Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Oncotarget ; 8(24): 38969-38977, 2017 Jun 13.
Article in English | MEDLINE | ID: mdl-28465494

ABSTRACT

Interferon regulatory factor 2 (IRF-2) is known to play a pivotal role in the development and progression of several malignancies. As a crucial member of interferon regulatory factor family, the association between the expression of IRF-2 and clinical prognostic significance has not been fully explored in colorectal cancer (CRC). The purpose of our study was to investigate the expression profile of IRF-2 in CRC and to examine its association with clinical features. The expression levels of IRF-2 in 18 paired CRC and non-cancerous colorectal tissues were measured by quantitative real-time PCR (qRT-PCR) and those in 4 paired samples by Western blotting. The results showed a significant increase in IRF-2 mRNA expression and protein expression in CRC tissues compared to those in paired normal tissues. Besides, high expression of IRF-2 was significantly associated with distant metastasis (P = 0.041) and preoperative serum CEA level (P = 0.045). Kaplan-Meier survival analysis showed that patients with high expression of IRF-2 had a significantly worse overall survival than those with low expression of IRF-2 (P = 0.006). Further multivariate analysis indicated that IRF-2 and TNM stage were independent prognostic factors for overall survival in patients with CRC. Our study primarily suggests IRF-2 as a potential prognostic biomarker in CRC.


Subject(s)
Adenocarcinoma/pathology , Biomarkers, Tumor/analysis , Colorectal Neoplasms/pathology , Interferon Regulatory Factor-2/biosynthesis , Adenocarcinoma/mortality , Adult , Aged , Colorectal Neoplasms/mortality , Disease-Free Survival , Female , Humans , Interferon Regulatory Factor-2/analysis , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Proportional Hazards Models
2.
Gut ; 63(4): 665-73, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23787026

ABSTRACT

BACKGROUND: IL-7 and IL-15 are produced by hepatocytes and are critical for the expansion and function of CD8 T cells. IL-15 needs to be presented by IL-15Rα for efficient stimulation of CD8 T cells. METHODS: We analysed the hepatic levels of IL-7, IL-15, IL-15Rα and interferon regulatory factors (IRF) in patients with chronic hepatitis C (CHC) (78% genotype 1) and the role of IRF1 and IRF2 on IL-7 and IL-15Rα expression in Huh7 cells with or without hepatitis C virus (HCV) replicon. RESULTS: Hepatic expression of both IL-7 and IL-15Rα, but not of IL-15, was reduced in CHC. These patients exhibited decreased hepatic IRF2 messenger RNA levels and diminished IRF2 staining in hepatocyte nuclei. We found that IRF2 controls basal expression of both IL-7 and IL-15Rα in Huh7 cells. IRF2, but not IRF1, is downregulated in cells with HCV genotype 1b replicon and this was accompanied by decreased expression of IL-7 and IL-15Rα, a defect reversed by overexpressing IRF2. Treating Huh7 cells with IFNα plus oncostatin M increased IL-7 and IL-15Rα mRNA more intensely than either cytokine alone. This effect was mediated by strong upregulation of IRF1 triggered by the combined treatment. Induction of IRF1, IL-7 and IL-15Rα by IFNα plus oncostatin M was dampened in replicon cells but the combination was more effective than either cytokine alone. CONCLUSIONS: HCV genotype 1 infection downregulates IRF2 in hepatocytes attenuating hepatocellular expression of IL-7 and IL-15Rα. Our data reveal a new mechanism by which HCV abrogates specific T-cell responses and point to a novel therapeutic approach to stimulate anti-HCV immunity.


Subject(s)
Hepacivirus/physiology , Hepatitis C, Chronic/physiopathology , Hepatocytes/physiology , Interferon Regulatory Factors/physiology , Blotting, Western , CD8-Positive T-Lymphocytes/physiology , Gene Expression Regulation, Viral/genetics , Gene Expression Regulation, Viral/physiology , Genotype , Hepacivirus/genetics , Hepacivirus/metabolism , Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/virology , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Interferon Regulatory Factor-1/biosynthesis , Interferon Regulatory Factor-1/physiology , Interferon Regulatory Factor-2/biosynthesis , Interferon Regulatory Factor-2/physiology , Interleukin-15/biosynthesis , Interleukin-15/physiology , Interleukin-15 Receptor alpha Subunit/biosynthesis , Interleukin-15 Receptor alpha Subunit/physiology , Interleukin-7/biosynthesis , Interleukin-7/physiology , Real-Time Polymerase Chain Reaction , Virus Replication/physiology
3.
J Cell Sci ; 126(Pt 9): 1969-80, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23487038

