51
|
Jung KH, Chu K, Lee ST, Yoon HJ, Chang JY, Nam WS, Yoon SH, Cho JY, Yu KS, Jang IJ, Kim M, Lee SK, Roh JK. Prolonged Use of Aspirin Alters Human and Rat Intestinal Cells and Thereby Limits the Absorption of Clopidogrel. Clin Pharmacol Ther 2011; 90:612-9. [DOI: 10.1038/clpt.2011.163] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
52
|
Kim IY, Park SY, Kang Y, Thapa D, Choi HG, Kim JA. Role of nonsteroidal anti-inflammatory drug-activated gene-1 in docetaxel-induced cell death of human colorectal cancer cells with different p53 status. Arch Pharm Res 2011; 34:323-30. [DOI: 10.1007/s12272-011-0219-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 11/10/2010] [Accepted: 11/18/2010] [Indexed: 12/21/2022]
|
53
|
Lin L, Tan RX. Cross-kingdom actions of phytohormones: a functional scaffold exploration. Chem Rev 2011; 111:2734-60. [PMID: 21250668 DOI: 10.1021/cr100061j] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Lan Lin
- Institute of Functional Biomolecules, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, P. R. China
| | | |
Collapse
|
54
|
Bathaie SZ, Nikfarjam L, Rahmanpour R, Moosavi-Movahedi AA. Spectroscopic studies of the interaction of aspirin and its important metabolite, salicylate ion, with DNA, A·T and G·C rich sequences. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2010; 77:1077-1083. [PMID: 20869297 DOI: 10.1016/j.saa.2010.08.078] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Revised: 08/01/2010] [Accepted: 08/26/2010] [Indexed: 05/29/2023]
Abstract
Among different biological effects of acetylsalicylic acid (ASA), its anticancer property is controversial. Since ASA hydrolyzes rapidly to salicylic acid (SA), especially in the blood, interaction of both ASA and SA (as the small molecules) with ctDNA, oligo(dA·dT)15 and oligo(dG·dC)15, as a possible mechanism of their action, is investigated here. The results show that the rate of ASA hydrolysis in the absence and presence of ctDNA is similar. The spectrophotometric results indicate that both ASA and SA cooperatively bind to ctDNA. The binding constants (K) are (1.7±0.7)×10(3) M(-1) and (6.7±0.2)×10(3) M(-1) for ASA and SA, respectively. Both ligands quench the fluorescence emission of ethidium bromide (Et)-ctDNA complex. The Scatchard plots indicate the non-displacement based quenching (non-intercalative binding). The circular dichroism (CD) spectra of ASA- or SA-ctDsNA complexes show the minor distortion of ctDNA structure, with no characteristic peaks for intercalation of ligands. Tm of ctDNA is decreased up to 3°C upon ASA binding. The CD results also indicate more distortions on oligo(dG·dC)15 structure due to the binding of both ASA and SA in comparison with oligo(dA·dT)15. All data indicate the more affinity for SA binding with DNA minor groove in comparison with ASA which has more hydrophobic character.
Collapse
Affiliation(s)
- S Z Bathaie
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box: 14115-111, Tehran, Iran.
| | | | | | | |
Collapse
|
55
|
Hawcroft G, Loadman PM, Belluzzi A, Hull MA. Effect of eicosapentaenoic acid on E-type prostaglandin synthesis and EP4 receptor signaling in human colorectal cancer cells. Neoplasia 2010; 12:618-27. [PMID: 20689756 PMCID: PMC2915406 DOI: 10.1593/neo.10388] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Revised: 04/30/2010] [Accepted: 05/03/2010] [Indexed: 11/18/2022]
Abstract
The omega-3 polyunsaturated fatty acid eicosapentaenoic acid (EPA), in the free fatty acid (FFA) form, has been demonstrated to reduce adenoma number and size in patients with familial adenomatous polyposis. However, the mechanistic basis of the antineoplastic activity of EPA in the colorectum remains unclear. We tested the hypothesis that EPA-FFA negatively modulates synthesis of and signaling by prostaglandin (PG) E(2) in human colorectal cancer (CRC) cells. EPA-FFA induced apoptosis of cyclooxygenase (COX)-2-positive human HCA-7 CRC cells in vitro. EPA-FFA in cell culture medium was incorporated rapidly into phospholipid membranes of HCA-7 human CRC cells and acted as a substrate for COX-2, leading to reduced synthesis of PGE(2) and generation of PGE(3). Alone, PGE(3) bound and activated the PGE(2) EP4 receptor but with reduced affinity and efficacy compared with its "natural" ligand PGE(2). However, in the presence of PGE(2), PGE(3) acted as an antagonist of EP4 receptor-dependent 3',5' cyclic adenosine monophosphate induction in naturally EP4 receptor-positive LoVo human CRC cells and of resistance to apoptosis in HT-29-EP4 human CRC cells overexpressing the EP4 receptor. We conclude that EPA-FFA drives a COX-2-dependent "PGE(2)-to-PGE(3) switch" in human CRC cells and that PGE(3) acts as a partial agonist at the PGE(2) EP4 receptor.
Collapse
Affiliation(s)
- Gillian Hawcroft
- Leeds Institute of Molecular Medicine, St James's University Hospital, Leeds, United Kingdom.
| | | | | | | |
Collapse
|
56
|
Singh A, Rathaur S. Combination of DEC plus aspirin induced mitochondrial mediated apoptosis in filarial parasite Setaria cervi. Biochimie 2010; 92:894-900. [DOI: 10.1016/j.biochi.2010.03.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2009] [Accepted: 03/24/2010] [Indexed: 02/01/2023]
|
57
|
Ettarh R, Cullen A, Calamai A. NSAIDs and Cell Proliferation in Colorectal Cancer. Pharmaceuticals (Basel) 2010; 3:2007-2021. [PMID: 27713339 PMCID: PMC4036654 DOI: 10.3390/ph3072007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Revised: 05/17/2010] [Accepted: 06/22/2010] [Indexed: 12/21/2022] Open
Abstract
Colon cancer is common worldwide and accounts for significant morbidity and mortality in patients. Fortunately, epidemiological studies have demonstrated that continuous therapy with NSAIDs offers real promise of chemoprevention and adjunct therapy for colon cancer patients. Tumour growth is the result of complex regulation that determines the balance between cell proliferation and cell death. How NSAIDs affect this balance is important for understanding and improving treatment strategies and drug effectiveness. NSAIDs inhibit proliferation and impair the growth of colon cancer cell lines when tested in culture in vitro and many NSAIDs also prevent tumorigenesis and reduce tumour growth in animal models and in patients, but the relationship to inhibition of tumour cell proliferation is less convincing, principally due to gaps in the available data. High concentrations of NSAIDs are required in vitro to achieve cancer cell inhibition and growth retardation at varying time-points following treatment. However, the results from studies with colon cancer cell xenografts are promising and, together with better comparative data on anti-proliferative NSAID concentrations and doses (for in vitro and in vivo administration), could provide more information to improve our understanding of the relationships between these agents, dose and dosing regimen, and cellular environment.
Collapse
Affiliation(s)
- Raj Ettarh
- School of Medicine & Medical Science, University College Dublin, C206 Health Sciences Building, Belfield, Dublin 4, Ireland.
| | - Anthony Cullen
- School of Medicine & Medical Science, University College Dublin, C206 Health Sciences Building, Belfield, Dublin 4, Ireland.
| | - Alvise Calamai
- School of Medicine & Medical Science, University College Dublin, C206 Health Sciences Building, Belfield, Dublin 4, Ireland.
| |
Collapse
|
58
|
Upreti RK, Kannan A, Pant AB. Experimental impact of aspirin exposure on rat intestinal bacteria, epithelial cells and cell line. Hum Exp Toxicol 2010; 29:833-43. [DOI: 10.1177/0960327110363333] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Aspirin, a commonly used therapeutic non-steroidal anti-inflammatory drug (NSAID) is known to cause gastric mucosal damage. Intestinal bacteria having a regulatory effect on intestinal homeostasis play significant role in NSAID-induced intestinal injury. Bacteria and specific cell lines are considered to be suitable for toxicity screening and testing of chemicals. Therefore, to evaluate and compare in vitro toxicity, cultures of rat intestinal epithelial cells (IEC), isolated bacteria and IEC-6 cell line were assessed for viability, morphometric analysis, membrane transport enzymes and structural constituents for membrane damage, dehydrogenase activity test for respiratory and energy producing processes and esterase activity test for intra- and extra-cellular degradation, following the post exposure to aspirin (0—50 µg mL- 1). Similar pattern of dose-dependent changes in these parameters were observed in three types of cells. Similar in situ effects on IEC validated the in vitro findings. These findings indicate that higher aspirin concentrations may alter cellular functions of IEC and gut bacteria. Furthermore, results suggest that gut bacteria and IEC-6 cell line can be used for the initial screening of gastrointestinal cellular toxicity caused by NSAIDs.
Collapse
Affiliation(s)
- Raj K Upreti
- Biomembrane Toxicology, Indian Institute of Toxicology Research (National Laboratory of Council of Scientific & Industrial Research, New Delhi-India), MG Marg, Lucknow - 226 001, India,
| | - A. Kannan
- Biomembrane Toxicology, Indian Institute of Toxicology Research (National Laboratory of Council of Scientific & Industrial Research, New Delhi-India), MG Marg, Lucknow - 226 001, India
| | - AB Pant
- Biomembrane Toxicology, Indian Institute of Toxicology Research (National Laboratory of Council of Scientific & Industrial Research, New Delhi-India), MG Marg, Lucknow - 226 001, India
| |
Collapse
|
59
|
Dennis T, Fanous M, Mousa S. Natural products for chemopreventive and adjunctive therapy in oncologic disease. Nutr Cancer 2010; 61:587-97. [PMID: 19838932 DOI: 10.1080/01635580902825530] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Nutritional supplements or complementary and alternative medicines (CAM) are currently being investigated for their use in preventing, inhibiting, and reversing the progression of cancer. Natural agents and their derivatives such as vitamin A, selenium, green tea, resveratrol, aspirin, and probiotics have potential benefits in chemoprevention. There is also growing evidence for the use of natural products as adjunctive therapy alongside conventional cancer treatments. Nutritional supplements expenditures demonstrated greater growth than pharmaceuticals, with approximately 80% of cancer patients using natural products. Current issues with nutritional supplements use in cancer treatment include insufficient or conflicting evidence, poor quality control, potential interactions with chemotherapy, and potential efficacy in relation to changes in certain biomarkers, but long-term implications remain largely unresolved. Continued research is needed to lend credibility to these potentially valuable naturally driven supplements in the prevention and potentially in the treatment of cancer in conjunction with standard pharmaceuticals.
Collapse
Affiliation(s)
- Tiffany Dennis
- Albany College of Pharmacy, The Pharmaceutical Research Institute, Albany, New York, USA
| | | | | |
Collapse
|
60
|
Li CJ, Chang JK, Chou CH, Wang GJ, Ho ML. The PI3K/Akt/FOXO3a/p27Kip1 signaling contributes to anti-inflammatory drug-suppressed proliferation of human osteoblasts. Biochem Pharmacol 2009; 79:926-37. [PMID: 19883628 DOI: 10.1016/j.bcp.2009.10.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Revised: 10/21/2009] [Accepted: 10/21/2009] [Indexed: 12/17/2022]
Abstract
Akt has been reported to suppress p27(Kip1) promoter activity through Forkhead box O (FOXO) in different kinds of cells. Previous studies indicated that anti-inflammatory drugs up-regulated p27(Kip1), and this effect might play an important role in anti-inflammatory drug-induced cell cycle arrest of human osteoblasts (hOBs). In this study, we hypothesized that these drugs might increase p27(Kip1) expression in hOBs by altering the Akt/FOXO signaling. We tested this hypothesis by examining the influences of three anti-inflammatory drugs on the levels and/or activities of Akt, FOXO and p27(Kip1) as well as the relationship between these factors and proliferation of hOBs. We tested the effects of indomethacin (10(-5) and 10(-4)M), celecoxib (10(-6) and 10(-5)M), and dexamethasone (10(-7) and 10(-6)M) using PI3K inhibitor, LY294002 (10(-5)M) as the basis of comparison. The three drugs suppressed the canonical level of phosphorylated Akt in hOBs. This was accompanied by elevated FOXO3a level and increased promoter activity, mRNA expression and protein level of p27(Kip1). Furthermore, the anti-inflammatory drugs suppressed the EGF-induced increases in proliferation, phosphorylation, and nucleus translocation of Akt. Simultaneously, they suppressed EGF-induced decreases of FOXO3a nucleus accumulation and p27(Kip1) mRNA expression. On the other hand, FOXO silencing significantly attenuated the drug-induced up-regulation of p27(Kip1) and suppression of proliferation in hOBs. To the best of our knowledge, this study represents the first to demonstrate that Akt/FOXO3a/p27(Kip1) pathway contributes to suppression of hOB proliferation by anti-inflammatory drugs. We suggest that anti-inflammatory drugs suppress hOB proliferation, at least partly, through inactivating Akt, activating FOXO3a, and eventually up-regulating p27(Kip1) expression.
Collapse
Affiliation(s)
- Ching-Ju Li
- Department of Physiology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | | | | | |
Collapse
|
61
|
Demiralay EC, Alsancak G, Ozkan SA. Determination of pK
a
values of nonsteroidal antiinflammatory drug-oxicams by RP-HPLC and their analysis in pharmaceutical dosage forms. J Sep Sci 2009; 32:2928-36. [DOI: 10.1002/jssc.200900234] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
62
|
Thurnher, Maia Bakroeva, Gertraud S D. Non-steroidal Anti-inflammatory Drugs Induce Apoptosis in Head and Neck Cancer Cell Lines. Acta Otolaryngol 2009. [DOI: 10.1080/00016480127365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
63
|
Guo Q, Wu M, Lian P, Liao M, Xiao Z, Wang X, Shen S. Synergistic effect of indomethacin and NGX6 on proliferation and invasion by human colorectal cancer cells through modulation of the Wnt/β-catenin signaling pathway. Mol Cell Biochem 2009; 330:71-81. [DOI: 10.1007/s11010-009-0102-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2009] [Accepted: 03/30/2009] [Indexed: 11/29/2022]
|
64
|
Swiderski J, Withrow S. A novel surgical stapling technique for rectal mass removal: a retrospective analysis. J Am Anim Hosp Assoc 2009; 45:67-71. [PMID: 19258417 DOI: 10.5326/0450067] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Both benign and malignant rectal masses occur in dogs. The mainstay of treatment is surgical excision with adjuvant therapy based on histopathological diagnosis and completeness of removal. Location of the mass within the rectum helps dictate the approach used. This paper describes the use of a novel technique for removal of rectal masses involving the distal third of the rectum in seven dogs. To perform this technique, the rectum is prolapsed and stay sutures are placed to maintain prolapse. A thoracoabdominal stapling device is placed at the base of the mass with a minimum of 0.5- to 1-cm margins, and the mass is amputated. Mean time to veterinarian follow-up was 564 days, and no dog had recurrence of disease during this time.
Collapse
Affiliation(s)
- Jen Swiderski
- Colorado State University Veterinary Medical Center, Cancer Center, Fort Collins, 80523, USA
| | | |
Collapse
|
65
|
Stewart GD, Nanda J, Brown DJG, Riddick ACP, Ross JA, Habib FK. NO-sulindac inhibits the hypoxia response of PC-3 prostate cancer cells via the Akt signalling pathway. Int J Cancer 2009; 124:223-32. [PMID: 18924134 DOI: 10.1002/ijc.23934] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nitric oxide-donating non-steroidal anti-inflammatory drugs are safer than traditional NSAIDs and inhibit the growth of prostate cancer cells with greater potency than NSAIDs. In vivo, prostate cancer deposits are found in a hypoxic environment which induces resistance to chemotherapy. The aim of this study was to assess the effects and mechanism of action of a NO-NSAID called NO-sulindac on the PC-3 prostate cancer cell line under hypoxic conditions. NO-sulindac was found to have pro-apoptotic, cytotoxic, and anti-invasive effect on PC-3 cells under normoxia and hypoxia. NO-sulindac was significantly more cytotoxic than sulindac at all oxygen levels. The sulindac/linker and NO-releasing subunits both contributed to the cytotoxic effects of NO-sulindac. Resistance of PC-3 cells to NO-sulindac was induced as the oxygen concentration declined. Hypoxia-induced chemoresistance was reversed by knocking-down hypoxia-inducible factor-1alpha (HIF-1alpha) mRNA using RNAi. Nuclear HIF-1alpha levels were upregulated at 0.2% oxygen but reduced by treatment with NO-sulindac, as was Akt phosphorylation. NO-sulindac treatment of hypoxic PC-3 cells transfected with a reporter construct, downregulated activation of the hypoxia response element (HRE) promoter. Co-transfection of PC-3 cells with the HRE promoter reporter construct and myr-Akt (constitutively active Akt) plasmids reversed the NO-sulindac induced reduction in HRE activation. Real-time polymerase chain reaction analysis of hypoxic, NO-sulindac treated PC-3 cells showed downregulation of lysyl oxidase and carbonic anhydrase IX mRNA expression. Collectively, these novel findings demonstrate that NO-sulindac directly inhibits the hypoxia response of PC-3 prostate cancer cells by inhibiting HIF-1alpha translation via the Akt signalling pathway. The ability of NO-sulindac to inhibit tumour adaption to hypoxia has considerable relevance to the future management of prostate cancer with the same cellular properties as PC-3.
Collapse
Affiliation(s)
- Grant D Stewart
- Prostate Research Group, Edinburgh Cancer Research Centre, University of Edinburgh, Edinburgh, Scotland, United Kingdom.
| | | | | | | | | | | |
Collapse
|
66
|
Strillacci A, Griffoni C, Sansone P, Paterini P, Piazzi G, Lazzarini G, Spisni E, Pantaleo MA, Biasco G, Tomasi V. MiR-101 downregulation is involved in cyclooxygenase-2 overexpression in human colon cancer cells. Exp Cell Res 2008; 315:1439-47. [PMID: 19133256 DOI: 10.1016/j.yexcr.2008.12.010] [Citation(s) in RCA: 190] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2008] [Revised: 12/16/2008] [Accepted: 12/16/2008] [Indexed: 12/14/2022]
Abstract
Overexpressed cyclooxygenase-2 (COX-2) strongly contributes to the growth and invasiveness of tumoral cells in patients affected by colorectal cancer (CRC). It has been demonstrated that COX-2 overexpression depends on different cellular pathways involving both transcriptional and post-transcriptional regulations. We assumed that COX-2 expression could be regulated also by microRNAs (miRNAs) since these short RNA molecules participate to the fine regulation of several genes implicated in cell growth and differentiation. In this paper, we report the inverse correlation between COX-2 and miR-101 expression in colon cancer cell lines and we demonstrated in vitro the direct inhibition of COX-2 mRNA translation mediated by miR-101. Moreover, this correlation was supported by data collected ex vivo, in which colon cancer tissues and liver metastases derived from CRC patients were analyzed. These findings provide a novel molecular insight in the modulation of COX-2 at post-transcriptional level by miR-101 and strengthen the observation that miRNAs are highly implicated in the control of gene expression. An impairment of miR-101 levels could represent one of the leading causes of COX-2 overexpression in colon cancer cells.
Collapse
Affiliation(s)
- Antonio Strillacci
- Department of Experimental Biology, University of Bologna, via Selmi 3, 40126 Bologna, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Gonçalves P, Araújo JR, Pinho MJ, Martel F. Modulation of butyrate transport in Caco-2 cells. Naunyn Schmiedebergs Arch Pharmacol 2008; 379:325-36. [PMID: 19023563 DOI: 10.1007/s00210-008-0372-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2008] [Accepted: 10/30/2008] [Indexed: 02/06/2023]
Abstract
The aim of this study was to investigate the putative influence of some pharmacological agents and drugs of abuse upon the apical uptake of butyrate (BT) into Caco-2 cells. The apical uptake of (14)C-BT by Caco-2 cells was (1) time and concentration dependent, (2) pH dependent, (3) Na(+) independent and Cl(-) dependent, (4) energy dependent, (5) inhibited by several BT structural analogues (acetate, propionate, alpha-ketobutyrate, pyruvate, lactate), (6) insensitive to the anion exchange inhibitors DIDS and SITS and (7) inhibited by the monocarboxylate transport (MCT) inhibitors NPPB and pCMB. These characteristics are compatible with an involvement of MCT1-mediated transport. Acutely, uptake of a low concentration of (14)C-BT (10 microM) was reduced by acetaldehyde, acetylsalicylic acid, indomethacin, caffeine and theophylline and increased by MDMA. Chronically, uptake was increased by caffeine and decreased by tetrahydrocannabinol and MDMA; reverse transcription quantitative real-time PCR analysis showed that these three compounds decreased the mRNA levels of MCT1. Acutely, acetaldehyde, indomethacin and MDMA reduced the uptake of a high concentration of (14)C-BT (20 mM), and acetylsalicylic acid increased it. Chronically, none of the compounds affected uptake. Acetaldehyde, indomethacin and propionate seem to be competitive inhibitors of (14)C-BT uptake. Acetylsalicylic acid simultaneously increased the K (m) and the V (max) of (14)C-BT uptake. In conclusion, MCT1-mediated transport of (14)C-BT in Caco-2 cells is modulated by either acute or chronic exposure to some pharmacological agents and drugs of abuse (acetaldehyde, acetylsalicylic acid, indomethacin, caffeine, theophylline and the drugs of abuse tetrahydrocannabinol and MDMA).
Collapse
Affiliation(s)
- Pedro Gonçalves
- Department of Biochemistry (U38-FCT), Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
| | | | | | | |
Collapse
|
68
|
Aspirin Has Antitumor Effects via Expression of Calpain Gene in Cervical Cancer Cells. JOURNAL OF ONCOLOGY 2008; 2008:285374. [PMID: 19266085 PMCID: PMC2648633 DOI: 10.1155/2008/285374] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2008] [Revised: 07/17/2008] [Accepted: 07/22/2008] [Indexed: 11/17/2022]
Abstract
Aspirin and other nonsteroidal anti-inflammatory drugs show efficacy in the prevention of cancers. It is known that they can inhibit cyclooxygenases, and some studies have shown that they can induce apoptosis. Our objective in this study was to investigate the mechanism by which aspirin exerts its apoptosis effects in human cervical cancer HeLa cells. The effect of aspirin on the gene expression was studied by differential mRNA display RT-PCR. Among the isolated genes, mu-type calpain gene was upregulated by aspirin treatment. To examine whether calpain mediates the antitumor effects, HeLa cells were stably transfected with the mammalian expression vector pCR3.1 containing mu-type calpain cDNA (pCRCAL/HeLa), and tumor formations were measured in nude mice. When tumor burden was measured by day 49, HeLa cells and pCR/HeLa cells (vector control) produced tumors of 2126 mm3 and 1638 mm3, respectively, while pCRCAL/HeLa cells produced markedly smaller tumor of 434 mm3 in volume. The caspase-3 activity was markedly elevated in pCRCAL/HeLa cells. The increased activity levels of caspase-3 in pCRCAL/HeLa cells, in parallel with the decreased tumor formation, suggest a correlation between caspase-3 activity and calpain protein. Therefore, we conclude that aspirin-induced calpain mediates an antitumor effect via caspase-3 in cervical cancer cells.
Collapse
|
69
|
Schwab M, Reynders V, Loitsch S, Shastri YM, Steinhilber D, Schröder O, Stein J. PPARgamma is involved in mesalazine-mediated induction of apoptosis and inhibition of cell growth in colon cancer cells. Carcinogenesis 2008; 29:1407-1414. [PMID: 18544567 DOI: 10.1093/carcin/bgn118] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
PURPOSE Mesalazine has been identified as a candidate chemopreventive agent in colon cancer prophylaxis because of its pro-apoptotic and anti-proliferative effects. However, the precise mechanisms of action are not entirely understood. The aim of our study was to investigate the involvement of peroxisome proliferator-activated receptor gamma (PPARgamma) in mesalazine's anticarcinogenic actions in colorectal cancer cells. EXPERIMENTAL DESIGN The effects of mesalazine on cell cycle distribution, cell count, proliferation and caspase-mediated apoptosis were examined in Caco-2, HT-29 and HCT-116 cells used as wild-type, dominant-negative PPARgamma mutant and empty vector cultures. We focused on caspase-3 activity, cleavage of poly(ADP-ribose) polymerase (PARP), caspase-8 and caspase-9, as well as on expression of survivin, X-linked inhibitor of apoptosis (Xiap), phosphatase and tensin homolog deleted from chromosome ten (PTEN) and c-Myc. Techniques employed included transfection assays, immunoblotting, flow cytometry analysis, colorimetric and fluorometric assays. RESULTS Mesalazine caused a time- and dose-dependent decrease in both cell growth and proliferation. Growth inhibition was accompanied by a G1/G0 arrest, a significant increase in PTEN, caspase-3 activity, cleavage of PARP and caspase-8, whereas the expressions of Xiap, survivin and c-Myc were decreased simultaneously. Cleavage of caspase-9 was not observed. Moreover, PPARgamma expression and activity were elevated. The growth-inhibitory effect of mesalazine was partially reduced in dominant-negative PPARgamma mutant cells, whereas the expression of c-Myc was not affected. Mesalazine-mediated increased caspase-3 activity, the expression of PTEN, cleavage of PARP and caspase-8 as well as reduced levels of survivin and Xiap were completely abolished in the PPARgamma mutant cell lines. CONCLUSION This study clearly demonstrates that mesalazine-mediated pro-apoptotic and anti-proliferative actions are regulated via PPARgamma-dependent and -independent pathways in colonocytes.
Collapse
Affiliation(s)
- Markus Schwab
- First Department of Medicine-ZAFES, Johann Wolfgang Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | | | | | | | | | | | | |
Collapse
|
70
|
Lee KJ, Jung YS, Kim WH, Yoon TI, Joo HJ, Soh EY. Cyclooxygenase-2 expression in human thyroid disease. J Endocrinol Invest 2008; 31:111-8. [PMID: 18362501 DOI: 10.1007/bf03345576] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
OBJECTIVES Cyclooxygenase (COX)-2, which is the inducible form of the COX enzyme for prostaglandin synthesis and a key mediator of epithelial cell growth, has been shown to be up-regulated in gastrointestinal cancers. Additionally, regular intake of other non-steroidal anti-inflammatory drugs (NSAID) is known to decrease the incidence of these cancers. Therefore, the goals of the present study were to determine the possible involvement of COX-2 in human thyroid diseases. METHODS We used immunohistochemical staining and Western blot analysis to characterize the expression of COX-2 proteins in thyroid tissues from 64 patients with thyroiditis, benign tumors, and malignant tumors with or without metastasis. Immunoreactivity scores were calculated by multiplication of the determined grades. RESULTS COX-2 proteins were not expressed in normal thyroid tissues. However, each type of tumor tissue showed intense bands of COX-2 protein expression in Western blot analyses, and the immunoreactivity scores were 7.67+/-1.17 (SD) for thyroiditis, 7.87+/-0.9 for benign tumors, 7.53+/-1.53 for follicular cancer, 7.63+/-1.11 for papillary cancer without metastasis, and 7.17+/-1.55 for papillary cancer with metastasis. No significant differences were found in the levels of COX-2 expression between different tumor tissue types. CONCLUSION No significant correlations were observed between clinical and/or pathological characteristics of thyroid tumors and the intensity of COX-2 protein expression. In addition, we found no difference in COX-2 protein expression between thyroiditis and thyroid tumors. Thus, up-regulation of COX-2 protein synthesis in human thyroid diseases does not appear to be of clinical significance.
