Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters











Database
Language
Publication year range
1.
Cancer Sci ; 101(7): 1610-7, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20367642

ABSTRACT

Toll-like receptor 3 (TLR3) has gained recognition as a novel molecular target for cancer therapy because TLR3 activation by its synthetic ligand poly I:C directly causes tumor cell death. Recently, we reported that tumor suppressor p53 increases the expression of TLR3 in several tumor cell lines. Another study also showed that interferon-alpha (IFN-alpha) up-regulates TLR3 expression. We thus hypothesized that various anticancer drugs such as p53-activating reagents and IFNs may potentiate poly I:C-induced tumor cell death through the up-regulation of TLR3 expression. Here, we screened several anticancer drugs that, together with poly I:C, effectively cause tumor cell death in colon carcinoma HCT116 cells. We found that the DNA-damaging reagent 5-fluorouracil (5-FU) increased TLR3 mRNA expression and potentiated poly I:C-induced apoptosis in HCT116 p53(+/+) cells but had only minimal effect in p53(-/-) cells, indicating a p53-dependent pathway. On the other hand, IFN-alpha increased poly I:C-induced apoptosis and the TLR3 mRNA level in HCT116 p53(+/+) and p53(-/-) cell lines. Furthermore, the combination of poly I:C, 5-FU and IFN-alpha induced the highest apoptosis in HCT116 p53(+/+) and p53(-/-) cells. Taken together, these data suggest that the anticancer drugs increased TLR3 expression and subsequently potentiated poly I:C-induced apoptosis likely via p53-dependent and -independent pathways. Considering that the p53 status in malignant cells is heterogeneous, this combination approach may provide a highly effective tumor therapy.


Subject(s)
Apoptosis/drug effects , Poly I-C/pharmacology , Toll-Like Receptor 3/biosynthesis , Adenocarcinoma/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line , Cell Line, Tumor , Colorectal Neoplasms/genetics , DNA Damage/drug effects , Fluorouracil/pharmacology , Humans , Interferon-alpha/pharmacology , Interferon-beta/pharmacology , Kidney , Lung Neoplasms/genetics , Mice , Poly I-C/genetics , Toll-Like Receptor 3/drug effects , Toll-Like Receptor 3/genetics
2.
Mol Cell Biol ; 28(21): 6557-67, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18779317

ABSTRACT

Toll-like receptors (TLRs) are important sensors of microbial pathogens and mediators of innate immune responses. Although the signal transduction of TLRs is well elucidated, their basal regulation is largely unexplored. Here we show that the tumor suppressor p53 positively regulates the transcription of TLR3, a receptor for viral double-stranded RNA and poly(I-C), by binding to the p53 site in the TLR3 promoter. TLR3 expression was lower in HCT116 p53(-/-) cells than in HCT116 p53(+/+) cells. Activation of p53 by 5-fluorouracil increased the TLR3 mRNA in epithelial cell lines with wild-type p53 but not in cell lines harboring mutant p53. Knockdown of p53 by small interfering RNA decreased the TLR3 expression. TLR3 mRNA was also lower in liver and intestine of p53(-/-) mice than in p53(+/+) mice. Furthermore, the poly(I-C)-induced phosphorylation of IkappaB-alpha, nuclear translocation of NF-kappaB, and phosphorylation of interferon regulatory transcription factor 3, were drastically reduced in HCT116 p53(-/-) cells, indicating a dysregulation of the two signaling pathways governed by TLR3. Consequently, induction of interleukin-8 and beta interferon after poly(I-C) stimulation was impaired in HCT116 p53(-/-) cells. These results suggest that p53 influences TLR3 expression and function and highlight a role of p53 in innate immune response in epithelial cells.


Subject(s)
Epithelial Cells/metabolism , Toll-Like Receptor 3/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Cytokines/genetics , Cytokines/metabolism , Epithelial Cells/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intestinal Mucosa/metabolism , Liver/metabolism , Mice , Poly I-C/pharmacology , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 3/metabolism , Transcriptional Activation/drug effects
3.
Int J Oncol ; 32(6): 1317-24, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18497994

ABSTRACT

Hyperthermia is used as one of the treatment modalities for various types of cancer, but the acquisition of thermotolerance in cancer cells, through the induction of heat shock proteins (Hsps), renders hyperthermia less effective. Among the Hsp family members, Hsp27 is frequently associated with thermotolerance and chemoresistance. Thus, down-regulation of Hsp27 expression during hyperthermic or chemotherapeutic applications is a promising approach to efficient tumor treatment. In the present study, we found that the cytokine interferon-gamma (IFN-gamma) suppresses the basal, the heat shock-induced and the cisplatin-induced expression of Hsp27 in HSC-2 (oral squamous carcinoma) and A549 (lung cancer) cells but not in 16HBE14o- (normal bronchial epithelial cells). Neither IFN-alpha nor IFN-beta affected Hsp27 expression, suggesting the specificity of IFN-gamma. We also demonstrate here that IFN-gamma suppresses Hsp27 basal transcription and promoter activity, and this is mediated specifically through one of the two Sp1 sites in the proximal region of the Hsp27 promoter. More importantly, pre-treatment of cells with IFN-gamma enhanced the induction of cell death by hyperthermia and cisplatin treatments in the tumor cell lines, HSC-2 and A549, but has no effect in 16HBE14o-, indicating a tumor cell-specific effect of IFN-gamma. Furthermore, the combination treatment of hyperthermia and IFN-gamma suppressed tumor growth in vivo more effectively than hyperthermia treatment alone. Together, our findings propose that IFN-gamma could be a useful potentiator of hyperthermia and cisplatin in cancer therapy.


Subject(s)
Apoptosis/drug effects , Heat-Shock Proteins/metabolism , Hyperthermia, Induced , Interferon-gamma/pharmacology , Lung Neoplasms/pathology , Mouth Neoplasms/pathology , Neoplasm Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cells, Cultured , Cisplatin/pharmacology , Combined Modality Therapy , Down-Regulation/drug effects , Female , Gene Expression Regulation, Neoplastic , HSP27 Heat-Shock Proteins , Humans , In Vitro Techniques , Lung Neoplasms/metabolism , Mice , Mice, Inbred ICR , Mice, Nude , Molecular Chaperones , Mouth Neoplasms/metabolism , Promoter Regions, Genetic/genetics , Recombinant Proteins , Sp1 Transcription Factor/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL