Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Life Sci ; 283: 119849, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34343539

ABSTRACT

AIMS: Cardiotoxicity of doxorubicin frequently complicates treatment outcome. Aberrantly activated calcium/calmodulin pathway can eventually trigger signaling cascades that mediate cardiotoxicity. Therefore, we tested the hypothesis that trifluoperazine, a strong calmodulin antagonist, may alleviate this morbidity. MATERIALS AND METHODS: Heart failure and cardiotoxicity were assessed via echocardiography, PCR, immunohistochemistry, histopathology, Masson's trichrome staining and transmission electron microscopy. Whereas liver and kidney structural and functional alterations were evaluated histopathologically and biochemically. KEY FINDINGS: Results revealed that combination treatment with trifluoperazine could overcome doxorubicin-induced heart failure with reduced ejection fraction. Moreover, heart weight/body weight ratio and histopathological examination showed that trifluoperazine mitigated doxorubicin-induced cardiac atrophy, inflammation and myofibril degeneration. Transmission electron microscopy further confirmed the marked restoration of the left ventricular ultrastructures by trifluoperazine pretreatment. In addition, Masson's trichrome staining revealed that trifluoperazine could significantly inhibit doxorubicin-induced left ventricular remodeling by fibrosis. Of note, doxorubicin induced the expression of myocardial nuclear NF-κB-p65 and caspase-3 which were markedly inhibited by trifluoperazine, suggesting that cardioprotection conferred by trifluoperazine involved, at least in part, suppression of NF-κB and apoptosis. Furthermore, biochemical and histopathological examinations showed that trifluoperazine improved doxorubicin-induced renal and hepatic impairments both functionally and structurally. SIGNIFICANCE: In conclusion, the present in vivo study is the first to provide evidences underscoring the protective effects of trifluoperazine that may pave the way for repurposing this calmodulin antagonist in ameliorating organ toxicity by doxorubicin.


Subject(s)
Apoptosis/drug effects , Cardiotoxicity , Cardiotoxins/adverse effects , Doxorubicin/adverse effects , Myocardium/metabolism , Transcription Factor RelA/metabolism , Trifluoperazine/pharmacology , Animals , Cardiotoxicity/drug therapy , Cardiotoxicity/metabolism , Cardiotoxicity/pathology , Cardiotoxins/pharmacology , Caspase 3/metabolism , Doxorubicin/pharmacology , Male , Mice , Myocardium/pathology
2.
Med Hypotheses ; 146: 110468, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33385878

ABSTRACT

Currently, no single medication has been approved for the management of coronavirus disease-2019 (COVID-19) caused by the new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Therefore, drug repositioningby investigating the use of existing drugs for management of COVID-19 patients is considered a desperate need. Tramadol is a commonly prescribed analgesic drug for treatment of moderate to severe pain with less potential for dependence and respiratory depression. Multiple evidence support that tramadol is a promising drug for treatment of COVID-19 patients. Herein, we discuss the possible beneficial effects of using tramadol against SARS-CoV-2 infection and their underlying mechanism of action. The anti-inflammatory effect of tramadol may help to suppress the COVID-19 related cytokine storm through decreasing interleukin (IL)-6, tumor necrosis factor-alpha (TNF-α), and C-reactive protein (CRP). Besides, tramadol activates natural killer (NK) and T-cells and enhances IL-2 secretion, which produce immune-enhancing effect against SARS-CoV-2. Recent studies confirmed that COVID-19 patients with acute respiratory failure showed increased fibrin formation and polymerization that may lead to thrombosis. Tramadol owing to its hypocoagulable effect may protect against venous thromboembolism in these patients. Moreover, tramadol can exert a cardioprotective effect via decreasing lactate dehydrogenase (LDH) level which is elevated in most of patients with COVID-19. Furthermore, the severity and mortality of COVID-19 have been correlated with old age patients, which may be due to the lack of antioxidant mechanisms and increased oxidative damage. Tramadol could protect COVID-19 patient from disease complications by increases the antioxidant enzymes superoxide dismutase and glutathione peroxidase while diminished malondialdehyde. More interestingly, tramadol as an effective analgesic and antitussive may have a beneficial effect on COVID-19 patients suffering from cough, headache, ache, and pain. The tramadol anti-psychotic effect may also protect against psychiatric disorders associated with SARS-CoV-2 infection. Moreover, tramadol has bactericidal activity against a wide range of pathogens including Pseudomonas aeruginosa which is common in severe COVID-19 patients leading to pneumonia with worse clinical outcomes. Therefore, we hypothesize that tramadol might be a promising adjuvant therapeutic option against SARS-CoV-2 infection. Based on that, tramadol should be considered as adjuvant therapy for COVID-19 clinical trials.


Subject(s)
COVID-19 Drug Treatment , Tramadol/pharmacology , Analgesics, Opioid/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Antidepressive Agents/pharmacology , Antioxidants/pharmacology , COVID-19/complications , COVID-19/physiopathology , Drug Repositioning , Humans , Immunologic Factors/pharmacology , Models, Biological , Pandemics , SARS-CoV-2
3.
Toxicol Appl Pharmacol ; 404: 115185, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32771489

ABSTRACT

The development of resistance to endocrine therapy of estrogen receptor alpha (ERα)-positive breast cancer is inevitable, necessitating the introduction of alternative treatment strategies. Therefore, the current study was carried out to investigate the in vivo efficacy and tolerability of nilotinib/rosuvastatin novel combination against ERα-positive breast carcinoma. Results showed that treatment of tumor-bearing mice with nilotinib/rosuvastatin exerted a significant antitumor activity. Mechanistically, the combination treatment efficiently inhibited the in vivo ERα protein expression, whereas ERα mRNA levels were unaffected suggesting a posttranslational regulation. In addition, the combination treatment markedly downregulated the expression of two ERα downstream target genes: C3 and pS2 confirming the inhibition of ERα signaling in vivo. Further, nilotinib/rosuvastatin combination strongly induced apoptosis evidenced by a marked caspase-3 cleavage and downregulation of tumor nitric oxide levels. Moreover, histopathology showed significant declines in mitotic figures and tumor giant cells implying the in vivo capability of the combination treatment to interfere with cancer cell proliferation and persistence. Of note, the combination treatment abrogated nilotinib-induced hypercholesterolemia and did not adversely affect the liver function or body weight. Overall, the present study provided evidences that warrant further assessment of nilotinib/rosuvastatin combination as an alternative therapeutic modality for ERα-positive breast cancer.


Subject(s)
Adenocarcinoma/veterinary , Estrogen Receptor alpha/antagonists & inhibitors , Mammary Neoplasms, Animal/drug therapy , Pyrimidines/therapeutic use , Rosuvastatin Calcium/therapeutic use , Adenocarcinoma/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Gene Expression Regulation, Neoplastic/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Mice , Pyrimidines/administration & dosage , Rosuvastatin Calcium/administration & dosage
4.
Biomed Pharmacother ; 105: 215-224, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29857301

ABSTRACT

Biodegradable PLGA nanoparticles, loaded with 5-fluorouracil (5FU), were prepared using a double emulsion method and characterised in terms of mean diameter, zeta potential, entrapment efficiency and in vitro release. Poly (vinyl alcohol) was used to modify both internal and external aqueous phases and shown have a significant effect on nanoparticulate size, encapsulation efficiency and the initial burst release. Addition of poly (ethylene glycol) to the particle matrix, as part of the polymeric backbone, improved significantly the encapsulation efficiency. 5FU-loaded NPs were spherical in shape and negatively charged with a size range of 185-350 nm. Biological evaluation was performed in vivo using a solid Ehrlich carcinoma (SEC) murine model. An optimised 5FU-loaded formulation containing PEG as part of a block copolymer induced a pronounced reduction in tumour volume and tumour weight, together with an improved percentage tumour growth inhibition. Drug-loaded nanoparticles showed no significant toxicity or associated changes on liver and kidney function in tested animals, whereas increased alanine aminotransferase, aspartate aminotransferase and serum creatinine were observed in animals treated with free 5FU. Histopathological examination demonstrated enhanced cytotoxic action of 5FU-loaded nanoparticles when compared to the free drug. Based on these findings, it was concluded that nano-encapsulation of 5FU using PEGylated PLGA improved encapsulation and sustained in vitro release. This leads to increased anti-tumour efficacy against SEC, with a reduction in adverse effects.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Ehrlich Tumor/drug therapy , Fluorouracil/therapeutic use , Nanoparticles/chemistry , Polymers/chemistry , Animals , Carcinoma, Ehrlich Tumor/blood , Carcinoma, Ehrlich Tumor/pathology , Drug Compounding , Female , Fluorouracil/pharmacology , Mice , Nanoparticles/ultrastructure , Particle Size , Polyethylene Glycols/chemistry , Polyglycolic Acid , Polyvinyl Alcohol/chemistry , Tumor Burden
5.
Anticancer Res ; 35(6): 3235-43, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26026083

ABSTRACT

BACKGROUND/AIM: The current study investigated the mechanisms underlying the antitumor activity of SB265610, a cysteine-amino acid-cysteine (CXC) chemokines receptor 2 (CXCR2) antagonist. MATERIALS AND METHODS: Cell-cycle progression and regulatory molecules were assessed by flow cytometry, immunoblotting, real-time PCR and immunoprecipitation. Target validation was achieved via RNA interference. RESULTS: G1 arrest induced by SB265610 occurred at concentrations lacking CXCR2 selectivity, persisted upon interleukin 8 (IL8) challenge, and did not affect IL8 downstream target expression. Profiling of G1 regulators revealed cyclin-dependent kinase 2 (CDK2) (Thr160) hypophosphorylation, cyclin D3 gene down-regulation, and p21 post-translational induction. However, only cyclin D3 and CDK2 contributed towards G1 arrest. Furthermore, SB265610 induced a sustained phosphorylation of the p38MAPK. Pharmacological interference with p38MAPK significantly abrogated SB265610-induced G1 arrest and normalized the expression of cyclin D3, with restoration of its exclusive binding to CDK6, but with weak recovery of CDK2 (Thr160) hypo-phosphorylation. CONCLUSION: The present study described the mechanisms for the anti-proliferative activity of SB265610 which may be of value in IL8-rich tumor microenvironments.


Subject(s)
Cyclin D3/biosynthesis , Cyclin-Dependent Kinase 2/biosynthesis , Phenylurea Compounds/administration & dosage , Prostatic Neoplasms/genetics , Receptors, Interleukin-8B/biosynthesis , Triazoles/administration & dosage , Cell Cycle Checkpoints/drug effects , Cyclin D3/genetics , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , G1 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-8/genetics , Male , Phosphorylation , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Receptors, Interleukin-8B/genetics , Tumor Microenvironment/drug effects , p38 Mitogen-Activated Protein Kinases/genetics
6.
Mol Oncol ; 9(1): 309-22, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25245324

ABSTRACT

Novel combinations aiming at maximizing the efficacy of bortezomib are highly valued in the clinic. Therefore the current study investigated the outcomes of combining bortezomib with dipyridamole, a well-known antiplatelet. The co-treatment exerted a synergistic lethality in a panel of human leukemia/lymphoma cell lines of different origin. Mechanistically, dipyridamole did not modulate the proteasome inhibitory activity of bortezomib. However, dipyridamole triggered an endoplasmic reticulum (ER) stress, and co-treatment with bortezomib resulted in higher levels of ER stress than either monotherapies. Relieving ER stress with the protein translation inhibitor, cycloheximide suppressed cell death. Moreover, the enhanced ER stress by the co-treatment was associated with an aggravation of reactive oxygen species (ROS) generation and glutathione (GSH) depletion. Replenishing GSH pools significantly scavenged ROS and rescued the cells. Importantly, the cytotoxicity of the co-treatment was executed mainly via the mitochondrial apoptotic pathway with an efficient suppression of the key anti-apoptotic regulators, Mcl-1, Bcl-xl, Bcl-2 and XIAP, driving the independence of the co-treatment-induced apoptosis of a single apoptotic trigger. Furthermore, the intrinsic potential of bortezomib to inhibit important pro-survival pathways was enhanced by dipyridamole in a GSH/ROS-dependent manner. Interestingly, dipyridamole abrogated JAK2 phosphorylation indirectly and selectively in cancer cells, and the co-treatment-induced cytotoxicity was preserved in a model of stromal-mediated chemoresistance. In nude mice, the antitumor activity of the co-treatment surpassed that of bortezomib monotherapy despite that synergy was lacking. In summary, findings of the present study provided a preclinical rationale which warrants further clinical evaluation of bortezomib/dipyridamole novel combination in hematologic malignancies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Hematologic Neoplasms/drug therapy , Animals , Bortezomib/pharmacology , Dipyridamole/pharmacology , HL-60 Cells , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Humans , Jurkat Cells , K562 Cells , Mice , Mice, Nude , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , U937 Cells , Xenograft Model Antitumor Assays
7.
Oncol Rep ; 31(2): 947-54, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24276615

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is known to cause apoptosis in several types of malignant tumor cells through its interaction with the death domain-containing receptor, death receptor 5 (DR5). In the present study, we showed that co-treatment with troglitazone (TGZ), a synthetic ligand of peroxisome proliferator-activated receptor γ (PPARγ), and TRAIL synergistically induced apoptosis through DR5 upregulation in human colon cancer DLD-1 cells. TGZ elevated DR5 expression at the promoter level through the CCAAT/enhancer-binding protein homologous protein (CHOP) binding site. These results suggest that combined treatment with TGZ and TRAIL may be promising as a new therapy against malignant tumors.


Subject(s)
Apoptosis/genetics , Chromans/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Thiazolidinediones/pharmacology , Transcription Factor CHOP/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/pharmacology , Binding Sites , Cell Line, Tumor , Colonic Neoplasms/pathology , Drug Synergism , Endoplasmic Reticulum Stress/physiology , Humans , PPAR gamma , Promoter Regions, Genetic , Protein Binding , RNA Interference , RNA, Small Interfering , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Transcription Factor CHOP/biosynthesis , Transcription Factor CHOP/genetics , Troglitazone
8.
Biochem Pharmacol ; 85(12): 1741-52, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23611835

ABSTRACT

SB225002 (SB) is an IL-8 receptor B (IL-8RB) antagonist that has previously been shown to inhibit IL-8-based cancer cell invasion, and to possess in vivo anti-inflammatory and anti-nociceptive effects. The present study presented an evidence for the cell cycle-targeting activity of SB in a panel of p53-mutant human cancer cell lines of different origin, and investigated the underlying molecular mechanisms. A combination of cell cycle analysis, immunocytometry, immunoblotting, and RNA interference revealed that SB induced a BubR1-dependent mitotic arrest. Mechanistically, SB was shown to possess a microtubule destabilizing activity evidenced by hyperphosphorylation of Bcl2 and BclxL, suppression of microtubule polymerization and induction of a prometaphase arrest. Molecular docking studies suggested that SB has a good affinity toward vinblastine-binding site on ß-tubulin subunit. Of note, SB265610 which is a close structural analog of SB225002 with a potent IL-8RB antagonistic activity did not exhibit a similar antimitotic activity. Importantly, in P-glycoprotein overexpressing NCI/Adr-Res cells the antitumor activity of SB was unaffected by multidrug resistance. Interestingly, the mechanisms of SB-induced cell death were cell-line dependent, where in invasive hepatocellular carcinoma HLE cells the significant contribution of BAK-dependent mitochondrial apoptosis was demonstrated. Conversely, SB activated p38 MAPK signaling in colorectal adenocarcinoma cells SW480, and pharmacologic inhibition of p38 MAPK activity revealed its key role in mediating SB-induced caspase-independent cell death. In summary, the present study introduced SB as a promising antitumor agent which has the potential to exert its activity through dual mechanisms involving microtubules targeting and interference with IL-8-drivin cancer progression.


