma cells. Dig Dis Sci 2005; 50:1025-32. [PMID: 15986848 DOI: 10.1007/s10620-005-2698-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Various biochemical, clinical and epidemiological studies have shown that aspirin (ASA) and other nonsteroidal anti-inflammatory drugs (NSAIDs) demonstrate antineoplastic properties, particularly in the gastrointestinal tract, inhibiting the proliferation of colorectal cancer cells. The mechanism of action may be prostaglandin mediated through inhibition of the COX enzymatic system. This includes two iso-enzymes, COX-I and COX-II, working in concert with the activation of apoptosis, activation of immune surveillance, inhibition of proliferation, and inhibition of carcinogen activation. 5-Fluorouracil (5-FU) has demonstrated activity against colorectal cancer, leading to apoptosis of neoplastic cells. We evaluated the effects of varying doses of ASA (0.5, 1, 1.5 mM), both as a single agent and in combination with 5-FU (50 microg) in HT-29, a colon adenocarcinoma cell line. Proliferation assays showed that aspirin at a concentration of 1 mM inhibits cell growth. Cells treated with ASA, both alone and in combination with 5-FU, demonstrated apoptotic activity with the up-regulation of Bax protein, which is consistent with 5-FU anticancer treatment. Furthermore, there was synergistic cell death with ASA and 5-FU. DNA fragmentation, TUNEL, and trypan blue exclusion methods indicated that a combination of ASA and 5-FU induces apoptosis in cells in a time- and concentration-dependent manner. This study serves to further elucidate the mechanism of action of ASA, and ASA in combination with 5-FU, in colorectal cancer as evidenced by its effect on the HT-29 cell line.
Collapse
Affiliation(s)
- Hassan Ashktorab
- Department of Medicine, Division of Hematology/Oncology, Howard University Cancer Center, Howard University, Washington, DC 20059, USA.
| | | | | | | | | |
Collapse
|
105
|
Subhashini J, Mahipal SVK, Reddanna P. Anti-proliferative and apoptotic effects of celecoxib on human chronic myeloid leukemia in vitro. Cancer Lett 2005; 224:31-43. [PMID: 15911099 DOI: 10.1016/j.canlet.2004.11.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2004] [Revised: 10/30/2004] [Accepted: 11/02/2004] [Indexed: 01/04/2023]
Abstract
Celecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor, is the only non-steroidal anti-inflammatory drug so far which has been approved by the FDA for adjuvant treatment of patients with familial adenomatous polyposis. The molecular mechanism responsible for the anti-cancer effects of celecoxib is not fully understood. There is little data on the potential role of COX-2 in lymphoma pathogenesis. In view of the reported induction of apoptosis in cancer cells by cyclooxygenase-2 inhibitors, the present study is undertaken to test the effect of celecoxib on human chronic myeloid leukemia cell line, K562 and other hematopoietic cancer cell lines like Jurkat (human T lymphocytes), HL60 (human promyelocytic leukemia) and U937 (human macrophage). Treatment of these cells with celecoxib (10-100 microM) dose-dependently, reduced cell growth with arrest of the cell cycle at G0/G1 phase and induction of apoptosis. Further mechanism of apoptosis induction was elucidated in detail in K562 cell line. Apoptosis was mediated by release of cytochrome c into the cytoplasm and cleavage of poly (ADP-ribose) polymerase-1 (PARP-1). This was followed by DNA fragmentation. The level of anti-apoptotic protein Bcl-2 was decreased without any change in the pro-apoptotic Bax. Celecoxib also inhibited NF-kB activation. Celecoxib thus potentiates apoptosis as shown by MTT assay, cytochrome c leakage, PARP cleavage, DNA fragmentation, Bcl-2 downregulation and possibly by inhibiting NF-kB activation.
Collapse
Affiliation(s)
- J Subhashini
- Department of Animal Sciences, School of Life Sciences, University of Hyderabad, Hyderabad 500 046, India
| | | | | |
Collapse
|
106
|
Kobayashi S, Nantz R, Kitamura T, Higashikubo R, Horikoshi N. Combined inhibition of extracellular signal-regulated kinases and HSP90 sensitizes human colon carcinoma cells to ionizing radiation. Oncogene 2005; 24:3011-9. [PMID: 15735687 DOI: 10.1038/sj.onc.1208508] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Indomethacin, a common nonsteroidal anti-inflammatory drug, has been shown to enhance radiation-mediated cell-killing effect through the activation of p38 mitogen-activated protein kinase (MAPK). We found that indomethacin strongly reduced the basal level of extracellular signal-regulated kinases 1 and 2 (ERK1/2) in HT-29 human colon carcinoma cells. The inhibition of ERK1/2 by indomethacin was only observed in cells with high basal activities of ERK1/2 such as HT-29 cells, but not in cells with low basal activities, such as HeLa. Cell cycle analysis of HT-29 cells exposed with indomethacin showed a partial G1/S arrest and slow DNA synthesis. However, the treatment with NS398, a specific COX-1/2 inhibitor, failed to show any effect on cell cycle, indicating that the inhibition of COX-1/2 is not responsible for cell cycle arrest. Since U0126, a specific inhibitor for MEK1/2, also induced a partial G1/S arrest, the G1/S arrest induced by indomethacin is, at least in part, caused by the inhibition of ERK1/2. Cell proliferation of HT-29 was inhibited by the treatment of U0126 but not in HeLa cells, and the treatment of HT-29 cells with U0126 enhanced radiation sensitivity possibly due to the accumulation of cells in G1 phase. We found that 17-allylamino-17-demethoxygeldanamycin, a geldanamycin delivative, radiosensitized HT-29 cells at a relatively low dose of irradiation, and indomethacin and U0126 further enhanced this effect. Therefore, tumor cells with elevated ERK1/2 activity can be effectively sensitized to radiation treatment by a combinational inhibition of HSP90 and MAPK activity.
Collapse
Affiliation(s)
- Shinichiro Kobayashi
- Department of Radiation Oncology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO 63108, USA
| | | | | | | | | |
Collapse
|
107
|
Ilsley JNM, Nakanishi M, Flynn C, Belinsky GS, De Guise S, Adib JN, Dobrowsky RT, Bonventre JV, Rosenberg DW. Cytoplasmic Phospholipase A2Deletion Enhances Colon Tumorigenesis. Cancer Res 2005; 65:2636-43. [PMID: 15805260 DOI: 10.1158/0008-5472.can-04-3446] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cellular pools of free arachidonic acid are tightly controlled through enzymatic release of the fatty acid and subsequent utilization by downstream enzymes including the cyclooxygenases. Arachidonic acid cleavage from membrane phospholipids is accomplished by the actions of phospholipase A(2) (PLA(2)). Upon release, free arachidonic acid provides substrate for the synthesis of eicosanoids. However, under certain conditions, arachidonic acid may participate in ceramide-mediated apoptosis. Disruption of arachidonic acid homeostasis can shift the balance of cell turnover in favor of tumorigenesis, via overproduction of tumor-promoting eicosanoids or alternatively by limiting proapoptotic signals. In the following study, we evaluated the influence of genetic deletion of a key intracellular phospholipase, cytoplasmic PLA(2) (cPLA(2)), on azoxymethane-induced colon tumorigenesis. Heterozygous and null mice, upon treatment with the organotropic colon carcinogen, azoxymethane, developed a significant (P < 0.05) increase in colon tumor multiplicity (7.2-fold and 5.5-fold, respectively) relative to their wild-type littermates. This enhanced tumor sensitivity may be explained, in part, by the attenuated levels of apoptosis observed by terminal deoxynucleotidyl transferase-mediated nick end labeling staining within the colonic epithelium of heterozygous and null mice ( approximately 50% of wild type). The lower frequency of apoptotic cells corresponded with reduced ceramide levels (69% and 46% of wild-type littermates, respectively). Remarkably, increased tumorigenesis resulting from cPLA(2) deletion occurred despite a significant reduction in prostaglandin E(2) production, even in cyclooxygenase-2-overexpressing tumors. These data contribute new information that supports a fundamental role of cPLA(2) in the control of arachidonic acid homeostasis and cell turnover. Our findings indicate that the proapoptotic role of cPLA(2) in the colon may supercede its contribution to eicosanoid production in tumor development.
Collapse
Affiliation(s)
- Jillian N M Ilsley
- Program in Colorectal Cancer, Center for Molecular Medicine, University of Connecticut Health Center, Farmington, Connecticut 06030, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
108
|
Kolluri SK, Corr M, James SY, Bernasconi M, Lu D, Liu W, Cottam HB, Leoni LM, Carson DA, Zhang XK. The R-enantiomer of the nonsteroidal antiinflammatory drug etodolac binds retinoid X receptor and induces tumor-selective apoptosis. Proc Natl Acad Sci U S A 2005; 102:2525-30. [PMID: 15699354 PMCID: PMC548323 DOI: 10.1073/pnas.0409721102] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Prostate cancer is often slowly progressive, and it can be difficult to treat with conventional cytotoxic drugs. Nonsteroidal antiinflammatory drugs inhibit the development of prostate cancer, but the mechanism of chemoprevention is unknown. Here, we show that the R-enantiomer of the nonsteroidal antiinflammatory drug etodolac inhibited tumor development and metastasis in the transgenic mouse adenocarcinoma of the prostate (TRAMP) model, by selective induction of apoptosis in the tumor cells. This proapoptotic effect was associated with loss of the retinoid X receptor (RXRalpha) protein in the adenocarcinoma cells, but not in normal prostatic epithelium. R-etodolac specifically bound recombinant RXRalpha, inhibited RXRalpha transcriptional activity, and induced its degradation by a ubiquitin and proteasome-dependent pathway. The apoptotic effect of R-etodolac could be controlled by manipulating cellular RXRalpha levels. These results document that pharmacologic antagonism of RXRalpha transactivation is achievable and can have profound inhibitory effects in cancer development.
Collapse
|
109
|
Kim KS, Yoon JH, Kim JK, Baek SJ, Eling TE, Lee WJ, Ryu JH, Lee JG, Lee JH, Yoo JB. Cyclooxygenase inhibitors induce apoptosis in oral cavity cancer cells by increased expression of nonsteroidal anti-inflammatory drug-activated gene. Biochem Biophys Res Commun 2005; 325:1298-303. [PMID: 15555568 DOI: 10.1016/j.bbrc.2004.10.176] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2004] [Indexed: 02/06/2023]
Abstract
We have investigated whether NAG-1 is induced in oral cavity cancer cells by various NSAIDs and if apoptosis induced by NSAIDs can be linked directly with the induction of NAG-1. NAG-1 expression was increased by diclofenac, aceclofenac, indomethacin, ibuprofen, and sulindac sulfide, in the order of NAG-1 induction, but not by acetaminophen, piroxicam or NS-398. Diclofenac was the most effective NAG-1 inducer. Incubation with diclofenac inhibited cell proliferation and induced apoptosis. The expression of NAG-1 was observed in advance of the induction of apoptosis. Conditioned medium from NAG-1-overexpressing Drosophila cells inhibited SCC 1483 cells proliferation and induced apoptosis. In summary, some NSAIDs induce NAG-1 expression in oral cavity cancer cells and the induced NAG-1 protein appears to mediate apoptosis. Therefore, NSAIDs may be considered as a possible chemopreventive agent against oral cavity cancer.