Collapse
Affiliation(s)
- K J Lee
- Department of Surgery, School of Medicine, Ajou University, Suwon, South Korea
| | | | | | | | | | | |
Collapse
|
71
|
Ribeiro G, Benadiba M, Colquhoun A, de Oliveira Silva D. Diruthenium(II,III) complexes of ibuprofen, aspirin, naproxen and indomethacin non-steroidal anti-inflammatory drugs: Synthesis, characterization and their effects on tumor-cell proliferation. Polyhedron 2008. [DOI: 10.1016/j.poly.2007.12.011] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
72
|
BURNETT BRUCEP, SILVA STACIA, MESCHES MICHAELH, WILSON STEVEN, JIA QI. SAFETY EVALUATION OF A COMBINATION, DEFINED EXTRACT OF SCUTELLARIA BAICALENSIS AND ACACIA CATECHU. J Food Biochem 2007. [DOI: 10.1111/j.1745-4514.2007.00142.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
73
|
Goldin N, Heyfets A, Reischer D, Flescher E. Mitochondria-mediated ATP depletion by anti-cancer agents of the jasmonate family. J Bioenerg Biomembr 2007; 39:51-7. [PMID: 17549642 DOI: 10.1007/s10863-006-9061-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Jasmonates are plant stress hormones that induce suppression of proliferation and death in cancer cells, while being selectively inactive towards non-transformed cells. Jasmonates can overcome apoptotic blocks and exert cytotoxic effects on drug-resistant cells expressing p53 mutations. Jasmonates induce a rapid depletion of ATP in cancer cells. Indeed, this steep drop occurs when no signs of cell death are detectable yet. Experiments using modulators of ATP synthesis via glycolysis or oxidative phosphorylation suggest that the latter is the pathway suppressed by jasmonates. Consequently, the direct effects of jasmonates on mitochondria were evaluated. Jasmonates induced cytochrome c release and swelling in mitochondria isolated from cancer cells but not from normal ones. Thus, the selectivity of jasmonates against cancer cells is rooted at the mitochondrial level, and probably exploits differences between mitochondria from normal versus cancer cells. These findings position jasmonates as promising anti-cancer drugs acting via energetic depletion in neoplastic cells.
Collapse
Affiliation(s)
- Natalia Goldin
- Department of Human Microbiology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | | | |
Collapse
|
74
|
Bock JM, Menon SG, Goswami PC, Sinclair LL, Bedford NS, Domann FE, Trask DK. Relative non-steroidal anti-inflammatory drug (NSAID) antiproliferative activity is mediated through p21-induced G1 arrest and E2F inhibition. Mol Carcinog 2007; 46:857-64. [PMID: 17415779 DOI: 10.1002/mc.20318] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
This study was performed to compare the relative antineoplastic activity of 10 different non-steroidal anti-inflammatory drugs (NSAIDs) in clinical use, and to investigate the underlying mechanisms of this activity in a squamous cell carcinoma of the head and neck model (SCCHN). A standard 5-day MTT assay was used to calculate IC(50) values in UM-SCC-1 cells for 10 NSAIDs, including celecoxib, rofecoxib, sulindac sulfide, sulindac sulfone, indomethacin, ketoprofen, flurbiprofen, naproxen, piroxicam, and aspirin. Celecoxib, a COX-2 specific inhibitor, was by far the most potent NSAID, with an IC(50) of 39.9 +/- 1.1 microM, followed by sulindac sulfide (116.5 +/- 2.34 microM). Celecoxib and sulindac sulfide also induced more activation of caspase-3 than any other NSAID. Cell cycle analysis showed that celecoxib and sulindac sulfide both induced a 3-fold increase in G(1) phase distribution, and this correlated with strong induction of p21(waf1/cip1), inhibition of cyclin D1, and hypophosphorylation of Rb. Celecoxib and sulindac sulfide treatment induced strong downstream inhibition of E2F transactivating activity as determined by a luciferase reporter assay. These data demonstrate the wide range of activity of various NSAID agents, and reveal a mechanism of action through cell cycle inhibition and induction of apoptosis.
Collapse
Affiliation(s)
- Jonathan M Bock
- Department of Otolaryngology--Head and Neck Surgery, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | |
Collapse
|
75
|
Tan XL, Nieters A, Hoffmeister M, Beckmann L, Brenner H, Chang-Claude J. Genetic polymorphisms in TP53, nonsteroidal anti-inflammatory drugs and the risk of colorectal cancer: evidence for gene-environment interaction? Pharmacogenet Genomics 2007; 17:639-45. [PMID: 17622940 DOI: 10.1097/fpc.0b013e3280d5121c] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Substantial evidence indicates that nonsteroidal anti-inflammatory drugs protect against colorectal cancer by altering cell cycle progression and/or inducing apoptosis, whereas p53 protein is crucial to maintaining cell-cycle arrest and regulating DNA repair, differentiation, and apoptosis. Genetic variants in TP53 gene might therefore influence colorectal cancer risk and modify the effects of nonsteroidal anti-inflammatory drugs. We assessed the association of TP53 Arg72Pro and p53PIN3 polymorphisms with colorectal cancer risk and their possible interaction with nonsteroidal anti-inflammatory drug use. METHODS We included 467 cases and 563 controls from a population-based case-control study. Multivariate logistic regression analysis was used to estimate the association between genotypes, environmental exposures and colorectal cancer risk, adjusting for potential confounders. RESULTS Odds ratios of colorectal cancer were 0.75 (95% confidence interval, 0.57-0.99) for TP53 72Pro carriers compared with those homozygous for the TP53 72Arg allele and 0.78 (95% confidence interval, 0.58-1.05) for p53PIN3 A2 carriers compared with p53PIN3 A1A1. Risks differed by nonsteroidal anti-inflammatory drug use. For both investigated TP53 polymorphisms, we found that the colorectal cancer risk associated with regular nonsteroidal anti-inflammatory drug use was statistically significantly modified by the TP53 genotype (P values for interaction=0.049 and 0.034, respectively), whereby a substantial protective effect of nonsteroidal anti-inflammatory drug use was observed for homozygous carriers of the 72Arg allele and of the PIN3 A1 allele (odds ratio 0.44; 95% confidence interval, 0.30-0.65 and odds ratio, 0.45; 95% confidence interval, 0.31-0.65). The interaction between nonsteroidal anti-inflammatory drugs and TP53 genetic polymorphisms was confirmed by haplotype analysis. CONCLUSIONS These data suggest that the TP53 genotype may modify the influence of nonsteroidal anti-inflammatory drug use on the risk of colorectal cancer. A direct proof of functional analysis is warranted to confirm these findings.
Collapse
Affiliation(s)
- Xiang-Lin Tan
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
76
|
Kleban J, Mikes J, Szilárdiová B, Koval J, Sacková V, Solár P, Horváth V, Hofmanová J, Kozubík A, Fedorocko P. Modulation of Hypericin Photodynamic Therapy by Pretreatment with 12 Various Inhibitors of Arachidonic Acid Metabolism in Colon Adenocarcinoma HT-29 Cells. Photochem Photobiol 2007; 83:1174-85. [PMID: 17880512 DOI: 10.1111/j.1751-1097.2007.00127.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
One proposal to increase the efficiency of photodynamic therapy (PDT) is to accompany photosensitization with other treatment modalities, including modulation of arachidonic acid (AA) metabolism. The aim of this study was to evaluate the effectiveness of a combined modality approach employing 48 and 24 h pretreatment with various inhibitors of lipoxygenase (LOX; nordihydroguaiaretic acid, esculetin, AA-861, MK-886 and baicalein), cyclooxygenase (COX; diclofenac, flurbiprofen, ibuprofen, indomethacin, SC-560 and rofecoxib) and cytochrome P450-monooxygenase (proadifen) pathways, followed by hypericin-mediated PDT. Cytokinetic parameters like MTT assay, adherent and floating cell numbers, viability and cell cycle distribution analysis were examined 24 h after hypericin activation. Pretreatment of human colon cancer cells HT-29 prior to PDT with 5-LOX inhibitor MK-886 as well as 5, 12-LOX and 12-LOX inhibitors (esculetin and baicalein, respectively) resulted in significant and dose-dependent effects on all parameters tested. Pretreatment with diclofenac, flurbiprofen, ibuprofen and indomethacin, the nonspecific COX inhibitors, promoted hypericin-mediated PDT, but these effects were probably COX-independent. In contrast, application of SC-560 and rofecoxib, specific inhibitors of COX-1 and COX-2, respectively, attenuated PDT. Inhibition of P450 monooxygenase with proadifen implied also the significance of this metabolic pathway in cell survival and cell resistance to hypericin photocytotoxicity. In conclusion, our results testify that application of diverse inhibitors of AA metabolism may have different consequences on cellular response to hypericin-mediated PDT and that some of them could be considered for potentiation of PDT.
Collapse
Affiliation(s)
- Ján Kleban
- Institute of Biology and Ecology, Faculty of Sciences, P. J. Safárik University, Kosice, Slovakia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Camici M. Guanylin peptides and colorectal cancer (CRC). Biomed Pharmacother 2007; 62:70-6. [PMID: 17582727 DOI: 10.1016/j.biopha.2007.04.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2007] [Accepted: 04/26/2007] [Indexed: 12/27/2022] Open
Abstract
Agonists of guanylyl-C receptor, such as guanylin/uroguanylin, are correlated not only with the intestinal cell epithelial physiology but also with the colorectal cancer tumorigenesis. Activation of the second intracellular messenger cyclic guanosine monophosphate by guanylyl cyclase-C receptor results in a complex intracellular signalling cascade involving the phosphodiesterase, the ion channels and the protein kinase. After an analytical review of relevant new knowledge, new diagnostic and therapeutic approaches for colorectal cancer are discussed.