Subject(s)
Antineoplastic Agents/pharmacology , Microtubules/drug effects , Phenylurea Compounds/pharmacology , Tubulin Modulators/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Humans , Microtubules/metabolism , Phenylurea Compounds/chemistry , Receptors, Interleukin-8/antagonists & inhibitors , Receptors, Interleukin-8/metabolism , Tubulin Modulators/chemistry , U937 Cells
9.
Mol Cancer Ther ; 11(10): 2294-300, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22784708

ABSTRACT

Methylglyoxal is an essential component in glycolysis and is known to be an inducer of apoptosis. Glyoxalase I (GLO1) metabolizes and inactivates methylglyoxal. GLO1 is known to be overexpressed in cancer cells and causes resistance to anticancer agents. We show for the first time that methylglyoxal treatment or the silencing of GLO1 enhances sensitivity to the promising anticancer agent TRAIL in malignant tumor cells. Methylglyoxal suppressed the expression of antiapoptotic factors, X-linked inhibitor of apoptosis protein (XIAP), survivin, cIAP1, Bcl-2, and Bcl-xL, without affecting TRAIL receptors, DR4 and DR5. Knockdown of XIAP or survivin by siRNA also enhanced TRAIL-induced apoptosis, indicating that downregulation of XIAP and survivin expression by methylglyoxal contributes to the enhancement of TRAIL activity. Furthermore, methylglyoxal decreased NF-κB activity with or without TRAIL treatment. On the other hand, the knockdown of GLO1 by siRNA enhanced TRAIL-induced apoptosis via the downregulation of XIAP and survivin expression. In conclusion, our results strongly suggest that sensitivity to TRAIL is increased by inhibition of the glyoxalase pathway and that the combination of TRAIL with methylglyoxal or glyoxalase inhibitors may be useful for a novel combination chemotherapy.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Down-Regulation/drug effects , Lactoylglutathione Lyase/antagonists & inhibitors , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/metabolism , Caspases/metabolism , Cell Line, Tumor , Down-Regulation/genetics , Drug Screening Assays, Antitumor , G1 Phase/drug effects , G1 Phase/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Lactoylglutathione Lyase/metabolism , NF-kappa B/metabolism , Neoplasms/enzymology , Neoplasms/genetics , Pyruvaldehyde/pharmacology , Survivin , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism
10.
Int J Oncol ; 38(2): 365-74, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21165560

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is one of the most promising anti-cancer agents, but some tumor types develop resistance to TRAIL. Here, we report that chetomin, an inhibitor of hypoxia-inducible factors, is a potent enhancer of TRAIL-induced apoptosis. TRAIL or chetomin alone weakly induced apoptosis, but the combination of chetomin and TRAIL synergistically induced apoptosis in prostate cancer PC-3 cells. The combination of chetomin and TRAIL induces the activation of caspase-3, -8, -9 and -10. Among the apoptotic factors related to the TRAIL pathway, chetomin markedly decreased the X-linked inhibitor of apoptosis (XIAP) protein levels in a dose-dependent manner, but other IAP family members, TRAIL receptors and Bcl-2 family members were not altered by chetomin. Using XIAP siRNA instead of chetomin, down-regulation of XIAP sensitized PC-3 cells to TRAIL-induced apoptosis. Conversely, transient transfection of XIAP reduced the apoptotic response to combined treatment with chetomin and TRAIL. Treatment with chetomin induced a rapid decrease in XIAP protein levels but had no effect on XIAP mRNA levels. Since chetomin-mediated XIAP down-regulation was completely prevented by proteasome inhibitors, it was suggested that chetomin induces the degradation of the XIAP protein in a proteasome-dependent manner. Additionally, chetomin also sensitized renal cancer Caki-1 cells and bladder cancer UM-UC-3 cells to TRAIL-induced apoptosis via down-regulation of XIAP. Co-treatment of chetomin and TRAIL did not enhance apoptosis in normal peripheral blood mononuclear cells (PBMC). Taken together, these findings suggest that TRAIL and chetomin synergistically induce apoptosis in human urogenital cancer cells through a mechanism that involves XIAP down-regulation by chetomin.