Collapse
Affiliation(s)
- Kyung-Su Kim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, 134 Shinchon-dong, Seodaemoon-gu, Seoul, Republic of Korea.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
110
|
Chang JK, Wang GJ, Tsai ST, Ho ML. Nonsteroidal anti-inflammatory drug effects on osteoblastic cell cycle, cytotoxicity, and cell death. Connect Tissue Res 2005; 46:200-10. [PMID: 16546823 DOI: 10.1080/03008200500344025] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Previous studies indicated that nonsteroidal anti-inflammatory drugs (NSAIDs) suppress bone repair, growth, and remodeling in vivo. Our previous in vitro study demonstrated that indomethacin and ketorolac inhibited osteoblast proliferation. In this study, we further investigated the influences of 4 NSAIDs on cell cycle kinetics, cytotoxicity, and cell death pattern in osteoblast cultures from rat fetal calvaria. Our results showed that NSAIDs significantly arrested cell cycle at the G(0)/G(1) phase and induced cytotoxicity and cell death of osteoblasts. Apoptosis was more pronounced than necrosis caused by NSAIDs. Among these NSAIDs, piroxicam showed the least effect to produce osteoblastic dysfunction. Moreover, we found that the cytotoxic and apoptotic effects of NSAIDs on osteoblasts might not be prostaglandin related. These results suggest that the NSAID effects on cell cycle arrest and cell death induction in osteoblasts may be one of the important mechanisms contributing to their suppressive effect on bone formation.
Collapse
Affiliation(s)
- Je-Ken Chang
- Department of Orthopaedics, School of Medicine, Orthopaedic Research Center, Kaohsiung Medical University Chug-Ho Memorial Hospital, Kaohsiung, Taiwan
| | | | | | | |
Collapse
|
111
|
Hu Y, Le Leu RK, Young GP. Sulindac corrects defective apoptosis and suppresses azoxymethane-induced colonic oncogenesis inp53 knockout mice. Int J Cancer 2005; 116:870-5. [PMID: 15849741 DOI: 10.1002/ijc.21107] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The acute apoptotic response to genotoxic carcinogens (AARGC) might be important for controlling the subsequent colonic mutational load and progression through oncogenesis. We have found previously that AARGC is p53-dependent with a gene-dosage effect, and that decreased AARGC in p53(+/-) and p53(-/-) mice is associated with increased susceptibility to carcinogen-induced oncogenesis. We tested the ability of sulindac to reverse these defects. The effect of sulindac on azoxymethane (AOM)-induced apoptosis was measured in colonic epithelium in wild-type, p53(+/-) and p53(-/-) mice, 8 hr after a single AOM injection. Sulindac supplementation (0.5 +/- 0.1 mg/day) restored defective AARGC in p53(+/-) but not in p53(-/-) mice. For effect on colon tumor development, sulindac treatment was started at age 4 weeks in wild-type, p53(+/-) and p53(-/-) mice; three weekly AOM injections were commenced at 6 weeks of age to induce tumors. Sulindac reduced significantly tumor incidence and multiplicity in wild-type mice (17% and 0.3 tumors/mouse compared to 36% and 0.8 respectively without drug), in p53(+/-)mice (38% and 0.8 compared to 64% and 1.63) and in p53(-/-) mice (63% and 1.0 compared to 90% and 1.74). Although loss of p53 function impairs the apoptotic response to AOM-induced DNA damage, sulindac is capable of partly restoring this defect. As sulindac also reverses the increased risk of oncogenesis due to p53 dysfunction, its enhancement of the apoptotic response to initiating mutations might act to reduce mutational load driving oncogenesis. Sulindac is an effective chemopreventive agent in the presence of p53 dysfunction.
Collapse
Affiliation(s)
- Ying Hu
- Department of Medicine Flinders University of South Australia, Adelaide, Australia.
| | | | | |
Collapse
|
112
|
Galfi P, Neogrady Z, Amberger A, Margreiter R, Csordas A. Sensitization of colon cancer cell lines to butyrate-mediated proliferation inhibition by combined application of indomethacin and nordihydroguaiaretic acid. ACTA ACUST UNITED AC 2005; 29:276-85. [PMID: 15936596 DOI: 10.1016/j.cdp.2004.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2004] [Accepted: 12/01/2004] [Indexed: 11/20/2022]
Abstract
The aim of the study was to investigate the effects of nonsteroidal anti-inflammatory drugs (NSAIDs) on histone deacetylase-mediated proliferation inhibition. In the colon cancer cell line HT29 butyrate-mediated proliferation inhibition was enhanced by the additional presence of indomethacin (IM) and/or nordihydroguaiaretic acid (NDGA). Sensitisation to butyrate-mediated proliferation inhibition was abolished by the general caspase inhibitor Z-VAD-fmk, however, only IM-induced cell detachment was prevented by the caspase inhibitor but not that induced by NDGA or NDGA plus IM. In contrast to the parental cell line HT29, in the methotrexate-resistant sub-lines HT29-12 and HT29-21, IM counteracted butyrate-mediated proliferation inhibition, which was abrogated by NDGA. In all the investigated cell lines, proliferation inhibition was most effectively achieved under the combined application of butyrate with IM and NDGA, suggesting that inhibition of both cyclooxygenase (COX) and lipoxygenase (LOX) isoenzymes is needed for proliferation inhibition by NSAIDs in tumour cells.
Collapse
Affiliation(s)
- Peter Galfi
- Institute of Physiology and Biochemistry, Faculty of Veterinary Sciences, Szent-István University, Budapest, Hungary
| | | | | | | | | |
Collapse
|
113
|
Luo ZQ, Wu SM, Xiao SD, Tong JF, Luo HY, Shen GF. Effects of Helicobacter pylori and NSAIDs on cytokinetics of gastric epithelium. Shijie Huaren Xiaohua Zazhi 2004; 12:2594-2598. [DOI: 10.11569/wcjd.v12.i11.2594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of Helicobacter pylori (H pylori ) and (NSAID) indomethacin and aspirin on the proliferation and apoptosis of gastric epithelial cells
METHODS: Gastric cancer cell line AGS cells were co-cultured with H pylori and/or NSAID, and then proliferation of AGS was examined by MTT assay, Western blotting and detection of proliferating cell nuclear antigen (PCNA). At the same time, cell apoptosis was detected with the FITC-Annexin-V/PI double staining, DNA gel eletrophoresis and transmission electron microscopy were used to confirm the results.
RESULTS: The CagA-positive H pylori strain NCTC11637 could enhance cell proliferation. Besides, low concentrations (range from 3.2×107 CFU/L to 4×109 CFU/L) of H pylori could promote proliferation (P <0.05) of AGS cell line while high concentrations (>2 ×1010 CFU/L) could inhibit the growth of AGS cells (P <0.05). When incubated with H pylori and NSAID together, proliferation of AGS cells was inhibited. Indomethacin and aspirin increased apoptosis of AGS cells significantly (P <0.05). No obvious apoptosis was observed in the H pylori-infected cells. When cells were co-cultured with H pylori and NSAID, the percentage of apoptosis also increased significantly (P <0.05), but the percentage was lower than that of cells incubated with NSAID alone. These results were confirmed by transmission electron microscopy and DNA gel electrophoresis.
CONCLUSION: CagA(+)H pylori strains are more prone to enhance the proliferation of gastric epithelial cells than cagA(-) H pylori strains. The effects of H pylori on the cell growth are associated with the concentrations of H pylori. NSAID can inhibit the gastric epithelial cell proliferation.
Collapse
Affiliation(s)
- Zhi-Qin Luo
- Department of Internal Medicine, Zhoupu Hospital, Nanhui District, Shanghai 201318, China
| | - Shu-Ming Wu
- Shanghai Institute of Digestive System Diseases, Department of Gastroenterology, Affiliated Renji Hospital, Shanghai Second Medical University, Shanghai 200001, China
| | - Shu-Dong Xiao
- Shanghai Institute of Digestive System Diseases, Department of Gastroenterology, Affiliated Renji Hospital, Shanghai Second Medical University, Shanghai 200001, China
| | - Ju-Fang Tong
- Shanghai Institute of Digestive System Diseases, Department of Gastroenterology, Affiliated Renji Hospital, Shanghai Second Medical University, Shanghai 200001, China
| | - Hong-Yu Luo
- Shanghai Institute of Digestive System Diseases, Department of Gastroenterology, Affiliated Renji Hospital, Shanghai Second Medical University, Shanghai 200001, China
| | - Guan-Feng Shen
- Shanghai Institute of Digestive System Diseases, Department of Gastroenterology, Affiliated Renji Hospital, Shanghai Second Medical University, Shanghai 200001, China
| |
Collapse
|
114
|
Huguenin S, Fleury-Feith J, Kheuang L, Jaurand MC, Bolla M, Riffaud JP, Chopin DK, Vacherot F. Nitrosulindac (NCX 1102): a new nitric oxide-donating non-steroidal anti-inflammatory drug (NO-NSAID), inhibits proliferation and induces apoptosis in human prostatic epithelial cell lines. Prostate 2004; 61:132-41. [PMID: 15305336 DOI: 10.1002/pros.20081] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The aim of our study was to explore the anti-tumoral potential of the Nitric Oxide-Donating Non-Steroidal Anti-Inflammatory Drugs (NO-NSAID) NCX1102 (nitrosulindac), on three human prostatic epithelial cell lines at varying degree of transformation (PNT1A, LNCaP, and PC3). METHODS Cytotoxicity, anti-proliferative effects, cell-cycle alterations, morphological changes, and apoptosis were investigated after treatment with nitrosulindac in comparison to the native molecule sulindac. Involvement of the polyamine pathway in the action of nitrosulindac was also examined. RESULTS Nitrosulindac but not sulindac exerted a cytotoxic effect on all cell lines and an anti-proliferative effect on LNCaP and PC3 cells only. Nitrosulindac differentially altered the cell cycle, induced mitotic arrest and displayed a pro-apoptotic activity in all cell lines. Finally, the polyamine pathway does not seem to be involved in the mechanism of nitrosulindac action. CONCLUSIONS Our results demonstrate the anti-proliferative and proapoptotic activity of nitrosulindac on prostate cancer cell lines and suggest its potential interest for new strategies in the management of prostate cancer.