Collapse
Affiliation(s)
- Marcello Camici
- Internal Medicine Department, University of Pisa, Via Roma 67, 56126 Pisa, Italy.
| |
Collapse
|
78
|
Rijcken FEM, Hollema H, van der Zee AGJ, van der Sluis T, Boersma-van Ek W, Kleibeuker JH. Sulindac treatment in hereditary non-polyposis colorectal cancer. Eur J Cancer 2007; 43:1251-6. [PMID: 17434727 DOI: 10.1016/j.ejca.2007.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2006] [Revised: 02/20/2007] [Accepted: 03/05/2007] [Indexed: 12/21/2022]
Abstract
UNLABELLED Non-steroidal anti-inflammatory drugs, e.g. sulindac have been extensively studied for chemoprevention in familial adenomatous polyposis, but not in hereditary non-polyposis colorectal cancer (HNPCC). We evaluated these effects in HNPCC using surrogate end-points for cancer risk. In a randomised double-blind cross-over study, 22 subjects (9 female; age 30-66 years, mean 44), all ascertained or probable mutation carriers for HNPCC, were included. Sulindac 150 mg b.i.d. and placebo were given for 4 weeks each, with 4 weeks in between, with biopsies taken from ascending, transverse and sigmoid colon and rectum by colonoscopy after both periods. Proliferation was determined by Ki-67 staining and apoptosis by staining of cytokeratin 18 cleavage products. Expression of cyclins B1, D3 and E and p21, p27, bax, bcl2 and cox-2 was studied immunohistochemically. Proliferation was higher during sulindac treatment than drug placebo treatment in ascending and transverse colon, but not in sigmoid and rectum. Apoptosis was not affected. Besides an increase in cyclin D3, no differences were found in expression of regulating proteins in the proximal colon. CONCLUSION Sulindac induces an increase in epithelial cell proliferation in the proximal colon of subjects with HNPCC. Since colorectal cancer predominantly arises in the proximal colon in HNPCC, these results cast doubts on the potential chemopreventive effects of sulindac in HNPCC.
Collapse
Affiliation(s)
- Fleur E M Rijcken
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | | | | | | | | | | |
Collapse
|
79
|
Hostanska K, Jürgenliemk G, Abel G, Nahrstedt A, Saller R. Willow bark extract (BNO1455) and its fractions suppress growth and induce apoptosis in human colon and lung cancer cells. ACTA ACUST UNITED AC 2007; 31:129-39. [PMID: 17418981 DOI: 10.1016/j.cdp.2007.03.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/03/2007] [Indexed: 12/21/2022]
Abstract
BACKGROUND Recently, there have been extensive efforts to evaluate the chemopreventive role of substances present in natural products. The aim of this study was to examine the effects of the main groups of compounds (salicylalcohol derivates, flavonoids, proanthocyanidins), and salicin isolated from willow bark extract BNO 1455 on proliferation and apoptosis in human colon and cancer cells. METHODS We used human colon cyclooxygenase-2 (COX-2)-positive HT 29 and (COX-2)-negative HCT 116 or lung COX-2 proficient A 549 and low COX-2 expressing SW2 cells. After treatment for 72 h with various concentrations of single substances and acetylsalicylic acid (ASA) as control, inhibition of cell growth and cytotoxicity were measured by colorimetric WST-1 assay and propidium iodide uptake by flow cytometry, respectively. Apoptotic cells were identified by annexin V adhesion using flow cytometry. RESULTS Studies on dose-dependent effects of BNO 1455 and its fractions showed anti-proliferative activity of all compounds with 50% maximal growth inhibitory concentrations (GI(50)) between 33.3 and 103.3 microg/ml for flavonoids and proanthocyanidins fractions and 50.0-243.0 microg/ml for salicylalcohol derivates and extract. Apoptosis induction was confirmed by annexin V adherence and analysis of cell morphology based on light scattering characteristics using flow cytometry in all cell lines at GI(50). CONCLUSIONS We showed that willow bark extract BNO 1455 an its fractions inhibit the cell growth and promote apoptosis in human colon and lung cancer cell lines irrespective of their COX-selectivity.
Collapse
Affiliation(s)
- Katarina Hostanska
- University Hospital Zürich, Department of Internal Medicine, Institute for Complementary Medicine, FGel 102, Rämistrasse 100, CH-8091 Zürich, Switzerland.
| | | | | | | | | |
Collapse
|
80
|
Lev-Ari S, Kazanov D, Liberman E, Ben-Yosef R, Arber N. Down-regulation of PGE2 by physiologic levels of celecoxib is not sufficient to induce apoptosis or inhibit cell proliferation in human colon carcinoma cell lines. Dig Dis Sci 2007; 52:1128-33. [PMID: 17342386 DOI: 10.1007/s10620-006-9619-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2006] [Accepted: 09/14/2006] [Indexed: 01/20/2023]
Abstract
This study was performed to evaluate whether down-regulation of prostaglandin E(2) (PGE(2)) synthesis by celecoxib treatment is associated with inhibition of cell growth in human colon carcinoma cell lines. Physiologic concentrations of celecoxib (5-10 microM) inhibited 80% to 90% of PGE(2) production in HT-29 cells that express high levels of COX-2 protein. In these concentrations, celecoxib had a minor inhibitory effect (20-30%) on cell growth. There was a significant change in induction of apoptosis only at higher concentrations of celecoxib (>20 microM). Treatment by low concentrations of celecoxib did not alter the levels of COX-1, beta-catenin, P(27), Bcl-2, and Bcl-x proteins. The effect of celecoxib on cell growth inhibition was higher on the COX-2-positive HT-29 cell line (IC(50)=20 microM) than on the COX-2 deficient SW-480 cell line (IC(50)=35 microM). In conclusion, inhibition of PGE(2) synthesis is an early, but not sufficient, step in the mechanism of celecoxib-mediated cell growth inhibition. These results support the need for additional evaluation of independent COX-2 pathways of celecoxib in chemoprevention of CRC.
Collapse
Affiliation(s)
- Shahar Lev-Ari
- Integrated Cancer Prevention Center, Tel Aviv Sourasky Medical Center, Tel Aviv University, 6 Weizmann Street, Tel Aviv 64239, Israel
| | | | | | | | | |
Collapse
|
81
|
Marjanović M, Zorc B, Pejnović L, Zovko M, Kralj M. Fenoprofen and Ketoprofen Amides as Potential Antitumor Agents. Chem Biol Drug Des 2007; 69:222-6. [PMID: 17441909 DOI: 10.1111/j.1747-0285.2007.00494.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Following numerous experimental observations that various non-steroidal anti-inflammatory drugs have antitumor potentials, a series of fenoprofenamides (1a-g) and ketoprofenamides (2a-c) was tested on proliferation of different human tumor cell lines and normal human fibroblasts in vitro. Fenoprofen and ketoprofen showed modest antiproliferative activity, whereas the growth inhibitory activity of the tested amides clearly demonstrates that the substituents linked by an amide bond are essential for the significantly stronger cytostatic activity, probably because of a greater lipophilicity and/or better cell uptake. Additionally, it was shown that the most active derivatives (1d and 2a) induced cell cycle arrest at the G1 phase, as well as apoptosis.
Collapse
|
82
|
Mahdi JG, Alkarrawi MA, Mahdi AJ, Bowen ID, Humam D. Calcium salicylate-mediated apoptosis in human HT-1080 fibrosarcoma cells. Cell Prolif 2006; 39:249-60. [PMID: 16872361 PMCID: PMC6495909 DOI: 10.1111/j.1365-2184.2006.00390.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Salicylates are novel biologically active compounds that exhibit multiple therapeutic activities. The anti-cancer effectiveness of calcium salicylate has been investigated on human HT-1080 fibrosarcoma cell lines at relatively low concentrations (predominantly 0.4 mM) compared to those previously reported. Although low calcium salicylate concentrations did not retard tumour growth progression significantly, as measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and time-lapse assays, its cytotoxic characteristics were proven to be prominent by various morphological and immunocytological techniques. The results here demonstrate evidence for approximately 25% apoptosis after treatment with calcium salicylate, which up-regulatd the expression of p53, p21 and Bax, and down-regulated Bcl-2 in HT-1080 cells.
Collapse
Affiliation(s)
- J G Mahdi
- School of Biosciences, Cardiff University, PO Box 911, Cardiff CF10 3US, UK.
| | | | | | | | | |
Collapse
|
83
|
Hu Y, Le Leu RK, Young GP. Defective acute apoptotic response to genotoxic carcinogen in small intestine of APC(Min/+) mice is restored by sulindac. Cancer Lett 2006; 248:234-44. [PMID: 16950562 DOI: 10.1016/j.canlet.2006.07.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2006] [Revised: 07/27/2006] [Accepted: 07/28/2006] [Indexed: 01/28/2023]
Abstract
The effect of APC loss on azoxymethane (AOM)-induced apoptosis and cell proliferation, as well as their regulation by sulindac was examined in colon and small intestine in APC(Min/+) mice. APC(Min/+) mice showed increased epithelial proliferation in all regions, with significant impairment of apoptosis in small intestine, but not in colon. Sulindac administration restored defective apoptosis to normal. As the apoptotic defect occurred at the major site of intestinal tumor formation in APC(Min/+) mice and as it was restored to normal by a proven chemopreventive agent, this defect in apoptosis might be a key biological consequence of APC dysfunction contributing to tumor formation.