Subject(s)
Disulfides/pharmacology , Indole Alkaloids/pharmacology , Kidney Neoplasms/therapy , Prostatic Neoplasms/therapy , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Urinary Bladder Neoplasms/therapy , X-Linked Inhibitor of Apoptosis Protein/metabolism , Apoptosis/drug effects , Blotting, Western , Caspases/metabolism , Cell Proliferation/drug effects , Drug Synergism , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Male , Mycotoxins/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors , X-Linked Inhibitor of Apoptosis Protein/genetics
11.
Clin Cancer Res ; 16(8): 2320-32, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20371690

ABSTRACT

PURPOSE: The clinically relevant histone deacetylase inhibitors (HDI) valproic acid (VPA) and suberoylanilide hydroxamic acid exert variable antitumor activities but increase therapeutic efficacy when combined with other agents. The natural endogenous ligand of peroxisome proliferator-activated receptor gamma 15-deoxy-Delta(12,14)-prostaglandin J(2) (15d-PGJ(2)) is a potent antineoplastic agent. Therefore, we investigated whether these HDIs in combination with 15d-PGJ(2) could show synergistic antitumor activity in colon cancer DLD-1 cells. EXPERIMENTAL DESIGN: Cell viability was determined using a Cell Counting Kit-8 assay. Apoptosis and reactive oxygen species (ROS) generation were determined using flow cytometry analysis. Western blotting and real-time reverse transcription-PCR analysis were carried out to investigate the expression of apoptosis-related molecules. Mice bearing DLD-1 xenograft were divided into four groups (n = 5) and injected everyday (i.p.) with diluent, VPA (100 mg/kg), 15d-PGJ(2) (5 mg/kg), or a combination for 25 days. RESULTS: HDI/15d-PGJ(2) cotreatments synergistically induced cell death through caspase-dependent apoptosis in DLD-1 cells. Moreover, HDIs/15d-PGJ(2) caused histone deacetylase inhibition, leading to subsequent ROS generation and endoplasmic reticulum stress to decrease the expression of antiapoptotic molecules Bcl-X(L) and XIAP and to increase that of proapoptotic molecules CAAT/enhancer binding protein homologous protein and death receptor 5. Additionally, VPA/15d-PGJ(2) cotreatment induced ROS-dependent apoptosis in other malignant tumor cells and was more effective than a VPA or 15d-PGJ(2) monotherapy in vivo. CONCLUSIONS: Cotreatments with the clinically relevant HDIs and the endogenous peroxisome proliferator-activated receptor gamma ligand 15d-PGJ(2) are promising for the treatment of a broad spectrum of malignant tumors.


Subject(s)
Apoptosis , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Prostaglandin D2/analogs & derivatives , Animals , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Drug Synergism , Humans , Luciferases/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prostaglandin D2/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reactive Oxygen Species/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Vorinostat , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism , bcl-X Protein/genetics , bcl-X Protein/metabolism
12.
Mol Cancer Res ; 6(12): 1852-60, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19074830

ABSTRACT

Allopurinol has been used for the treatment of gout and conditions associated with hyperuricemia for several decades. We explored the potential of allopurinol on cancer treatment. Allopurinol did not expose cytotoxicity as a single treatment in human hormone refractory prostate cancer cell lines, PC-3 and DU145. However, allopurinol drastically induced apoptosis of PC-3 and DU145 in combination with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), which is a promising candidate for anticancer agent but its efficacy is limited by the existence of resistant cancer cells. We examined the underlying mechanism by which allopurinol overcomes the resistance of prostate cancer cells to TRAIL. Allopurinol up-regulated the expression of a proapoptotic TRAIL receptor, death receptor 5 (DR5). Allopurinol increased DR5 protein, mRNA, and promoter activity. Using DR5 small interfering RNA (siRNA), we showed that allopurinol-mediated DR5 up-regulation contributed to the enhancement of TRAIL effect by allopurinol. Furthermore, we examined the mechanism of allopurinol-mediated DR5 up-regulation. DR5 promoter activity induced by allopurinol was diminished by a mutation of a CAAT/enhancer binding protein homologous protein (CHOP)-binding site. In addition, allopurinol also increased CHOP expression, suggesting that allopurinol induced DR5 expression via CHOP. Allopurinol possesses the activity of a xanthine oxidase (XO) inhibitor. We used XO siRNA instead of allopurinol. XO siRNA also up-regulated DR5 and CHOP expression and sensitized the prostate cancer cells to TRAIL-induced apoptosis. Here, we show the novel potential of allopurinol in cancer treatment and indicate that the combination of allopurinol with TRAIL is effective strategy to expand the TRAIL-mediated cancer therapy.


Subject(s)
Allopurinol/pharmacology , Antimetabolites/pharmacology , Apoptosis/drug effects , Prostatic Neoplasms/drug therapy , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Therapy, Combination , Endoplasmic Reticulum/metabolism , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Promoter Regions, Genetic/physiology , Prostatic Neoplasms/pathology , RNA, Small Interfering , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Response Elements , Transcription Factor CHOP/genetics , Up-Regulation/drug effects , Xanthine Oxidase/genetics
13.
Cancer Res ; 68(21): 8918-27, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18974136

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is one of the most promising candidates for new cancer therapeutics. A current problem is that some cancers still remain resistant to TRAIL. We show for the first time that a naturally occurring flavonoid, baicalein, overcomes TRAIL resistance in cancer cells. The combination of baicalein and TRAIL effectively induced apoptosis in TRAIL-resistant colon cancer SW480 cells. Baicalein up-regulated the expression of death receptor 5 (DR5) among TRAIL receptors at the mRNA and protein levels. Suppression of this up-regulation with small interfering RNA (siRNA) efficiently reduced the apoptosis induced by TRAIL and baicalein, suggesting that the sensitization was mediated through DR5 induction. Moreover, baicalein also overcame TRAIL resistance with DR5 up-regulation in prostate cancer PC3 cells. Of note, the combination of TRAIL and baicalein hardly induced apoptosis in normal human cells, such as blood cells and hepatocytes. Baicalein increased DR5 promoter activity, and this enhanced activity was diminished by mutation of a CCAAT/enhancer-binding protein homologous protein (CHOP)-binding site in SW480 cells. In SW480 cells, CHOP siRNA blocked both functions of baicalein. CHOP expression was induced by baicalein in SW480 cells; however, in PC3 cells, baicalein scarcely induced CHOP and mutation of the CHOP-binding site did not abrogate the DR5 promoter activation by baicalein. Interestingly, baicalein induced reactive oxygen species (ROS) and a ROS scavenger prevented DR5 expression and TRAIL sensitization in PC3 but not SW480 cells. These results indicate that, using two different pathways, baicalein exposes cancer surveillance of TRAIL and overcomes TRAIL resistance in cancer cells.


Subject(s)
Flavanones/pharmacology , TNF-Related Apoptosis-Inducing Ligand/physiology , Apoptosis/drug effects , Apoptosis/physiology , Blotting, Western , Cell Line, Tumor , Down-Regulation , Humans , RNA, Messenger/genetics , RNA, Small Interfering , Reactive Oxygen Species/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/physiology , TNF-Related Apoptosis-Inducing Ligand/genetics , Transcription Factor CHOP/metabolism
14.
Biochem Biophys Res Commun ; 375(1): 129-33, 2008 Oct 10.
Article in English | MEDLINE | ID: mdl-18680719

ABSTRACT

Kaempferol is a natural compound contained in edible plants, and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising anti-cancer agent. Here, we show for the first time that the combined treatment with kaempferol and TRAIL drastically induced apoptosis in human colon cancer SW480 cells, compared to single treatments. Kaempferol markedly up-regulated TRAIL receptors, DR5 and DR4. DR5 but not DR4 siRNA efficiently blocked apoptosis induced by the co-treatment with kaempferol and TRAIL, indicating that DR5 up-regulation by kaempferol helps to enhance TRAIL actions. Moreover, we examined the combined effect on normal human cells. The co-treatment induced no apoptosis in normal human peripheral blood mononuclear cells and little apoptosis in normal human hepatocytes. These results suggest that kaempferol is useful for TRAIL-based treatments for cancer.