Collapse
Affiliation(s)
- Sandra Huguenin
- Equipe de recherche INSERM E 03-37 Oncogenèse des Tumeurs Respiratoires et Urogénitales, Faculté de Médecine, Rue du Général Sarrail, Créteil, Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
115
|
Muralidhar B, Carpenter KLH, Müller K, Skepper JN, Arends MJ. Potency of arachidonic acid in polyunsaturated fatty acid-induced death of human monocyte-macrophages: implications for atherosclerosis. Prostaglandins Leukot Essent Fatty Acids 2004; 71:251-262. [PMID: 15301796 DOI: 10.1016/j.plefa.2004.03.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2003] [Accepted: 03/17/2004] [Indexed: 12/23/2022]
Abstract
Evidence suggests that oxidation of LDL is involved in the progression of atherosclerosis by inducing apoptosis in macrophages. Polyunsaturated fatty acids (PUFAs) are prominent components of LDL and are highly peroxidisable. We therefore tested PUFAs for induction of apoptosis in human monocyte-macrophages in vitro. Arachidonic acid (AA) induced the highest levels of apoptosis followed by docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), despite DHA and EPA being more peroxidisable than AA. alpha-Linolenic acid induced lower levels of apoptosis. Linoleic and oleic acids were innocuous. Results of experiments with AA products and enzyme inhibitors suggest roles for peroxidation, cyclooxygenase and lipoxygenase in AA-induced apoptosis. Our results further suggest activation of PPARgamma by AA and DHA associated with apoptosis induction. These findings may be relevant to potential mechanisms of fatty acid influences on plaques and may suggest strategies for combating atherosclerosis progression.
Collapse
Affiliation(s)
- Balaji Muralidhar
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | | | | | | | | |
Collapse
|
116
|
Tsibouris P, Hendrickse MT, Isaacs PET. Daily use of non-steroidal anti-inflammatory drugs is less frequent in patients with Barrett's oesophagus who develop an oesophageal adenocarcinoma. Aliment Pharmacol Ther 2004; 20:645-55. [PMID: 15352913 DOI: 10.1111/j.1365-2036.2004.02150.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Non-steroidal anti-inflammatory drugs use may protect against development of oesophageal adenocarcinoma. AIM To define the consequences of non-steroidal anti-inflammatory drugs use in patients with Barrett's oesophagus. METHODS Records of all Barrett's oesophagus/oesophageal adenocarcinoma patients examined in Blackpool-Wyre-Fylde area were reviewed. All surviving patients completed validated questionnaires. RESULTS Use of non-steroidal anti-inflammatory drugs of any type and at any frequency was more prevalent in Barrett's oesophagus patients [147 (38%) Barrett's oesophagus vs. 30 (26%) oesophageal adenocarcinoma, P = 0.02]. Daily use of non-steroidal anti-inflammatory drugs was more prevalent in Barrett's oesophagus patients [88 (23%) Barrett's oesophagus vs. 14 (12%) oesophageal adenocarcinoma, P = 0.02], due to more prevalent consumption of non-aspirin non-steroidal anti-inflammatory drugs [48 (13%) Barrett's oesophagus vs. four (4%) oesophageal adenocarcinoma, P = 0.009]. There was no difference between the two groups in usage of either daily low-dose aspirin or of occasional non-steroidal anti-inflammatory drugs. In logistic regression analysis any use of non-steroidal anti-inflammatory drugs [odds ratio (OR) = 0571 (95% CI: 0.359-0.909), P = 0.018] and daily use of non-aspirin non-steroidal anti-inflammatory drugs [OR = 0.297 (95% CI: 0.097-0.911), P = 0.034] were significant protective factors. Non-steroidal anti-inflammatory drugs use did not affect the survival of oesophageal adenocarcinoma patients. Oesophageal adenocarcinoma and Barrett's oesophagus consuming non-steroidal anti-inflammatory drugs did not differ in upper gastrointestinal bleeding [26 (15%) non-steroidal anti-inflammatory drugs consumers vs. 29 (9%) non-consumers, P = 0.08], oesophageal ulcers [31 (18%) non-steroidal anti-inflammatory drug consumers vs. 49 (15%) non-consumers, P = 0.43] or stricturing [19 (11%) non-steroidal anti-inflammatory drug consumers vs. 41 (13%) non-consumers, P = 0.58]. CONCLUSIONS (i) Daily use of non-steroidal anti-inflammatory drugs is more prevalent in Barrett's oesophagus than oesophageal adenocarcinoma patients, because of a more prevalent use of non-aspirin non-steroidal anti-inflammatory drugs. (ii) Use of non-steroidal anti-inflammatory drugs in Barrett's oesophagus patients is safe if acid suppression is adequate.
Collapse
|
117
|
Gardner SH, Hawcroft G, Hull MA. Effect of nonsteroidal anti-inflammatory drugs on beta-catenin protein levels and catenin-related transcription in human colorectal cancer cells. Br J Cancer 2004; 91:153-63. [PMID: 15188006 PMCID: PMC2364748 DOI: 10.1038/sj.bjc.6601901] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Elevated β-catenin levels in human colorectal cancer (CRC) cells lead to increased trans-activation of ‘protumorigenic’ β-catenin/T-cell factor (TCF) target genes such as cyclin D1. Therefore, possible targets for the anti-CRC activity of nonsteroidal anti-inflammatory drugs (NSAIDs) are β-catenin and catenin-related transcription (CRT). We tested the antiproliferative activity and the effects on levels of β-catenin and cyclin D1 protein, as well as CRT (measured using a synthetic β-catenin/TCF-reporter gene [TOPflash]), of a panel of NSAIDs (indomethacin, diclofenac, sulindac sulphide and sulphone, rofecoxib; range 10–600 μM) on SW480 human CRC cells in vitro. Following NSAID treatment, there was no consistent relationship between reduced cell proliferation, induction of apoptosis and changes in β-catenin protein levels or CRT. All the NSAIDs, except rofecoxib, decreased nuclear β-catenin content and cyclin D1 protein levels in parallel with their antiproliferative activity. However, cyclin D1 downregulation occurred prior to a decrease in total β-catenin protein levels and there was no correlation with changes in CRT, suggesting the existence of CRT-independent effects of NSAIDs on cyclin D1 expression. In summary, NSAIDs have differential effects on β-catenin protein and CRT, which are unlikely to fully explain their effects on cyclin D1 and their antiproliferative activity on human CRC cells in vitro.
Collapse
Affiliation(s)
- S H Gardner
- Molecular Medicine Unit, University of Leeds, St James's University Hospital, Leeds LS9 7TF, UK
| | - G Hawcroft
- Molecular Medicine Unit, University of Leeds, St James's University Hospital, Leeds LS9 7TF, UK
| | - M A Hull
- Molecular Medicine Unit, University of Leeds, St James's University Hospital, Leeds LS9 7TF, UK
- Molecular Medicine Unit, University of Leeds, St James's University Hospital, Leeds LS9 7TF, UK. E-mail:
| |
Collapse
|
118
|
Yeh RK, Chen J, Williams JL, Baluch M, Hundley TR, Rosenbaum RE, Kalala S, Traganos F, Benardini F, del Soldato P, Kashfi K, Rigas B. NO-donating nonsteroidal antiinflammatory drugs (NSAIDs) inhibit colon cancer cell growth more potently than traditional NSAIDs: a general pharmacological property? Biochem Pharmacol 2004; 67:2197-205. [PMID: 15163551 DOI: 10.1016/j.bcp.2004.02.027] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2004] [Accepted: 02/06/2004] [Indexed: 11/23/2022]
Abstract
The novel nitric oxide-donating nonsteroidal antiinflammatory drugs (NO-NSAIDs), consisting of a traditional NSAID to which a NO releasing moiety is covalently attached, may have an important role in colon cancer prevention and/or treatment. Preclinical studies have shown that NO-aspirin (NO-ASA) is more potent than traditional ASA in preventing colon cancer. Preclinical and clinical studies have also documented its superior safety, compared to traditional ASA. To evaluate the role of this structural modification on the cancer cell growth inhibitory effect of NSAIDs, we studied seven pairs of traditional NSAIDs (ASA, salicylic acid, indomethacin, sulindac, ibuprofen, flurbiprofen, piroxicam) and their corresponding NO-NSAIDs. All NO-NSAIDs (except NO-piroxicam which is a salt and not a true NO-NSAID) have greater potency in inhibiting HT-29 and HCT-15 colon cancer cell growth compared to their NSAID counterparts: the IC(50)s of the NO-NSAIDs were enhanced between 7- and 689-fold in HT-29 cells and 1.7- to 1083-fold in HCT-15 cells over those of the corresponding NSAIDs. Their growth inhibitory effect is due to a profound cell kinetic effect consisting of reduced cell proliferation and enhanced cell death. Since HT-29 cells express cyclooxygenases but HCT-15 do not, this effect appears independent of cyclooxygenase in the colon cancer cells. Thus the structural modification of these traditional NSAIDs leading to NO-NSAIDs enhances their potency in inhibiting colon cancer cell growth. Our findings suggest that the enhanced potency imparted on NSAIDs by this structural modification represents a pharmacological property that may be a general one for this class of compounds.
Collapse
Affiliation(s)
- Raymond K Yeh
- American Health Foundation Cancer Center, Institute for Cancer Prevention, Valhalla, NY 06595, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
119
|
Gomes-Pereira M, Monckton DG. Chemically induced increases and decreases in the rate of expansion of a CAG*CTG triplet repeat. Nucleic Acids Res 2004; 32:2865-72. [PMID: 15155855 PMCID: PMC419613 DOI: 10.1093/nar/gkh612] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Somatic mosaicism of repeat length is prominent in repeat expansion disorders such as Huntington disease and myotonic dystrophy. Somatic mosaicism is age-dependent, tissue-specific and expansion-biased, and likely contributes toward the tissue-specificity and progressive nature of the symptoms. We propose that therapies targeted at somatic repeat expansion may have general utility in these disorders. Specifically, suppression of somatic expansion would be expected to be therapeutic, whilst reversion of the expanded mutant repeat to within the normal range would be predicted to be curative. However, the effects of genotoxic agents on the mutational properties of specific nuclear genes are notoriously difficult to define. Nonetheless, we have determined that chronic exposure over a three month period to a number of genotoxic agents can alter the rate of triplet repeat expansion in whole populations of mammalian cells. Interestingly, high doses of caffeine increased the rate of expansion by approximately 60%. More importantly, cytosine arabinoside, ethidium bromide, 5-azacytidine and aspirin all significantly reduced the rate of expansion by from 35 to 75%. These data establish that drug induced suppression of somatic expansion is possible. These data also suggest that highly unstable expanded simple sequence repeats may act as sensitive reporters of genotoxic assault in the soma.