Collapse
Affiliation(s)
- Ying Hu
- Department of Medicine, Flinders University of South Australia, Adelaide, Australia.
| | | | | |
Collapse
|
84
|
Maglietta R, D'Addabbo A, Piepoli A, Perri F, Liuni S, Pesole G, Ancona N. Selection of relevant genes in cancer diagnosis based on their prediction accuracy. Artif Intell Med 2006; 40:29-44. [PMID: 16920342 DOI: 10.1016/j.artmed.2006.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2006] [Revised: 06/01/2006] [Accepted: 06/06/2006] [Indexed: 10/24/2022]
Abstract
MOTIVATIONS One of the main problems in cancer diagnosis by using DNA microarray data is selecting genes relevant for the pathology by analyzing their expression profiles in tissues in two different phenotypical conditions. The question we pose is the following: how do we measure the relevance of a single gene in a given pathology? METHODS A gene is relevant for a particular disease if we are able to correctly predict the occurrence of the pathology in new patients on the basis of its expression level only. In other words, a gene is informative for the disease if its expression levels are useful for training a classifier able to generalize, that is, able to correctly predict the status of new patients. In this paper we present a selection bias free, statistically well founded method for finding relevant genes on the basis of their classification ability. RESULTS We applied the method on a colon cancer data set and produced a list of relevant genes, ranked on the basis of their prediction accuracy. We found, out of more than 6500 available genes, 54 overexpressed in normal tissues and 77 overexpressed in tumor tissues having prediction accuracy greater than 70% with p-value <or=0.05. CONCLUSIONS The relevance of the selected genes was assessed (a) statistically, evaluating the p-value of the estimate prediction accuracy of each gene; (b) biologically, confirming the involvement of many genes in generic carcinogenic processes and in particular for the colon; (c) comparatively, verifying the presence of these genes in other studies on the same data-set.
Collapse
Affiliation(s)
- Rosalia Maglietta
- Istituto di Studi sui Sistemi Intelligenti per l'Automazione, CNR Via Amendola 122/D-I, 70126 Bari, Italy
| | | | | | | | | | | | | |
Collapse
|
85
|
Pitari GM, Li T, Baksh RI, Waldman SA. Exisulind and guanylyl cyclase C induce distinct antineoplastic signaling mechanisms in human colon cancer cells. Mol Cancer Ther 2006; 5:1190-6. [PMID: 16731751 DOI: 10.1158/1535-7163.mct-05-0415] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The nonsteroidal anti-inflammatory drug sulindac is metabolized to sulindac sulfone (exisulind), an antineoplastic agent that inhibits growth and induces apoptosis in solid tumors. In colon cancer cells, the antineoplastic effects of exisulind have been attributed, in part, to induction of cyclic guanosine 3',5'-monophosphate (cGMP) signaling through inhibition of cGMP-specific phosphodiesterases, which elevates intracellular cGMP, and novel expression of cGMP-dependent protein kinase (PKG) Ibeta, the presumed downstream effector mediating apoptosis. Here, inhibition of proliferation and induction of cell death by exisulind was dissociated from cGMP signaling in human colon cancer cells. Accumulation of intracellular cGMP produced by an exogenous cell-permeant analogue of cGMP or a potent agonist of guanylyl cyclase C yielded cytostasis without cell death. Surprisingly, the antiproliferative effects of induced cGMP accumulation were paradoxically less than additive, rather than synergistic, when combined with exisulind. Further, although exisulind induced expression of PKG Ibeta, it did not elevate intracellular cGMP and its efficacy was not altered by inhibition or activation of PKG I. Rather, PKG I induced by exisulind may mediate desensitization of cytostasis induced by cGMP. Thus, cytotoxic effects of exisulind are independent of cGMP signaling in human colon cancer cells. Moreover, combination therapies, including exisulind and agents that induce cGMP signaling, may require careful evaluation in patients with colon cancer.
Collapse
Affiliation(s)
- Giovanni Mario Pitari
- Division of Clinical Pharmacology, Departments of Pharmacology and Experimental Therapeutics and Medicine, Thomas Jefferson University, 1100 Walnut Street MOB 810, Philadelphia, PA 19107, USA.
| | | | | | | |
Collapse
|
86
|
Dikshit P, Chatterjee M, Goswami A, Mishra A, Jana NR. Aspirin induces apoptosis through the inhibition of proteasome function. J Biol Chem 2006; 281:29228-35. [PMID: 16880202 DOI: 10.1074/jbc.m602629200] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Aspirin and other nonsteroidal anti-inflammatory drugs inhibit cell proliferation and induce apoptosis in various cancer cell lines, which is considered to be an important mechanism for their anti-tumor activity and prevention of carcinogenesis. However, the molecular mechanisms through which these compounds induce apoptosis are not well understood. Here we have found that aspirin treatment of the mouse Neuro 2a cells impaired the proteasome function and caused severe mitochondrial abnormalities. Treatment with aspirin lead to a dose- and time-dependent decrease in proteasome activity and an increase in the accumulation of ubiquitylated proteins in the cells, which correlated with its effect on cell death. Aspirin exposure also resulted in an increase in the half-life of pd1EGFP, a model substrate of proteasome, as well as various intracellular substrates like Bax, IkappaB-alpha, p53, and p27(kip1). Aspirin-induced proteasomal malfunction might be responsible, at least in part, for the down-regulation of NF-kappaB activity and neurite outgrowth. Finally, we have shown that aspirin treatment caused changes in the mitochondrial membrane potential, release of cytochrome c from mitochondria, and activation of caspase-9 and -3, which could be because of the proteasomal dysfunction.
Collapse
Affiliation(s)
- Priyanka Dikshit
- Cellular and Molecular Neuroscience Laboratory, National Brain Research Centre, Manesar, Gurgaon 122050, India
| | | | | | | | | |
Collapse
|
87
|
Wang Y, Chen X, Zhu W, Zhang H, Hu S, Cong X. GROWTH INHIBITION OF MESENCHYMAL STEM CELLS BY ASPIRIN: INVOLVEMENT OF THE WNT/β-CATENIN SIGNAL PATHWAY. Clin Exp Pharmacol Physiol 2006; 33:696-701. [PMID: 16895542 DOI: 10.1111/j.1440-1681.2006.04432.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
1. Mesenchymal stem cell (MSC) therapy is drawing increasing attention in cardiology. However, the effect of aspirin, an assistant medication used extensively in the treatment of cardiovascular diseases, on MSC is not clear. 2. In the present study, we investigated the effect of aspirin on the growth of MSC in vitro and the underlying mechanism of its action. 3. The 3-(4,5-dimethyl-2 thiazoyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay revealed that 1, 5 and 10 mmol/L aspirin inhibited the growth of MSC by 18, 37 and 62%, respectively. DNA synthesis of MSC was inhibited by 25, 57 and 90% following treatment with 1, 5 and 10 mmol/L aspirin, respectively, as determined by the tritiated thymidine incorporation assay. No cytotoxicity was observed based on Trypan blue dye exclusion and cell morphological observations. Western blot analysis demonstrated that the protein level of phosphorylated beta-catenin increased, whereas that of cyclin D1 decreased, after treatment of MSC with aspirin. Cell cycle analysis showed that aspirin failed to significantly alter the proportion of cells in different stages of the cell cycle. 4. These observations indicate that aspirin inhibits MSC proliferation and that the downregulation of the wnt/beta-catenin signal pathway may be involved in the growth inhibition of MSC by aspirin.
Collapse
Affiliation(s)
- Yiner Wang
- Research Center for Cardiovascular Regenerative Medicine, Cardiovascular Institute , Beijing, China
| | | | | | | | | | | |
Collapse
|
88
|
Neve RL, McPhie DL. The cell cycle as a therapeutic target for Alzheimer's disease. Pharmacol Ther 2006; 111:99-113. [PMID: 16274748 DOI: 10.1016/j.pharmthera.2005.09.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2005] [Accepted: 09/21/2005] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease worldwide. It is a progressive, incurable disease whose predominant clinical manifestation is memory loss, and which always ends in death. The classic neuropathological diagnostic markers for AD are amyloid plaques and neurofibrillary tangles, but our understanding of the role that these features of AD play in the etiology and progression of the disease remains incomplete. Research over the last decade has revealed that cell cycle abnormalities also represent a major neuropathological feature of AD. These abnormalities appear very early in the disease process, prior to the appearance of plaques and tangles. Growing evidence suggests that neuronal cell cycle regulatory failure, leading to apoptosis, may be a significant component of the pathogenesis of AD. A number of signaling pathways with the potential to activate aberrant cell cycle re-entry in AD have been described. The relationships among these signaling cascades, which involve the amyloid precursor protein (APP), cyclin-dependent kinases (cdks), and the cell cycle protein Pin1, have not yet been fully elucidated, but details of the individual pathways are beginning to emerge. This review summarizes the current state of knowledge with respect to specific neuronal signaling events that are thought to underlie cell cycle regulatory failure in AD brain. The elements of these pathways that represent potential new therapeutic targets for AD are described. Drugs and peptides that can inhibit molecular steps leading to AD neurodegeneration by intervening in the activation of cell cycle re-entry in neurons represent an entirely new approach to the development of treatments for AD.