Subject(s)
Apoptosis , Colonic Neoplasms/metabolism , Kaempferols/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Caspases/metabolism , Cell Line, Tumor , Humans , Poly(ADP-ribose) Polymerases/metabolism , RNA, Small Interfering/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Up-Regulation
15.
Cancer Sci ; 98(9): 1417-23, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17645780

ABSTRACT

Lipoxygenases induce malignant tumor progression and lipoxygenase inhibitors have been considered as promising anti-tumor agents. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is one of the most promising candidates for new cancer therapeutics. Combined treatment with nordihydroguaiaretic acid (NDGA), a lipoxygenase inhibitor, and TRAIL markedly induced apoptosis in Jurkat T-cell leukemia cells at suboptimal concentrations for each agent. The combined treatment efficiently activated caspase-3, -8 and -10, and Bid. The underling mechanism by which NDGA enhanced TRAIL-induced apoptosis was examined. NDGA did not change the expression levels of anti-apoptotic factors, Bcl-x(L), Bcl-2, cIAP-1, XIAP and survivin. The expression of death receptor-related genes was investigated and it was found that NDGA specifically up-regulated the expression of death receptor 5 (DR5) at mRNA and protein levels. Down-regulation of DR5 by small interfering RNA prevented the sensitizing effect of NDGA on TRAIL-induced apoptosis. Furthermore, NDGA sensitized prostate cancer and colorectal cancer cells to TRAIL-induced apoptosis. In contrast, NDGA neither enhanced TRAIL-induced apoptosis nor up-regulated DR5 expression in normal peripheral blood mononuclear cells. Another lipoxygenase inhibitor, AA861, also up-regulated DR5 and sensitized Jurkat and DU145 cells to TRAIL. These results indicate that lipoxygenase inhibitors augment the apoptotic efficiency of TRAIL through DR5 up-regulation in malignant tumor cells, and raise the possibility that the combination of lipoxygenase inhibitor and TRAIL is a promising strategy for malignant tumor treatment.


Subject(s)
Apoptosis/drug effects , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/pathology , Lipoxygenase Inhibitors/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/physiology , Apoptosis/genetics , Apoptosis/physiology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/physiology , Benzoquinones/pharmacology , Cell Line, Tumor , Down-Regulation/drug effects , Drug Synergism , Humans , Immunoglobulin Fc Fragments/genetics , Jurkat Cells , Leukemia, T-Cell/enzymology , Masoprocol/pharmacology , Mutant Chimeric Proteins/physiology , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand/physiology , Up-Regulation/drug effects , Up-Regulation/genetics
16.
Pharmacol Res ; 50(5): 487-98, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15458769

ABSTRACT

The potential anti-tumour activity of non-steroidal anti-inflammatory drugs (NSAIDS) has been previously discussed. This study was undertaken to assess the possible anti-tumour activity of the cyclooxygenase-2 (COX-2) inhibitor; celecoxib in an animal model of mammary carcinoma; the solid Ehrlich carcinoma (SEC). The possibility that celecoxib may modulate the anti-tumour activity of doxorubicin on the SEC was also studied. Some of the possible mechanisms underlying such modulation were investigated. The anti-tumour activity of celecoxib (25 mg kg(-1)), diclofenac (12.5 mg kg(-1)) and doxorubicin (2 mg kg(-1)) either alone or in combination were investigated on SEC in vivo through the assessment of tumour growth delay (TGD) and tumour volume (TV), changes in tumour DNA content and nitric oxide (NO) levels, immunohistochemical staining of the tumour suppressor gene product; p53 histopathological examination and determination of apoptotic index of SEC. In addition, the influence of these drugs on the DNA fragmentation pattern of Ehrlich carcinoma cells (ECC) was studied. It was found that both celecoxib and diclofenac lack the anti-tumour activity on SEC. In addition there was a significant increase in doxorubicin anti-tumour activity when administered in combination with celecoxib. Moreover, it was found that both celecoxib and diclofenac have the potential to inhibit the function of P-glycoprotein (P-gp) in ECC using rhodamine uptake and efflux assays. Therefore, the current study suggested the chemosensitizing potential of celecoxib in the SEC animal model of mammary tumour, which could be explained in part on the basis of inhibition of P-gp function, with possible enhancement of doxorubicin anti-tumour activity.


Subject(s)
Cyclooxygenase Inhibitors/therapeutic use , Doxorubicin/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Prostaglandin-Endoperoxide Synthases/metabolism , Pyrazoles/therapeutic use , Sulfonamides/therapeutic use , Animals , Celecoxib , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Doxorubicin/pharmacology , Drug Synergism , Drug Therapy, Combination , Female , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Mice , Pyrazoles/pharmacology , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...