Collapse
Affiliation(s)
- Mário Gomes-Pereira
- Institute of Biomedical and Life Sciences, University of Glasgow, Anderson College Building, 56 Dumbarton Road, Glasgow G11 6NU, UK
| | | |
Collapse
|
120
|
Cyclooxygenase (COX)-2-dependent effects of the inhibitor SC236 when combined with ionizing radiation in mammary tumor cells derived from HER-2/ neu mice. Mol Cancer Ther 2004. [DOI: 10.1158/1535-7163.417.3.4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Abstract
Cyclooxygenase (COX)-2-derived prostaglandins (PGs) are thought to contribute to tumor growth and resistance to radiation therapy. COX-2 protein expression is increased in many tumors including those of the breast. COX-2-derived PGs have been shown to protect cells from radiation damage. This study evaluated the role of COX-2-derived PG in radiation treatment by using the NMF11.2 mammary tumor cell line originally obtained from HER-2/neu mice that overexpress HER-2/neu. We determined whether the effects of the COX-2 inhibitor SC236 on cell growth, radiation-induced PGE2 production and COX expression, cell cycle redistribution, and vascular endothelial growth factor (VEGF) were acting through COX-2-dependent mechanisms. The NMF11.2 cells expressed both COX-1 and COX-2 protein and mRNA. The radiation treatment alone led to a dose-dependent increase in the levels of COX-2 mRNA and COX-2 protein, which was associated with an increase in the production of PGE2 and prostacyclin (PGI2). Treating NMF11.2 cells with high concentrations (20 μm) of SC236 for 48 h reduced the radiation-induced increase in COX-2 activity and also decreased cell growth. SC236 (20 μm) increased the accumulation of the cells in the radiosensitive G2-M phase of the cell cycle. However, a low concentration (5 μm) of SC236 was adequate to reduce COX-2 activity. The lower concentration of SC236 (5 μm) also decreased cell growth after a longer incubation period (96 h) and, in combination with a 2 or 5 Gy dose, led to an accumulation of cells in G2-M phase. Restoring PG to control values in cells treated with 5 μm SC236 prevented the growth inhibition and G2-M cell cycle arrest. Radiation treatment of NMF11.2 cells also increased VEGF protein expression and VEGF secretion in a dose-dependent manner, which was blocked in those cells pretreated with 20 μm SC236 but not in those pretreated with 5 μm SC236. These findings indicate that the COX-2 inhibitor SC236 reduced cell growth and arrested cells in the G2-M phase of the cell cycle by mechanisms that are both dependent and independent of PG production while its effects on VEGF appear to be independent of COX-2.
Collapse
|
121
|
Wallace JM. Nutritional and botanical modulation of the inflammatory cascade--eicosanoids, cyclooxygenases, and lipoxygenases--as an adjunct in cancer therapy. Integr Cancer Ther 2004; 1:7-37; discussion 37. [PMID: 14664746 DOI: 10.1177/153473540200100102] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Emerging on the horizon in cancer therapy is an expansion of the scope of treatment beyond cytotoxic approaches to include molecular management of cancer physiopathology. The goal in these integrative approaches, which extends beyond eradicating the affected cells, is to control the cancer phenotype. One key new approach appears to be modulation of the inflammatory cascade, as research is expanding that links cancer initiation, promotion, progression, angiogenesis, and metastasis to inflammatory events. This article presents a literature review of the emerging relationship between neoplasia and inflammatory eicosanoids (PGE2 and related prostaglandins), with a focus on how inhibition of their synthesizing oxidases, particularly cyclooxygenase (COX), offers anticancer actions in vitro and in vivo. Although a majority of this research emphasizes the pharmaceutical applications of nonsteroidal anti-inflammatory drugs and selective COX-2 inhibitors, these agents fail to address alternate pathways available for the synthesis of proinflammatory eicosanoids. Evidence is presented that suggests the inhibition of lipoxygenase and its by-products-LTB4, 5-HETE, and 12-HETE-represents an overlooked but crucial component in complementary cancer therapies. Based on the hypothesis that natural agents capable of modulating both lipoxygenase and COX may advance the efficacy of cancer therapy, an overview and discussion is presented of dietary modifications and selected nutritional and botanical agents (notably, omega-3 fatty acids, antioxidants, boswellia, bromelain, curcumin, and quercetin) that favorably influence eicosanoid production.
Collapse
Affiliation(s)
- Jeanne M Wallace
- Nutritional Solutions, Inc., 2935 North, 1000 East, North Logan, UT 84341, USA.
| |
Collapse
|
122
|
Petkova DK, Clelland C, Ronan J, Pang L, Coulson JM, Lewis S, Knox AJ. Overexpression of cyclooxygenase-2 in non-small cell lung cancer. Respir Med 2004; 98:164-72. [PMID: 14971881 DOI: 10.1016/j.rmed.2003.09.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Evidence is accumulating to suggest that the inducible isoenzyme of cyclooxygenase (COX)-2 is up-regulated in human cancers and epidemiological studies indicate that COX inhibitors may have a protective effect on the development of lung cancer. We used immunohistochemistry and Western blotting to investigate COX expression in lung tumour specimens and three lung cancer cell lines. Sixty-five archival lung tissue samples, including 46 squamous cell and 6 adenocarcinoma lung resections, and 13 small cell lung cancer (SCLC) biopsies were studied. Dense and intense cytoplasmic COX-2 staining was found in all 52 resections from non-small cell lung cancer (NSCLC). The staining was diffuse and much stronger than adjacent respiratory epithelium. COX-2 staining was relatively weak in the majority of the SCLC samples. The bronchial and bronchiolar epithelium in the surrounding normal lung structures showed uniform COX immunoreactivity with apical concentration of the stain. There was no increase in COX-1 staining in any tumour type. Western blot analysis of the cancer lines revealed significantly higher expression of COX-1 in CORL23 line and COX-2 in two NSCLC cell lines (MOR/P; A549) compared with the expression of COX-1 and COX-2 in cultured normal bronchial epithelial cells. Our findings demonstrated COX-2 overexpression in NSCLC.
Collapse
Affiliation(s)
- D K Petkova
- Division of Respiratory Medicine, City Hospital, University of Nottingham, Hucknall Road, Nottingham NG5 1PB, UK
| | | | | | | | | | | | | |
Collapse
|
123
|
Kovaríková M, Hofmanová J, Soucek K, Kozubík A. The effects of TNF-α and inhibitors of arachidonic acid metabolism on human colon HT-29 cells depend on differentiation status. Differentiation 2004; 72:23-31. [PMID: 15008823 DOI: 10.1111/j.1432-0436.2004.07201006.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The level of differentiation could influence sensitivity of colonic epithelial cells to various stimuli. In our study, the effects of TNF-alpha, inhibitors of arachidonic acid (AA) metabolism (baicalein, BA; indomethacin, INDO; niflumic acid, NA; nordihydroguaiaretic acid, NDGA), and/or their combinations on undifferentiated or sodium butyrate (NaBt)-differentiated human colon adenocarcinoma HT-29 cells were compared. NaBt-treated cells became growth arrested (blocked in G0/G1 phase of the cell cycle), and showed down-regulated Bcl-xL and up-regulated Bak proteins and increased expression of cyclooxygenase-2 (COX-2) and 5-lipoxygenase (5-LOX). These cells were more perceptive to anti-proliferative and apoptotic effects of TNF-alpha. Both inhibitors of LOX (BA and NDGA) and COX (INDO and NA) in higher concentrations modulated cell cycle changes accompanying NaBt-induced differentiation and induced various level of cell death in undifferentiated and differentiated cells. Most important is our finding that TNF-alpha action on proliferation and cell death can be potentiated by co-treatment of cells with AA metabolism inhibitors, and that these effects were more significant in undifferentiated cells. TNF-alpha and INDO co-treatment was associated with accumulation of cells in G0/G1 cell cycle phase, increased reactive oxygen species production, and elevated caspase-3 activity. These results indicate the role of differentiation status in the sensitivity of HT-29 cells to the anti-proliferative and proapoptotic effects of TNF-alpha, AA metabolism inhibitors, and their combinations, and imply promising possibility for novel anti-cancer strategies.
Collapse
Affiliation(s)
- Martina Kovaríková
- Laboratory of Cytokinetics, Institute of Biophysics, Academy of Sciences of the Czech Republic, CZ-612 65 Brno, Královopolská 135, Czech Republic
| | | | | | | |
Collapse
|
124
|
Salgueiro-Pagadigorria CL, Kelmer-Bracht AM, Bracht A, Ishii-Iwamoto EL. Naproxen affects Ca2+ fluxes in mitochondria, microsomes and plasma membrane vesicles. Chem Biol Interact 2004; 147:49-63. [PMID: 14726152 DOI: 10.1016/j.cbi.2003.10.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
There is substantial evidence that nonsteroidal anti-inflammatory drugs (NSAIDs) affect cellular processes regulated by Ca(2+) ions, including the metabolic responses of the liver to Ca(2+)-dependent hormones. The aim of the present study was to determine whether the effects of naproxen are mediated by a direct action on cellular Ca(2+) fluxes. The effects of naproxen on 45Ca(2+) fluxes in mitochondria, microsomes and inside-out plasma membrane vesicles were examined. Naproxen strongly impaired the mitochondrial capacity to retain 45Ca(2+) and inhibited also ATP-dependent 45Ca(2+) uptake by microsomes. Naproxen did not modify 45Ca(2+) uptake by inside-out plasma membrane vesicles, but it inhibited the hexokinase/glucose-induced Ca(2+) efflux from preloaded vesicles. Additional assays performed in isolated mitochondria revealed that naproxen causes mitochondrial uncoupling and swelling in the presence of Ca(2+) ions. These effects were prevented by EGTA, ruthenium red and cyclosporin A, indicating that naproxen acts synergistically with Ca(2+) ions by promoting the mitochondrial permeability transition. The experimental results suggest that naproxen may impair the metabolic responses to Ca(2+)-dependent hormones acting by at least two mechanisms: (1) by interfering with the supply of external Ca(2+) through a direct action on the plasma membrane Ca(2+) influx, and (2) by affecting the refilling of the agonist-sensitive internal stores, including endoplasmic reticulum and mitochondria.