Collapse
Affiliation(s)
- Rachael L Neve
- Department of Psychiatry, MRC 223, Harvard Medical School and McLean Hospital, Belmont, MA 02478, USA.
| | | |
Collapse
|
89
|
Abstract
Several epidemiological studies have indicated that the long-term use of NSAIDs, most of which are cyclo-oxygenase (COX) inhibitors, may reduce the risk of Alzheimer's disease. For this reason, anti-inflammatory COX-inhibiting NSAIDs have received increased attention in experimental and therapeutic trials for Alzheimer's disease. However, several recent efforts attempting to demonstrate a therapeutic effect of NSAIDs in Alzheimer's disease have largely failed. Clinicians and scientists currently believe that this lack of success may be attributable to two key problems: (i) clinical trials of NSAIDs have been conducted in patients with late-stage Alzheimer's disease, wherein advanced neurodegeneration may be refractory to anti-inflammatory drug treatment; and (ii) it is not known which of the large family of NSAIDs (i.e. COX-1, COX-2 or mixed inhibitors) is most efficacious in preventing Alzheimer's disease. The wide list of putative functions for COX in the brain, and the significant functional heterogeneity of NSAIDs, which appear to influence the beta-amyloid (Abeta) neuropathology associated with Alzheimer's disease via both COX-dependent and COX-independent pathways, complicate the interpretation of the mechanisms through which COX-inhibiting NSAIDs may beneficially influence Alzheimer's disease. As discussed in this review, for patients at high risk of developing Alzheimer's disease (e.g. those with mild cognitive impairment), preventative treatment with COX-inhibiting NSAIDs may ultimately represent a viable strategy in the management of clinical Alzheimer's disease. However, the recent evidence showing an increased risk of major cardiovascular events among patients treated with certain COX-1 and COX-2 inhibitors leaves many questions unanswered. We suggest that further investigation into the physiological role(s) of COXs in normal health and in disease conditions, and the identification of safer and better tolerated COX inhibitors, will provide renewed impetus to the application of anti-inflammatory strategies for the prevention and treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Lap Ho
- Department of Psychiatry, The Mount Sinai School of Medicine, Neuroinflammation Research Laboratories, New York, New York 10029, USA
| | | | | | | |
Collapse
|
90
|
Abstract
Systematic annotation of the primary targets of roughly 1000 known therapeutics reveals that over 700 of these modulate approximately 85 biological targets. We report the results of three analyses. In the first analysis, drug/drug similarities and target/target similarities were computed on the basis of three-dimensional ligand structures. Drug pairs sharing a target had significantly higher similarity than drug pairs sharing no target. Also, target pairs with no overlap in annotated drug specificity shared lower similarity than target pairs with increasing overlap. Two-way agglomerative clusterings of drugs and targets were consistent with known pharmacology and suggestive that side effects and drug-drug interactions might be revealed by modeling many targets. In the second analysis, we constructed and tested ligand-based models of 22 diverse targets in virtual screens using a background of screening molecules. Greater than 100-fold enrichment of cognate versus random molecules was observed in 20/22 cases. In the third analysis, selectivity of the models was tested using a background of drug molecules, with selectivity of greater than 80-fold observed in 17/22 cases. Predicted activities derived from crossing drugs against modeled targets identified a number of known side effects, drug specificities, and drug-drug interactions that have a rational basis in molecular structure.
Collapse
Affiliation(s)
- Ann E Cleves
- UCSF Cancer Research Institute and Department of Biopharmaceutical Sciences, University of California, San Francisco, California 94143, USA
| | | |
Collapse
|
91
|
Mahdi JG, Mahdi AJ, Mahdi AJ, Bowen ID. The historical analysis of aspirin discovery, its relation to the willow tree and antiproliferative and anticancer potential. Cell Prolif 2006; 39:147-55. [PMID: 16542349 PMCID: PMC6496865 DOI: 10.1111/j.1365-2184.2006.00377.x] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2005] [Accepted: 12/14/2005] [Indexed: 12/12/2022] Open
Abstract
For several millennia, the willow tree and salicin have been associated with salicylic acid, the key precursor molecule that has contributed to the discovery of acetylsalicylic acid, traded as aspirin. These molecules have been shown to possess phyto- and chemotherapeutic activities as analgesic drugs. In recent decades, aspirin has become the focus of extensive investigation into antiproliferative and anticancer activities. The historical steps that led to the discovery of aspirin, and its antiproliferative and anticancer potential are highlighted in this review.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/chemistry
- Anti-Inflammatory Agents, Non-Steroidal/history
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Antineoplastic Agents, Phytogenic/chemistry
- Antineoplastic Agents, Phytogenic/history
- Antineoplastic Agents, Phytogenic/therapeutic use
- Aspirin/chemistry
- Aspirin/history
- Aspirin/therapeutic use
- Benzyl Alcohols/chemistry
- Benzyl Alcohols/history
- Benzyl Alcohols/therapeutic use
- Glucosides
- History, 18th Century
- History, 19th Century
- History, 20th Century
- History, 21st Century
- History, Ancient
- History, Medieval
- Humans
- Salicylic Acid/chemistry
- Salicylic Acid/history
- Salicylic Acid/therapeutic use
- Salix/chemistry
Collapse
Affiliation(s)
- J G Mahdi
- School of Biosciences, Cardiff University, PO Box 911, Cardiff, CF10 3US, UK.
| | | | | | | |
Collapse
|
92
|
Knottenbelt C, Mellor D, Nixon C, Thompson H, Argyle DJ. Cohort study of COX-1 and COX-2 expression in canine rectal and bladder tumours. J Small Anim Pract 2006; 47:196-200. [PMID: 16573762 DOI: 10.1111/j.1748-5827.2006.00094.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVES To determine the role that cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2) play in malignant transformation in canine transitional cell carcinoma and rectal tumours. METHODS Histological sections of 21 canine rectal adenocarcinomas and 18 canine transitional cell carcinomas were stained for COX-1 and COX-2. Mann-Whitney non-parametric tests were applied to determine if there was any relationship between the percentage of cells expressing COX-1 or COX-2, and between COX-1 and COX-2 staining intensity and age, breed or sex. RESULTS For rectal adenocarcinomas, 19.0 per cent of the sections were negative for COX-1 and COX-2. A further 38.1 per cent of the sections were negative for COX-2 but positive for COX-1, and 38.1 per cent of the sections had rare or occasional single cells positive for COX-2. No significant differences were found in COX staining when compared with age, breed or sex. For transitional cell carcinomas, all of the sections were positive for COX-1 and COX-2. For COX-2 staining, 16.7 per cent had more than 30 per cent positive cells. For COX-1 staining, 38.9 per cent had more than 30 per cent positive cells. There was a significant increase in the percentage of COX-1 positive cells in small breed dogs (P = 0.0337). CLINICAL SIGNIFICANCE The variations in COX expression reported in this study may explain the differences in the clinical response of transitional cell carcinomas and rectal adenocarcinomas following treatment with non-steroidal anti-inflammatory drugs.
Collapse
Affiliation(s)
- C Knottenbelt
- Department of Veterinary Clinical Studies, Institute of Comparative Medicine, Faculty of Veterinary Medicine, University of Glasgow, Bearsden Road, Glasgow G61 1QH
| | | | | | | | | |
Collapse
|
93
|
Murray BK, Brown B, Scherer PM, Tomer DP, Garvin KR, Hughes BG, O'Neill KL. Induction of apoptosis in HT-29 human colon adenocarcinoma cells by 13-cis-retinoic acid and vitamin E succinate. Nutr Res 2006. [DOI: 10.1016/j.nutres.2006.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
94
|
Babbar N, Gerner EW, Casero RA. Induction of spermidine/spermine N1-acetyltransferase (SSAT) by aspirin in Caco-2 colon cancer cells. Biochem J 2006; 394:317-24. [PMID: 16262603 PMCID: PMC1386030 DOI: 10.1042/bj20051298] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Epidemiological, experimental and clinical results suggest that aspirin and other NSAIDs (non-steroidal anti-inflammatory drugs) inhibit the development of colon cancer. It has been shown that the NSAID sulindac induces apoptosis and suppresses carcinogenesis, in part, by a mechanism leading to the transcriptional activation of the gene encoding SSAT (spermidine/spermine N1-acetyltransferase), a rate-limiting enzyme in polyamine catabolism. In the present study, we show that a variety of NSAIDs, including aspirin, sulindac, ibuprofen and indomethacin, can induce SSAT gene expression in Caco-2 cells. Aspirin, at physiological concentrations, can induce SSAT mRNA via transcriptional initiation mechanisms. This induction leads to increased SSAT protein levels and enzyme activity. Promoter deletion analysis of the 5' SSAT promoter-flanking region led to the identification of two NF-kappaB (nuclear factor kappaB) response elements. Electrophoretic mobility-shift assays showed binding of NF-kappaB complexes at these sequences after aspirin treatment. Aspirin treatment led to the activation of NF-kappaB signalling and increased binding at these NF-kappaB sites in the SSAT promoter, hence providing a potential mechanism for the induction of SSAT by aspirin in these cells. Aspirin-induced SSAT ultimately leads to a decrease in cellular polyamine content, which has been associated with decreased carcinogenesis. These results suggest that activation of SSAT by aspirin and different NSAIDs may be a common property of NSAIDs that plays an important role in their chemopreventive actions in colorectal cancer.
Collapse
Affiliation(s)
- Naveen Babbar
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | | | | |
Collapse
|
95
|
Gomes-Pereira M, Monckton DG. Chemical modifiers of unstable expanded simple sequence repeats: what goes up, could come down. Mutat Res 2006; 598:15-34. [PMID: 16500684 DOI: 10.1016/j.mrfmmm.2006.01.011] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
A mounting number of inherited human disorders, including Huntington disease, myotonic dystrophy, fragile X syndrome, Friedreich ataxia and several spinocerebellar ataxias, have been associated with the expansion of unstable simple sequence DNA repeats. Despite a similar genetic basis, pathogenesis in these disorders is mediated by a variety of both loss and gain of function pathways. Thus, therapies targeted at downstream pathology are likely to be disease specific. Characteristically, disease-associated expanded alleles in these disorders are highly unstable in the germline and somatic cells, with a tendency towards further expansion. Whereas germline expansion accounts for the phenomenon of anticipation, tissue-specific, age-dependent somatic expansion may contribute towards the tissue-specificity and progressive nature of the symptoms. Thus, somatic expansion presents as a novel therapeutic target in these disorders. Suppression of somatic expansion should be therapeutically beneficial, whilst reductions in repeat length could be curative. It is well established that both cis- and trans-acting genetic modifiers play key roles in the control of repeat dynamics. Importantly, recent data have revealed that expanded CAG.CTG repeats are also sensitive to a variety of trans-acting chemical modifiers. These data provide an exciting proof of principle that drug induced suppression of somatic expansion might indeed be feasible. Moreover, as our understanding of the mechanism of expansion is refined more rational approaches to chemical intervention in the expansion pathway can be envisioned. For instance, the demonstration that expansion of CAG.CTG repeats is dependent on the Msh2, Msh3 and Pms2 genes, highlights components of the DNA mismatch repair pathway as therapeutic targets. In addition to potential therapeutic applications, the response of expanded simple repeats to genotoxic assault suggests such sequences could also have utility as bio-monitors of environmentally induced genetic damage in the soma.