Collapse
|
125
|
Feltelius N, Ekbom A, Blomqvist P. Cancer incidence among patients with ankylosing spondylitis in Sweden 1965-95: a population based cohort study. Ann Rheum Dis 2003; 62:1185-8. [PMID: 14644856 PMCID: PMC1754397 DOI: 10.1136/ard.2002.004721] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Ankylosing spondylitis (AS) is a chronic inflammatory joint disease which may lead to extra-articular complications. The disease associated cancer risk has been poorly explored. Recently, tumour necrosis factor alpha blockers have been found to be efficacious in AS, but their long term risk is unknown. OBJECTIVE To analyse a large national cohort of patients with AS to determine the overall cancer risk in AS as a background to the future introduction of new treatments. METHODS All patients with AS admitted to Swedish hospitals 1965-95 were linked through individual national registration numbers to the Swedish Cancer Register and National Death Register. Standardised incidence ratio (SIR) of cancer risk was calculated in 6621 people, monitored during 67 885 person-years. RESULTS No overall increase in cancer risk was found (SIR 1.05, 95% CI 0.94 to 1.17). Rectal cancer was less common (SIR 0.41, 95% CI 0.15 to 0.89) while unspecified kidney cancer was more common (SIR 5.90, 95% CI 1.61 to 15.1). Risks for colon, renal parenchymal, and renal pelvic cancer were not significantly increased. Laryngeal cancer was more common than expected, while lung cancer was not. Risks of haematopoietic malignancies were not increased. CONCLUSIONS No overall increase in cancer risk was found. The decreased risk of rectal cancer might be due to local application of NSAIDs, and the increased risk of unspecified kidney cancer to frequent radiological pelvic examinations. If information on disease characteristics, including HLA-B27, was available for individual patients with cancer, risk-benefit analysis of long term effects of new immunomodulation treatment might be improved.
Collapse
Affiliation(s)
- N Feltelius
- Unit of Rheumatology, Department of Medicine at Karolinska Hospital, Karolinska Institutet, Stockholm, Sweden.
| | | | | |
Collapse
|
126
|
Smartt HJM, Elder DJE, Hicks DJ, Williams NA, Paraskeva C. Increased NF-kappaB DNA binding but not transcriptional activity during apoptosis induced by the COX-2-selective inhibitor NS-398 in colorectal carcinoma cells. Br J Cancer 2003; 89:1358-65. [PMID: 14520472 PMCID: PMC2394298 DOI: 10.1038/sj.bjc.6601266] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) inhibit colorectal neoplasia, an effect that is associated with their ability to induce apoptosis. Although NSAIDs have been reported to inhibit NF-κB, more recent studies show activation of NF-κB by NSAIDs. NF-κB commonly shows antiapoptotic activity and is implicated in the therapeutic resistance of cancer cells. The effects of highly COX-2-selective NSAIDs such as NS-398 on NF-κB in colorectal tumour cells have not been reported. Therefore, we addressed whether NF-κB has a role in NS-398-induced apoptosis of colorectal cancer cells. Treatment of HT-29 colorectal carcinoma cells with doses of NS-398 (50–75 μM) known to induce apoptosis had no effect on NF-κB for up to 48 h. However after 72 and 96 h NF-κB DNA-binding activity was increased by NS-398, in parallel with apoptosis induction. NS-398-treated HT-29 cells showed increased p50 homodimer binding and an induction of p50/p65 heterodimers, as demonstrated by supershift assay. However, although NS-398 increased NF-κB DNA binding it did not increase NF-κB-dependent reporter activity and inhibition of NF-κB DNA binding did not enhance NS-398-induced apoptosis. This indicates that NF-κB activated by NS-398 is transcriptionally inactive and is an encouraging result for the use of COX-2-selective NSAIDs not only in chemoprevention but also as novel therapies for colon cancer.
Collapse
Affiliation(s)
- H J M Smartt
- Cancer Research UK Colorectal Tumour Biology Research Group, Department of Pathology and Microbiology, School of Medical Sciences, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - D J E Elder
- Cancer Research UK Colorectal Tumour Biology Research Group, Department of Pathology and Microbiology, School of Medical Sciences, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - D J Hicks
- Cancer Research UK Colorectal Tumour Biology Research Group, Department of Pathology and Microbiology, School of Medical Sciences, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - N A Williams
- Department of Pathology and Microbiology, Division of Immunology, School of Medical Sciences, University of Bristol, University Walk, Bristol BS8 1TD, UK
| | - C Paraskeva
- Cancer Research UK Colorectal Tumour Biology Research Group, Department of Pathology and Microbiology, School of Medical Sciences, University of Bristol, University Walk, Bristol BS8 1TD, UK
- Cancer Research UK Colorectal Tumour Biology Research Group, Department of Pathology and Microbiology, School of Medical Sciences, University of Bristol, University Walk, Bristol BS8 1TD, UK. E-mail:
| |
Collapse
|
127
|
Akunda JK, Lao HC, Lee CA, Sessoms AR, Slade RM, Langenbach R. Genetic deficiency or pharmacological inhibition of cyclooxygenase‐1 or ‐2 induces mouse keratinocyte differentiation in vitro and in vivo. FASEB J 2003; 18:185-7. [PMID: 14630706 DOI: 10.1096/fj.02-1192fje] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Previously we demonstrated that genetic deficiency of the cyclooxygenases (COX-1 or COX-2) altered keratinocyte differentiation in mouse skin [Tiano et. al. (2002) Cancer Res. 62, 3395-3401]. In this study, we show that topical application of SC-560 (a COX-1 selective inhibitor) or celecoxib (COX-2 selective) to TPA-treated wild-type skin caused fivefold increases in the number of basal keratinocytes expressing the early differentiation marker keratin 1 (K1). In contrast to skin, COX-2 not COX-1 was the major isoform expressed in cultured primary keratinocytes. COX-1 was predominantly expressed in detached, differentiated cells, whereas COX-2 was found in the attached, proliferating cells. High Ca++ medium induced K1 and COX-1 in wild-type keratinocytes but did not change COX-2 expression. As observed in skin, COX-1-/- and COX-2-/- primary keratinocytes expressed fivefold more K1 than wild-type cells. K1 levels in cultured wild-type keratinocytes were also increased by treatment with celecoxib and indomethacin. However, unlike its in vivo effect, SC-560, possibly due to low COX-1 expression in cultured mouse keratinocytes, did not increase K1 levels. Furthermore, no increases in apoptotic cell numbers were observed in COX-deficient keratinocytes or COX-inhibitor treated wild-type cells. Thus, a major effect of COX inhibitors and COX-deficiency is the induction of keratinocyte differentiation.
Collapse
Affiliation(s)
- Jacqueline K Akunda
- Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | | | | | | | | | |
Collapse
|
128
|
Hull MA, Gardner SH, Hawcroft G. Activity of the non-steroidal anti-inflammatory drug indomethacin against colorectal cancer. Cancer Treat Rev 2003; 29:309-20. [PMID: 12927571 DOI: 10.1016/s0305-7372(03)00014-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A substantial body of evidence from rodent colon carcinogenesis models, in vitro experiments with human colorectal cancer cells and limited clinical observations in humans suggest that the non-steroidal anti-inflammatory drug indomethacin has anti-colorectal cancer activity. However, although many mechanisms of the anti-neoplastic activity of indomethacin have been suggested, e.g., cyclooxygenase inhibition and peroxisome proliferator-activated receptor gamma activation, the precise relevance of the majority of in vitro pharmacological observations to the in vivo anti-neoplastic activity of indomethacin remains unclear. Herein, we review the existing literature describing the chemopreventative and chemotherapeutic efficacy of indomethacin against colorectal cancer, and draw together the disparate literature describing potential mechanisms of action of indomethacin in human colorectal cancer cells in vitro. Although indomethacin itself has significant adverse effects, including serious upper gastrointestinal toxicity, the development of novel derivatives that may have an improved safety profile means that further investigation of the anti-colorectal cancer activity of indomethacin is warranted.
Collapse
Affiliation(s)
- M A Hull
- Molecular Medicine Unit, University of Leeds, Clinical Sciences Building, St. James's University Hospital, Leeds, LS9 7TF, UK.
| | | | | |
Collapse
|
129
|
Chang JK, Chuang LY, Ho ML, Wang GJ. Effects of nonsteroidal anti-inflammatory drugs on transforming growth factor-beta expression and bioactivity in rat osteoblast-enriched culture. Kaohsiung J Med Sci 2003; 19:278-88. [PMID: 12873036 DOI: 10.1016/s1607-551x(09)70474-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) have been reported to suppress bone remodeling in normal and repaired bones. Our previous results indicated that ketorolac and indomethacin suppressed proliferation, stimulated early differentiation, and induced apoptosis in cultured osteoblasts. Transforming growth factor-beta (TGF-beta) has been reported to enhance proliferation, suppress differentiation, and prevent apoptosis in osteoblasts. We proposed that one pathway of NSAID effects on osteoblast function might be through inhibition of the expression and/or bioactivity of TGF-beta in osteoblasts. We tested the effects of ketorolac and indomethacin on the expression of TGF-beta1 mRNA and protein and the bioactivity of TGF-beta in osteoblast-enriched cultures derived from fetal calvaria. The effects of prostaglandin E1 (PGE1) and PGE2 on TGF-beta expression and bioactivity were also examined in order to understand more about the role of prostaglandins in osteoblast function. Simultaneously, we estimated whether these NSAID effects on osteoblasts were prostaglandin-related. The results showed that 24-hour treatments with both PGEs and theoretic therapeutic concentrations of ketorolac and indomethacin had no significant effects on the levels of either transcription or translation of TGF-beta or the post-translational function of TGF-beta in osteoblasts. These results suggest that NSAIDs do not affect osteoblast function through changes in TGF-beta action in osteoblasts.
Collapse
Affiliation(s)
- Je-Ken Chang
- Department of Orthopedics, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | | | |
Collapse
|
130
|
Horrobin DF. A low toxicity maintenance regime, using eicosapentaenoic acid and readily available drugs, for mantle cell lymphoma and other malignancies with excess cyclin D1 levels. Med Hypotheses 2003; 60:615-23. [PMID: 12710892 DOI: 10.1016/s0306-9877(03)00075-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mantle cell lymphoma is a difficult to treat non-Hodgkin's lymphoma (NHL) whose biochemistry is unusually well characterised. Almost all and perhaps all patients overexpress the cyclin D1 protein which is crucial in driving cells from the G1 to the S phase. This overexpression may be responsible for the refractoriness. Despite this understanding, treatments for mantle cell lymphoma are based on standard NHL regimes of cyclophosphamide, doxorubicin, vincristine and prednisone, perhaps supplemented with the monoclonal antibody rituximab. There has never been any attempt to direct treatment to the cyclin D1 mechanism or to angiogenesis which is now known to be important in all lymphomas. Both these targets lend themselves to long-term maintenance regimes of relatively low toxicity which can be used as adjuvants to standard therapy. Agents which have recently been shown to block cyclin D1 translation by regulating calcium levels are the unsaturated essential fatty acid, eicosapentaenoic acid (EPA), the antidiabetic thiazolidinediones, and the antifungal agent, clotrimazole. Two types of agent which have been shown to inhibit angiogenesis are the teratogen, thalidomide, and the selective inhibitors of cyclo-oxygenase 2 (COX-2). Retinoids exert synergistic effects with EPA and have been shown to inhibit both tumour growth and angiogenesis. The mechanisms of action of these various agents are discussed, and specific suggestions are made for low toxicity maintenance therapy of mantle cell lymphoma and of other tumours which overexpress cyclin D1.
Collapse
|
131
|
Chen WS, Liu JH, Wei SJ, Liu JM, Hong CY, Yang WK. Colon cancer cells with high invasive potential are susceptible to induction of apoptosis by a selective COX-2 inhibitor. Cancer Sci 2003; 94:253-8. [PMID: 12824918 PMCID: PMC11160193 DOI: 10.1111/j.1349-7006.2003.tb01429.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2002] [Revised: 12/24/2002] [Accepted: 01/07/2003] [Indexed: 11/30/2022] Open
Abstract
Cyclooxygenase-2 (COX-2) expression has been shown to correlate with the invasiveness of colon cancer cells. To further investigate this positive correlation and its possible therapeutic implications, a selective COX-2 inhibitor, etodolac, was tested on three variants of HT-29 colon cancer cell lines, HT-29/Inv1, HT-29/Inv2 and HT-29/Inv3, with graded increases of in vitro Matrigel invasive potential and COX-2 expression levels. HT-29 variants with higher invasive potential were found to be more sensitive to etodolac by in vitro growth inhibition assays, the estimated LD(50) being 0.5 mM for highly invasive HT-29/Inv2 and HT-29/Inv3 cells, 0.6 mM for slightly less invasive HT-29/Inv1, and 1.8 mM for the parental HT-29. Treatment of the highly invasive HT-29/Inv2 and Inv3 variants with as little as 0.1 mM etodolac in the growth medium produced signs of apoptosis, as detected by DNA fragmentation and TUNEL (terminal deoxynucleotidyl transferase dUTP-biotin nick end labeling) assay. In vivo experiments in SCID mice showed that etolodac inhibited the growth of subcutaneous tumors induced by HT-29/Inv3 cells significantly more than those by the parental HT-29 cells. These results suggest that COX-2 inhibitors have a potential role in prevention of tumor invasion in colon cancer patients.
Collapse
Affiliation(s)
- Wei-Shone Chen
- Division of Colorectal Surgery, Veterans General Hospital-Taipei and National Yang-Ming University, Taiwan.
| | | | | | | | | | | |
Collapse
|
132
|
Ordan O, Rotem R, Jaspers I, Flescher E. Stress-responsive JNK mitogen-activated protein kinase mediates aspirin-induced suppression of B16 melanoma cellular proliferation. Br J Pharmacol 2003; 138:1156-62. [PMID: 12684272 PMCID: PMC1573760 DOI: 10.1038/sj.bjp.0705163] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
1. Available anticancer drugs do not seem to modify the prognosis of metastatic melanoma. Salicylate and acetyl salicylic acid (aspirin) were found to suppress growth in a number of transformed cells, that is, prostate and colon. Therefore, we studied the direct effects of aspirin on metastatic B16 melanoma cells. 2. Aspirin at a plasma-attainable and nontoxic level suppressed the proliferation of B16 cells. 3. Aspirin induced the activation of p38 and c-Jun N-terminal kinase (JNK) mitogen-activated protein kinases. 4. Inhibition of JNK, but not p38, decreased the suppressive effect of aspirin upon the proliferation of B16 cells. 5. The aspirin-induced reduction in B16 proliferation was cumulative over time. 6. Aspirin and the chemotherapeutic drug 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) induced B16 cell death synergistically. 7. In addition to the murine B16 cell line, the proliferation of SK-28 human melanoma cells was also suppressed by aspirin. 8 In conclusion, aspirin suppresses the proliferation of metastatic B16 cells in a JNK-dependent mechanism.
Collapse
Affiliation(s)
- Orly Ordan
- Department of Human Microbiology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ronit Rotem
- Department of Human Microbiology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ilona Jaspers
- Center for Environmental Medicine and Lung Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7310, U.S.A
| | - Eliezer Flescher
- Department of Human Microbiology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
- Author for correspondence:
| |
Collapse
|
133
|
Steele VE, Hawk ET, Viner JL, Lubet RA. Mechanisms and applications of non-steroidal anti-inflammatory drugs in the chemoprevention of cancer. Mutat Res 2003; 523-524:137-44. [PMID: 12628511 DOI: 10.1016/s0027-5107(02)00329-9] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Biological and chemical irritants can be the cause of irritation in a variety of organ sites. It is becoming well understood that chronic irritation in any form can initiate and accelerate the cancer process in these same organs. This understanding comes in part from the many epidemiologic studies which point out that chronic inflammation correlates with increased risk of developing cancer in that organ which is affected. One of the hallmarks of chronic irritation is the increased activity in the arachidonic acid pathway which provides many of the necessary inflammatory biochemical mediators to this process. Arachidonic acid metabolism diverges down two main pathways, the cyclooxygenase (COX) and the lipoxygenase (LOX) pathways. The COX pathway leads to prostaglandin and thromboxane production and the LOX pathway leads to the leukotrienes (LTs) and hydroxyeicosatetraenoic acids (HETEs). These classes of inflammatory molecules exert profound biological effects which enhance the development and progression of human cancers. A large number of synthetic drugs and natural products have been discovered that block many of these key pathways. Much experimental evidence in animals has shown that inhibition of the key enzymes which drive these pathways can, in fact, prevent, slow or reverse the cancer process. The data are convincing in a number of organ sites including colon, breast, lung, bladder and skin. More recently, double-blinded randomize clinical trials in humans have shown the prevention of colonic polyps by anti-inflammatory agents. These studies have primarily used non-steroidal anti-inflammatory drugs (NSAIDS) which block the COX pathways. Recent preclinical studies indicate that the LOX pathway also may be an important target for cancer prevention strategy. The expression of high levels of these enzymes in cancerous tissues make them an obvious first target for cancer prevention strategies. As newer more specific drugs are developed with few adverse effects this important prevention strategy may become a reality.
Collapse
Affiliation(s)
- Vernon E Steele
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892-7322, USA.
| | | | | | | |
Collapse
|
134
|
Kim KY, Seol JY, Jeon GA, Nam MJ. The combined treatment of aspirin and radiation induces apoptosis by the regulation of bcl-2 and caspase-3 in human cervical cancer cell. Cancer Lett 2003; 189:157-66. [PMID: 12490308 DOI: 10.1016/s0304-3835(02)00519-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The antitumor mechanisms mediated by combined treatment of aspirin and radiation on human cervical cancer cells are unclear. In this paper, we studied whether aspirin and radiation induced apoptosis and whether the sensitivity to radiation was enhanced in aspirin-pretreated HeLa TG, human cervical cancer cells. We identified the regulation of apoptosis-responsive genes, bcl-2, caspase-3 and p53 after combined treatment. To investigate the growth inhibitory effect on HeLa TG cells after treatment of various nonsteroidal anti-inflammatory drugs (NSAIDs), we performed cell proliferation assay and colony-forming assay. In the presence of aspirin, sulindac and indomethacin, cell proliferation and colony formation were decreased in a time- and dose-dependent manner. According to flow cytometry analysis and Hoechst 33342 staining, we found that aspirin increased sub-G1 population and nuclear condensation of cervical cancer cells. Remarkably, the combined treatment decreased cell proliferation compared with treatment of 1 mM aspirin or 6 Gy radiation alone. Pretreatment of aspirin followed by irradiation also elevated the population of apoptotic cells. These results revealed that sensitivity to radiation was enhanced in aspirin-pretreated HeLa TG cells, and aspirin has the additive role for amplifying the radiotherapeutic effect in cervical cancer cells. Finally, combined treatment revealed bcl-2 repression and caspase-3 induction did not detect any change but p53 expression did. We have demonstrated that combined treatment of aspirin and radiation induces the antitumor effect mediated by bcl-2 and caspase-3 pathway in cervical cancer cells.
Collapse
Affiliation(s)
- Kye Young Kim
- Department of Biomedical Sciences, National Institute of Health, Seoul 122-701, South Korea.
| | | | | | | |
Collapse
|
135
|
Kim HJ, Wu HG, Park IA, Ha SW. High cyclooxygenase-2 expression is related with distant metastasis in cervical cancer treated with radiotherapy. Int J Radiat Oncol Biol Phys 2003; 55:16-20. [PMID: 12504031 DOI: 10.1016/s0360-3016(02)03821-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE The cyclooxygenase (COX)-2 enzyme has been shown to have an important role in carcinogenesis and apoptosis in various types of cancer. The purpose of this study was to evaluate the relationship between local recurrence or distant metastasis and COX-2 expression and apoptosis in cervical cancer patients treated with radical radiotherapy (RT). METHODS AND MATERIALS Twenty-two patients who were diagnosed with cervical cancer were enrolled in this study. All patients were treated with radical RT (external beam RT plus brachytherapy) at Seoul National University Hospital. The formalin-fixed, paraffin-embedded tissues of 11 patients who developed local recurrence (n = 3) or distant metastasis (n = 8) were compared with those of other patients who were disease free. Prognostic factors, including tumor size, lymph node metastatic status, and stage, were well balanced between the two groups. COX-2 expression was determined immunohistochemically, and apoptosis was assessed using in situ DNA nick end labeling (TUNEL)-based methods. RESULTS COX-2 expression was stronger in the local recurrence and distant metastasis patients than in those free of disease. COX-2 expression was shown to have a statistically significant influence on treatment failure by the Mann-Whitney U test (p = 0.015) and the Mantel-Haenszel chi-square test (p = 0.015), but its distribution did not correlate with apoptosis. Among the clinicopathologic factors, including stage, lymph node metastatic status, and tumor size, lymph node metastatic status was found to closely correlate with COX-2 expression by the Mann-Whitney U test (p = 0.045) and Mantel-Haenszel chi-square test (p = 0.065). CONCLUSION COX-2 is believed to be one of the important factors associated with lymph node involvement and treatment failure. Our results suggest that inhibiting COX-2 may decrease treatment failure in cervical cancer treated with RT, and that COX-2 inhibitor administration may play an adjuvant role in cervical cancer treatment.
Collapse
Affiliation(s)
- Hak Jae Kim
- Department of Therapeutic Radiology, Seoul National University College of Medicine, Seoul, South Korea
| | | | | | | |
Collapse
|
136
|
Affiliation(s)
- Yaser Rayyan
- Division of Gastroenterology, New York Medical College, NY, USA
| | | | | |
Collapse
|
137
|
Mølck AM, Poulsen M, Meyer O. The combination of 1alpha,25(OH2)-vitamin D3, calcium and acetylsalicylic acid affects azoxymethane-induced aberrant crypt foci and colorectal tumours in rats. Cancer Lett 2002; 186:19-28. [PMID: 12183071 DOI: 10.1016/s0304-3835(02)00285-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Effects of 1alpha,25(OH)(2)-vitamin D(3) and acetylsalicylic acid at various dietary levels of calcium (CaCO(3)) on development of aberrant crypt foci (ACF) and tumours in colon were examined in groups of 16 male F344 rats initiated with azoxymethane and observed for 16 weeks. Calcium was the most potent modulator of ACF development. The total number of ACF increased with low calcium and decreased with high calcium. The number of large ACF decreased with any addition of calcium, acetylsalicylic acid and 1alpha,25(OH)(2)-vitamin D(3). High levels of calcium alone or in combination with 1alpha,25(OH)(2)-vitamin D(3) increased the incidence of tumour-bearing animals. 1alpha,25(OH)(2)-vitamin D(3) and acetylsalicylic acid at 5,000 ppm calcium increased the incidence as well.
Collapse
Affiliation(s)
- Anne-Marie Mølck
- Institute of Food Safety and Toxicology, The Danish Veterinary and Food Administration, Mørkhøj Bygade 19, DK-2860 Søborg, Denmark.
| | | | | |
Collapse
|
138
|
Yamazaki R, Kusunoki N, Matsuzaki T, Hashimoto S, Kawai S. Aspirin and sodium salicylate inhibit proliferation and induce apoptosis in rheumatoid synovial cells. J Pharm Pharmacol 2002; 54:1675-9. [PMID: 12542898 DOI: 10.1211/002235702261] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Aspirin has been reported to induce apoptosis in a variety of cell lines. In this study, we examined whether aspirin and sodium salicylate inhibit cell growth and induce apoptosis in rheumatoid synovial cells. Synovial cells were obtained from patients with rheumatoid arthritis, and the cells were treated with aspirin or sodium salicylate (0.1-10 mM) for 24 h. Cell proliferation and viability were assessed by 5-bromo-2'-deoxyuridine incorporation and by 4-[3-(4-iodophenyl)-2-(4-nitrophenyl)-2H-5-tetrazolio]-1,3-benzene disulfonate (WST-1) assay, respectively. The apoptosis of synovial cells was identified by DNA fragmentation assay and terminal deoxynucleotidyl transferase-mediated dUTP nick end labelling (TUNEL) assay. Aspirin and sodium salicylate suppressed the proliferation (IC50 (concentration causing 50% inhibition of cell proliferation): 2.1 and 1.2 mM, respectively) and reduced the viability (IC50: 2.0 and 1.4 mM, respectively) of synovial cells in a concentration-dependent manner at 0.3-10 mM. Furthermore, they induced DNA fragmentation and increased the number of TUNEL-positive synovial cells. These results suggest that aspirin and sodium salicylate can inhibit the proliferation of rheumatoid synovial cells through induction of apoptosis.
Collapse
Affiliation(s)
- Ryuta Yamazaki
- Institute of Medical Science, St Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki-shi, Kanagawa 216-8512, Japan
| | | | | | | | | |
Collapse
|
139
|
Yamazaki R, Kusunoki N, Matsuzaki T, Hashimoto S, Kawai S. Selective cyclooxygenase-2 inhibitors show a differential ability to inhibit proliferation and induce apoptosis of colon adenocarcinoma cells. FEBS Lett 2002; 531:278-84. [PMID: 12417326 DOI: 10.1016/s0014-5793(02)03535-4] [Citation(s) in RCA: 109] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although the influence of selective cyclooxygenase (COX)-2 inhibitors on the proliferation of colon adenocarcinoma cells have been the subject of much investigation, relatively little research has compared the effects of different COX-2 inhibitors. Celecoxib strongly suppressed the proliferation of COX-2 expressing HT-29 cells at 10-40 microM. NS-398 and nimesulide also inhibited cell proliferation, whereas rofecoxib, meloxicam, and etodolac did not. Only celecoxib induced apoptosis of HT-29 cells, as detected on the basis of DNA fragmentation, TUNEL positivity, and caspase-3/7 activation. DNA fragmentation was also increasd in COX-2 non-expressing cell lines (SW-480 and HCT-116) by exposure to celecoxib for 6-24 h. All six COX-2 inhibitors suppressed the production of prostaglandin E(2) by HT-29 cells, suggesting that the pro-apoptotic effect of celecoxib was unrelated to inhibition of COX-2. Inactivation of Akt might explain the differential pro-apoptotic effect of these selective COX-2 inhibitors on colon adenocarcinoma cells.
Collapse
Affiliation(s)
- Ryuta Yamazaki
- Institute of Medical Science, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki-shi, Kanagawa, Japan
| | | | | | | | | |
Collapse
|
140
|
Irani J, Ravery V, Pariente JL, Chartier-Kastler E, Lechevallier E, Soulie M, Chautard D, Coloby P, Fontaine E, Bladou F, Desgrandchamps F, Haillot O. Effect of nonsteroidal anti-inflammatory agents and finasteride on prostate cancer risk. J Urol 2002; 168:1985-8. [PMID: 12394690 DOI: 10.1016/s0022-5347(05)64277-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE We examine the relationship of nonsteroidal anti-inflammatory drugs and finasteride on the risk of prostate cancer. MATERIALS AND METHODS Participants in this case control study using a prospective collection of data were drawn from consecutive patients who underwent prostate biopsy at 12 different departments of urology from January 1999 to June 2000. Medication use was assessed by self-questionnaire as well as questions about dietary and lifestyle factors that might be relevant for prostate cancer risk. RESULTS The study included 639 patients with prostate cancer and 659 cancer-free controls. Univariate analysis showed no significant impact of aspirin and finasteride on prostate cancer risk while the nonaspirin nonsteroidal anti-inflammatory drug users had a lower risk (OR 0.80, 95% CI 0.64-0.99). After adjusting for potential confounders, the protective effect of nonaspirin nonsteroidal anti-inflammatory drugs was no longer significant (OR, 0.84, 95% CI 0.66-1.07), while finasteride showed a significant protective effect (OR 0.58, 95% CI 0.37-0.92). CONCLUSIONS The results suggest that finasteride could have a chemopreventive role in prostate cancer. While aspirin did not show any impact on prostate cancer risk, the role of nonaspirin nonsteroidal anti-inflammatory drugs warrants further studies.
Collapse
Affiliation(s)
- J Irani
- University Hospital of Poitiers, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Effect of Nonsteroidal Anti-Inflammatory Agents and Finasteride on Prostate Cancer Risk. J Urol 2002. [DOI: 10.1097/00005392-200211000-00020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
142
|
Pasinetti GM. From cyclooxygenase activities to Alzheimer's disease neuropathology:experimental approaches and therapeutic interventions. Drug Dev Res 2002. [DOI: 10.1002/ddr.10096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
143
|
Shah T, Ryu S, Lee HJ, Brown S, Kim JH. Pronounced radiosensitization of cultured human cancer cells by COX inhibitor under acidic microenvironment. Int J Radiat Oncol Biol Phys 2002; 53:1314-8. [PMID: 12128134 DOI: 10.1016/s0360-3016(02)02889-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE To demonstrate the influence of pH on the cytotoxicity and radiosensitization by COX (cyclooxygenase) -1 and -2 inhibitors using established human cancer cells in culture. METHODS AND MATERIALS Nonselective COX inhibitor, ibuprofen (IB), and selective COX-2 inhibitor, SC-236, were used to determine the cytotoxicity and radiosensitization at varying pH of culture media. Human colon carcinoma cell line (HT-29) was exposed to the drug alone and in combination with radiation at different pH of the cell culture media. The end point was clonogenic ability of the single-plated cells after the treatment. RESULTS Cytotoxicity and radiosensitization of IB increased with higher drug concentration and longer exposure time. The most significant radiosensitization was seen with IB (1.5 mM) for 2-h treatment at pH 6.7 before irradiation. The dose-modifying factor as defined by the ratio of radiation doses required to achieve the same effect on cell survival was 1.8 at 10% survival level. In contrast, SC-236 (50 microM for 2-8 h) showed no pH-dependent cytotoxicity. There was modest increase in the cell killing at lower doses of radiation. CONCLUSION An acidic pH was an important factor affecting the increased cytotoxicity and radiosensitization by ibuprofen. Radiation response was enhanced at shoulder portion of the cell survival curve by selective COX-2 inhibitor.
Collapse
Affiliation(s)
- Tushar Shah
- Department of Radiation Oncology, Henry Ford Hospital, Detroit, MI 48202, USA
| | | | | | | | | |
Collapse
|
144
|
Yamazaki R, Kusunoki N, Matsuzaki T, Hashimoto S, Kawai S. Nonsteroidal anti-inflammatory drugs induce apoptosis in association with activation of peroxisome proliferator-activated receptor gamma in rheumatoid synovial cells. J Pharmacol Exp Ther 2002; 302:18-25. [PMID: 12065695 DOI: 10.1124/jpet.302.1.18] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) have been reported to induce apoptosis in a variety of cell lines. In this study, we examined the effect of NSAIDs on the growth and apoptosis of synovial cells from patients with rheumatoid arthritis and analyzed the activation of peroxisome proliferator-activated receptor gamma (PPARgamma) as a possible mechanism of action of NSAIDs. Cell proliferation and viability were assessed from 5-bromo-2'-deoxyuridine incorporation and by 4-[3-(4-iodophenyl)-2-(4-nitrophenyl)-2H-5-tetrazolio]-1,3-benzene disulfonate (WST-1) assay, respectively. The apoptosis of synovial cells was identified by DNA fragmentation assay and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assay. Indometacin, diclofenac, oxaprozin, and zaltoprofen reduced cell proliferation and induced apoptotic cell death in synovial cells, whereas ketoprofen and acetaminophen did not. N-[2-(cyclohexyloxyl)-4-nitrophenyl]-methanesulfonamide (NS-398), a selective cyclooxygenase-2 inhibitor, also inhibited cell proliferation, whereas it did not cause apoptosis. Rheumatoid synovial cells expressed PPARgamma mRNA, and the PPARgamma ligands 15-deoxy-Delta(12,14)-prostaglandin J(2) and troglitazone reduced the proliferation and induced apoptosis in synovial cells. Luciferase reporter assay demonstrated that not only PPARgamma ligands but also NSAIDs, which could induce apoptosis, increased the activation of PPARgamma in synovial cells. Furthermore, the ability of NSAIDs and PPARgamma ligands to stimulate the activation of PPARgamma correlated with their ability to decrease cell viability(r = 0.92, p < 0.01) and ability to induce DNA fragmentation (r = 0.97, p < 0.001) in synovial cells. These results suggest that PPARgamma is an attractive target for induction of apoptosis in rheumatoid synovial cells and that the activation of the PPARgamma pathway is associated with the apoptotic action of NSAIDs.
Collapse
Affiliation(s)
- Ryuta Yamazaki
- Institute of Medical Science, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki-shi, Kanagawa 216-8512, Japan
| | | | | | | | | |
Collapse
|
145
|
Menzel T, Schauber J, Kreth F, Kudlich T, Melcher R, Gostner A, Scheppach W, Lührs H. Butyrate and aspirin in combination have an enhanced effect on apoptosis in human colorectal cancer cells. Eur J Cancer Prev 2002; 11:271-81. [PMID: 12131661 DOI: 10.1097/00008469-200206000-00011] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Laboratory and epidemiological studies suggest that butyrate, a metabolic product of microbial fermentation of dietary fibre, and aspirin, a non-steroidal antiphlogistic drug, both reduce the risk of developing colon cancer. Notably, few data exist on potential interactions of these two substances. In this study, the effects of a butyrate-aspirin combination on human colon cancer cells were compared with treatment with aspirin or butyrate alone. Both substances decreased proliferation and induced differentiation and apoptosis. Butyrate reduced mutant p53 expression, whereas aspirin did not affect p53 expression. Butyrate-induced apoptosis correlated with an increase in Bak expression and a decrease in the expression of Bcl-XL. Aspirin had no effect on the investigated apoptosis-controlling factors. The antiproliferative and pro-apoptotic effects of the butyrate-aspirin combination were markedly enhanced. The combination resulted in a stronger decrease in the expression of PCNA and cdk2. Our data suggest that the anticarcinogenic effect of aspirin might effectively be augmented by combination with the short-chain fatty acid butyrate.
Collapse
Affiliation(s)
- T Menzel
- Division of Gastroenterology, Department of Medicine, University of Würzburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
146
|
Ricchi P, Di Matola T, Ruggiero G, Zanzi D, Apicella A, di Palma A, Pensabene M, Pignata S, Zarrilli R, Acquaviva AM. Effect of non-steroidal anti-inflammatory drugs on colon carcinoma Caco-2 cell responsiveness to topoisomerase inhibitor drugs. Br J Cancer 2002; 86:1501-9. [PMID: 11986787 PMCID: PMC2375372 DOI: 10.1038/sj.bjc.6600289] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2001] [Revised: 02/27/2002] [Accepted: 03/03/2002] [Indexed: 11/26/2022] Open
Abstract
Numerous studies demonstrate that the chemopreventive effect of non-steroidal anti-inflammatory drugs on colon cancer is mediated through inhibition of cell growth and induction of apoptosis. For these effects non-steroidal anti-inflammatory drugs have been recently employed as sensitising agents in chemotherapy. We have shown previously that treatments with aspirin and NS-398, a cyclo-oxygenase-2 selective inhibitor, affect proliferation, differentiation and apoptosis of the human colon adenocarcinoma Caco-2 cells. In the present study, we have evaluated the effects of aspirin and NS-398 non-steroidal anti-inflammatory drugs on sensitivity of Caco-2 cells to irinotecan (CPT 11) and etoposide (Vp-16) topoisomerase poisons. We find that aspirin co-treatment is able to prevent anticancer drug-induced toxicity, whereas NS-398 co-treatment poorly affects anticancer drug-induced apoptosis. These effects correlate with the different ability of aspirin and NS-398 to interfere with cell cycle during anticancer drug co-treatment. Furthermore, aspirin treatment is associated with an increase in bcl-2 expression, which persists in the presence of the anticancer drugs. Our data indicate that aspirin, but not NS-398, determines a cell cycle arrest associated with death suppression. This provides a plausible mechanism for the inhibition of apoptosis and increase in survival observed in anticancer drug and aspirin co-treatment.
Collapse
Affiliation(s)
- P Ricchi
- Dipartimento di Biologia e Patologia Cellulare e Molecolare L. Califano, Istituto di Endocrinologia ed Oncologia Sperimentale G. Salvatore del Consiglio Nazionale delle Ricerche, Università Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Chen X, Wan Y, Bai Q, Zhang W, Zhu H. Sodium salicylate-triggered apoptosis in HL-60 cells depends on caspase-8 activation. Int J Hematol 2002; 75:407-11. [PMID: 12041673 DOI: 10.1007/bf02982133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
For investigation of the killing and proapoptotic effects of sodium salicylate (Na-Sal) on HL-60 cells, the cytotoxic activity of Na-Sal was measured by means of MTT assay. Apoptosis was identified and analyzed with the help of transmission electron microscopy, annexin V staining, and DNA gel electrophoresis, and the association of caspase-8 activation with apoptosis was determined with the specific protease inhibitor IETD-fmk. After exposure of HL-60 cells to increasing concentrations of Na-Sal (0.5, 1, 3, 5, and 7 mmol/L) for 24 hours, the mean cell viability gradually dropped to 92%, 83%, 68%, 50%, and 42%. With treatment of target cells with 5-mmol/L (IC50) Na-Sal for 6, 12, 24, or 36 hours, the mean cell survival tapered to 91%, 81%, 48% (P <.05 versus control), and 14% (P <.05 versus control). Again incubated with 5-mmol/L Na-Sal for 12 or 24 hours, HL-60 cells displayed clear early or late signs of apoptosis, including (1) notable enhancement of phosphatidylserine externalization, (2) cell shrinkage, membrane blebbing, and eventual disintegration into numerous apoptotic bodies, and (3) formation of ladder DNA. The viability of HL-60 cells increased significantly during 24 or 36 hours of coculture with 100-micromol/L IETD-fmk in combination with 5-mmol/L Na-Sal compared with the viability when 5-mmol/L Na-Sal was used alone (P < .05). Moreover, the target cells showed a considerable decrease in phosphatidylserine exposure and DNA fragmentation after coincubation for 12 or 24 hours performed as described above. The findings presented herein strongly suggest that Na-Sal can exert potent killing and proapoptotic activity against HL-60 cells, and this effect appears to depend on caspase-8 activation.
Collapse
Affiliation(s)
- Xiequn Chen
- Department of Hematology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | | | | | | | | |
Collapse
|
148
|
Li HL, Chen DD, Li XH, Zhang HW, Lü JH, Ren XD, Wang CC. JTE-522-induced apoptosis in human gastric adenocarinoma cell line AGS cells by caspase activation accompanying cytochrome C release, membrane translocation of Bax and loss of mitochondrial membrane potential. World J Gastroenterol 2002; 8:217-23. [PMID: 11925595 PMCID: PMC4658354 DOI: 10.3748/wjg.v8.i2.217] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the role of the mitochondrial pathway in JTE-522-induced apoptosis and to investigate the relationship between cytochrome C release, caspase activity and loss of mitochondrial membrane potential (△Ψm).
METHODS: Cell culture, cell counting, ELISA assay, TUNEL, flow cymetry, Western blot and fluorometric assay were employed to investigate the effect of JTE-522 on cell proliferation and apoptosis in AGS cells and related molecular mechanism.
RESULTS: JTE-522 inhibited the growth of AGS cells and induced the apoptosis. Caspases 8 and 9 were activated during apoptosis as judged by the appearance of cleavage products from procaspase and the caspase activities to cleave specific fluorogenic substrates. To elucidate whether the activation of caspases 8 and 9 was required for the apoptosis induction, we examined the effect of caspase-specific inhibitors on apoptosis. The results showed that caspase inhibitors significantly inhibited the apoptosis induced by JTE-522. In addition, the membrane translocation of Bax and cytosolic release of cytochrome C accompanying with the decrease of the uptake of Rhodamin 123, were detected at an early stage of apoptosis. Furthermore, Bax translocation, cytochrome C release, and caspase 9 activation were blocked by Z-VAD.fmk and Z-IETD-CHO.
CONCLUSION: The present data indicate a crucial association between activation of caspases 8, 9, cytochrome C release, membrane translocation of Bax, loss of △Ψm and JTE-522-induced apoptosis in AGS cells.
Collapse
Affiliation(s)
- Hong-Liang Li
- Department of Pharmacology, Jinan University Pharmacy College, Guangzhou 510632, Guangdong Province, China.
| | | | | | | | | | | | | |
Collapse
|
149
|
Fingrut O, Flescher E. Plant stress hormones suppress the proliferation and induce apoptosis in human cancer cells. Leukemia 2002; 16:608-16. [PMID: 11960340 DOI: 10.1038/sj.leu.2402419] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2001] [Accepted: 10/08/2001] [Indexed: 01/04/2023]
Abstract
Cellular stressors induce various outcomes including inhibition of cell proliferation and cell death. Sodium salicylate (SA), a plant stress hormone, can suppress the proliferation or cause apoptosis in certain mammalian cancer cells. Plant stress hormones are activators of cellular responses, including cell death, to diverse stress situations in plants. Thus, we hypothesized that plant stress hormones share the ability to adversely affect cancer cells. We found that the plant stress hormone SA suppressed proliferation of lymphoblastic leukemia, prostate, breast and melanoma human cancer cells. Jasmonic acid (JA), a plant stress hormone belonging to the Jasmonate family, induced death in lymphoblastic leukemia cells and caused suppression of cell proliferation in the other human cancer cells mentioned above. Another member of the Jasmonate family, methyl jasmonate (MJ), induced death in each of the cell lines. Plant stress hormones did not affect normal human lymphocytes, in contrast to their strong effect on lymphoblastic leukemia cells. JA and MJ caused apoptotic death, as determined by characteristic nuclear morphology, flow cytometric DNA profile and elevation of caspase-3 activity. Finally, mice bearing EL-4 lymphoma and treated with MJ, survived for significantly (P = 0.00953) longer periods of time than untreated mice. These findings suggest that plant stress hormones may potentially be a novel class of anti-cancer drugs.
Collapse
Affiliation(s)
- O Fingrut
- Department of Human Microbiology, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | | |
Collapse
|
150
|
Fehlberg S, Trautwein S, Göke A, Göke R. Bisphenol A diglycidyl ether induces apoptosis in tumour cells independently of peroxisome proliferator-activated receptor-gamma, in caspase-dependent and -independent manners. Biochem J 2002; 362:573-8. [PMID: 11879183 PMCID: PMC1222420 DOI: 10.1042/0264-6021:3620573] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear transcription factors which are involved in many biological processes, such as regulation of cell differentiation, lipid metabolism, inflammation and cell death. PPARs consist of three families, PPAR-alpha, PPAR-delta and PPAR-gamma. Bisphenol A diglycidyl ether (BADGE) has been described as a pure antagonist of PPAR-gamma. However, recent data also revealed PPAR-gamma-agonistic activities of BADGE. Here we show that BADGE kills transformed cells by apoptosis and promotes the cytotoxic effects of tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) and indomethacin. The cytotoxic effect of BADGE does not require PPAR-gamma expression and is mediated in caspase-dependent and caspase-independent manners.
Collapse
Affiliation(s)
- Sebastian Fehlberg
- Clinical Research Unit for Gastrointestinal Endocrinology, University of Marburg, 35033 Marburg, Germany
| | | | | | | |
Collapse
|