Collapse
Affiliation(s)
- Mário Gomes-Pereira
- Inserm U383, Clinique Maurice Lamy, Hôpital Necker Enfants Malades, 149 rue de Sèvres, 75015 Paris, France
| | | |
Collapse
|
96
|
Yamazaki T, Muramoto M, Oe T, Morikawa N, Okitsu O, Nagashima T, Nishimura S, Katayama Y, Kita Y. Diclofenac, a non-steroidal anti-inflammatory drug, suppresses apoptosis induced by endoplasmic reticulum stresses by inhibiting caspase signaling. Neuropharmacology 2006; 50:558-67. [PMID: 16388830 DOI: 10.1016/j.neuropharm.2005.10.016] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2005] [Revised: 10/07/2005] [Accepted: 10/26/2005] [Indexed: 12/21/2022]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) are frequently used in the treatment of inflammation and pain. In many reports, NSAIDs have induced apoptosis in a variety of cell lines such as colon cancer cells. On the other hand, more recently a few reports have found that NSAIDs protect against apoptosis. Here we investigate endoplasmic reticulum (ER)-stress-induced apoptosis of neuronal cells. The aim of this study is to examine the involvement of NSAIDs, in particular diclofenac, on ER-stress-induced apoptosis of human neuroblastoma SH-SY5Y cells. Diclofenac significantly suppressed SH-SY5Y cell death induced by two types of ER-stress-inducing agents: thapsigargin, an inhibitor of Ca2+-ATPase on the endoplasmic reticulum membrane, and tunicamycin, a glycosylation blocker. Other NSAIDs, such as indomethacin, ibuprofen, aspirin, and ketoprofen, also suppressed ER-stress-induced SH-SY5Y cell death. The dose-dependent anti-apoptotic effect of diclofenac did not correlate with the reduction of prostaglandin release. Administration of prostaglandin E2, which was a primary product of arachidonic metabolism, showed no effects against anti-apoptotic effects produced by diclofenac. Thapsigargin and tunicamycin each significantly activated caspase-3, -9, and -2 in the intrinsic apoptotic pathway in SH-SY5Y cells. Diclofenac suppressed the activation of caspases induced by both ER stresses. Thapsigargin and tunicamycin decreased the mitochondrial membrane potential in SH-SY5Y cells. Diclofenac suppressed the mitochondrial depolarization induced by both ER stresses. Diclofenac inhibited ER-stress-induced apoptosis of SH-SY5Y cells by suppressing the activation of caspases in the intrinsic apoptotic pathway. This is the first report to find that diclofenac has protective effects against ER-stress-induced apoptosis.
Collapse
Affiliation(s)
- Takao Yamazaki
- Pharmacology Research Labs, Astellas Pharma Inc., Tsukuba, Ibaraki 300-2698, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Abstract
Since salicylate, a plant stress hormone, suppresses the growth of various types of cancer cells, it was deemed of interest to investigate whether the jasmonate family of plant stress hormones is endowed with anti-cancer activities. Cell lines representing a wide spectrum of malignancies, including prostate, breast and lung, exhibit sensitivity to the cytotoxic effects of methyl jasmonate (MJ). Jasmonates induced death in leukemic cells isolated from the blood of chronic lymphocytic leukemia (CLL) patients and increased significantly the survival of lymphoma-bearing mice. Among the naturally occurring jasmonates, MJ is the most active, while the synthetic methyl-4,5-didehydrojasmonate, was approximately 29-fold more active than MJ. The cytotoxic activity of MJ is independent of transcription and translation. Studies have suggested several mechanisms of action. It appears that while prolonged exposures to relatively low concentrations of jasmonates induce growth arrest and re-differentiation in myeloid leukemia cells, higher concentrations of MJ induce direct perturbation of cancer cell mitochondria, leading to the release of cytochrome c and eventual cell death. A most important characteristic of jasmonates is their ability to selectively kill cancer cells while sparing normal cells. Even within a mixed population of normal and leukemic cells derived from the blood of CLL patients, MJ killed preferentially the leukemic cells. In conclusion, jasmonates present a unique class of anti-cancer compounds which deserves continued research at the basic and pharmaceutical levels in order to yield novel chemotherapeutic agents against a range of neoplastic diseases.
Collapse
Affiliation(s)
- Eliezer Flescher
- Department of Human Microbiology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
98
|
Lamm W, Vormittag L, Turhani D, Erovic BM, Czembirek C, Eder-Czembirek C, Thurnher D. The effect of nimesulide, a selective cyclooxygenase-2 inhibitor, on Ets-1 and Ets-2 expression in head and neck cancer cell lines. Head Neck 2005; 27:1068-72. [PMID: 16240325 DOI: 10.1002/hed.20285] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The protooncogenes Ets-1 and Ets-2 are involved in carcinogenesis of different tumors. Nimesulide, a selective cyclooxygenase-2 (COX-2) inhibitor, has antiproliferative effects on tumor cells. The question arises whether nimesulide influences Ets-1 and Ets-2 synthesis in head and neck tumors. METHODS Expression of Ets-1 and Ets-2 was analyzed in tumor tissues by immunohistochemistry. The influence of nimesulide and an extracellular signal-regulated kinase (ERK) inhibitor on cell proliferation of two head and neck cancer cell lines and Ets-1 and Ets-2 expression was determined by automated cell counting and Western blotting, respectively. RESULTS Immunohistochemistry showed a high expression of Ets-1 and Ets-2 in tumor tissues. In both cell lines, Ets-1 and Ets-2 expression were reduced after 24 and 48 hours by nimesulide. CONCLUSION Both Ets-1 and Ets-2 are overexpressed in head and neck cancer specimens. Inhibition of Ets-1 and Ets-2 expression in head and neck cancer cell lines by nimesulide might explain the proapoptotic property of this COX-2 inhibitor.
Collapse
Affiliation(s)
- Wolfgang Lamm
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, Waehringer Gürtel 18-20, 1090 Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
99
|
Kudryavtsev IA, Gudkova MV, Pavlova OM, Oreshkin AE, Myasishcheva NV. Lipoxygenase Pathway of Arachidonic Acid Metabolism in Growth Control of Tumor Cells of Different Type. BIOCHEMISTRY (MOSCOW) 2005; 70:1396-403. [PMID: 16417464 DOI: 10.1007/s10541-005-0275-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The influence of inhibitors of different lipoxygenases (LOX) on the growth of human tumor cells with different profiles of synthesized eicosanoids was studied. The studied LOX inhibitors had virtually no influence on the growth of A549 cells actively synthesizing cyclooxygenase and lipoxygenase metabolites of arachidonic acid (AA). The inhibitor of 12-LOX, baicalein, significantly inhibited proliferation in cultures of A431 epidermoid carcinoma cells with a characteristic domination of the major lipoxygenase metabolite of AA, 12-hydroxyeicosatetraenoic acid (12-HETE), in the profile of synthesized eicosanoids and reduced to 70% the incorporation of [3H]thymidine into DNA. Treatment of these cultures with 12-HETE virtually restored the growth potential of the tumor cells. The findings suggest that the lipoxygenase metabolite of AA, 12-HETE, is a growth-limiting factor for tumor cells of definite type.
Collapse
Affiliation(s)
- I A Kudryavtsev
- Institute of Carcinogenesis, Blokhin Russian Cancer Research Center, Russian Academy of Medical Sciences, Moscow, 115478, Russia.
| | | | | | | | | |
Collapse
|
100
|
Christofis P, Katsarou M, Papakyriakou A, Sanakis Y, Katsaros N, Psomas G. Mononuclear metal complexes with Piroxicam: Synthesis, structure and biological activity. J Inorg Biochem 2005; 99:2197-210. [PMID: 16176832 DOI: 10.1016/j.jinorgbio.2005.07.020] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2005] [Revised: 07/28/2005] [Accepted: 07/30/2005] [Indexed: 12/21/2022]
Abstract
Piroxicam (=Hpir) is a non-steroidal anti-inflammatory and an anti-arthritic drug. VO(2+), Mn(2+), Fe(3+), MoO(2)(2+) and UO(2)(2+) complexes with deprotonated piroxicam have been prepared and characterized with the use of infrared, UV-Vis, nuclear magnetic resonance and electron paramagnetic resonance spectroscopies. The experimental data suggest that piroxicam acts as a deprotonated bidentate ligand in all complexes and is coordinated to the metal ion through the pyridine nitrogen and the amide oxygen. Molecular mechanics calculations in the gas state have been performed in order to propose a model for the Fe(3+), VO(2+) and MoO(2)(2+) complexes. Potential anticancer cytostatic and cytotoxic effects of piroxicam complexes with VO(2+), Mn(2+) and MoO(2)(2+) on human promyelocytic leukemia HL-60 cells have been investigated. Among all complexes, only VO(pir)(2)(H(2)O) clearly induces apoptosis after 24-h incubation, whereas piroxicam induces apoptosis after 57-h incubation.
Collapse
Affiliation(s)
- Petros Christofis
- Institute of Physical Chemistry, NCSR Demokritos, 15310 Aghia Paraskevi Attikis, GR15310 Athens, Greece
| | | | | | | | | | | |
Collapse
|