ABSTRACT

The B-aggressive lymphoma-1 protein and ADP-ribosyltransferase BAL1/ARTD9 has been recently identified as a risk-related gene product in aggressive diffuse large B-cell lymphoma (DLBCL). BAL1 is constitutively expressed in a subset of high-risk DLBCLs with an active host inflammatory response and has been suggested to be associated with interferon-related gene expression. Here we identify BAL1 as a novel oncogenic survival factor in DLBCL and show that constitutive overexpression of BAL1 in DLBCL tightly associates with intrinsic interferon-gamma (IFNγ) signaling and constitutive activity of signal transducer and activator of transcription (STAT)-1. Remarkably, BAL1 stimulates the phosphorylation of both STAT1 isoforms, STAT1α and STAT1ß, on Y701 and thereby promotes the nuclear accumulation of the antagonistically acting and transcriptionally repressive isoform STAT1ß. Moreover, BAL1 physically interacts with both STAT1α and STAT1ß through its macrodomains in an ADP-ribosylation-dependent manner. BAL1 directly inhibits, together with STAT1ß, the expression of tumor suppressor and interferon response factor (IRF)-1. Conversely, BAL1 enhances the expression of the proto-oncogenes IRF2 and B-cell CLL/lymphoma (BCL)-6 in DLBCL. Our results show for the first time that BAL1 represses the anti-proliferative and pro-apoptotic IFNγ-STAT1-IRF1-p53 axis and mediates proliferation, survival and chemo-resistance in DLBCL. As a consequence constitutive IFNγ-STAT1 signaling does not lead to apoptosis but rather to chemo-resistance in DLBCL overexpressing BAL1. Our results suggest that BAL1 may induce an switch in STAT1 from a tumor suppressor to an oncogene in high-risk DLBCL.


Subject(s)
Apoptosis , Cell Proliferation , Interferon Regulatory Factor-1/metabolism , Interferon-gamma/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Neoplasm Proteins/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-2/biosynthesis , Interferon Regulatory Factor-2/genetics , Interferon-gamma/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Neoplasm Proteins/genetics , Poly(ADP-ribose) Polymerases , Protein Isoforms/genetics , Protein Isoforms/metabolism , STAT1 Transcription Factor/genetics , Tumor Suppressor Protein p53/genetics
4.
Tumour Biol ; 33(1): 247-55, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22119988

ABSTRACT

Pancreatic cancer is one of the most malignant diseases in the world. Interferon regulator factor 2 (IRF-2), an interferon regulatory factor, has been known to act as an oncogene in distinct types of cancer. In this study, we found that the expression of IRF-2 was up-regulated in primary pancreatic cancer samples and associated with tumor size, differentiation, tumor-node-metastasis stage, and survival of the patients. In pancreatic cancer cells, knockdown on the expression of IRF-2 inhibited cell growth in the liquid culture and on the soft agar. Mechanistically, IRF-2 modulated the growth of pancreatic cancer cells through regulating proliferation and apoptosis effectors, such as cyclin D1 and BAX. Collectively, these results suggest that IRF-2 plays an important role in the tumorigenesis of pancreatic cancer and down-regulation of IRF-2 would be a new treatment target for pancreatic cancer.


Subject(s)
Cell Proliferation , Cell Transformation, Neoplastic/pathology , Interferon Regulatory Factor-2/biosynthesis , Pancreas/pathology , Pancreatic Neoplasms/pathology , Adult , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Female , Gene Knockdown Techniques , Humans , Interferon Regulatory Factor-2/genetics , Male , Middle Aged , Pancreas/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Up-Regulation
5.
Cancer Res ; 68(4): 1136-43, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18281489

ABSTRACT

IFN-gamma is an antitumor cytokine that inhibits cell proliferation and induces apoptosis after engagement with the IFN-gamma receptors (IFNGR) expressed on target cells, whereas IFN regulatory factor 2 (IRF-2) is able to block the effects of IFN-gamma by repressing transcription of IFN-gamma-induced genes. Thus far, few studies have explored the influences of IFN-gamma on human esophageal cancer cells. In the present study, therefore, we investigated in detail the functions of IFN-gamma in esophageal cancer cells. The results in clinical samples of human esophageal cancers showed that the level of IFN-gamma was increased in tumor tissues and positively correlated with tumor progression and IRF-2 expression, whereas the level of IFNGR1 was decreased and negatively correlated with tumor progression and IRF-2 expression. Consistently, in vitro experiments showed that low concentration of IFN-gamma induced the expression of IRF-2 with potential promotion of cell growth, and moreover, IRF-2 was able to suppress IFNGR1 transcription in human esophageal cancer cells by binding a specific motif in IFNGR1 promoter, which lowered the sensitivity of esophageal cancer cells to IFN-gamma. Taken together, our results disclosed a new IRF-2-mediated inhibitory mechanism for IFN-gamma-induced pathway in esophageal cancer cells: IFN-gamma induced IRF-2 up-regulation, then up-regulated IRF-2 decreased endogenous IFNGR1 level, and finally, the loss of IFNGR1 turned to enhance the resistance of esophageal cancer cells to IFN-gamma. Accordingly, the results implied that IRF-2 might act as a mediator for the functions of IFN-gamma and IFNGR1 in human esophageal cancers.


Subject(s)
Esophageal Neoplasms/metabolism , Interferon Regulatory Factor-2/metabolism , Interferon-gamma/metabolism , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Feedback , Gene Expression , Humans , Interferon Regulatory Factor-2/biosynthesis , Interferon Regulatory Factor-2/genetics , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Receptors, Interferon/antagonists & inhibitors , Receptors, Interferon/biosynthesis , Receptors, Interferon/genetics , Transcription, Genetic , Up-Regulation , Interferon gamma Receptor
6.
Cancer Res ; 67(6): 2535-43, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17363571

ABSTRACT

IFN regulatory factor (IRF)-1 and IRF-2 are generally regarded as a tumor suppressor and an oncoprotein, respectively. However, little is known about their expression and function in esophageal squamous cell carcinomas (ESCC). In our present work, IRF-1 expression was decreased and IRF-2 expression was increased in ESCCs compared with matched normal esophageal tissues. Moreover, statistical data indicated that IRF-2 expression was tightly correlated with progression of ESCCs. As expected, overexpression of either IRF-1 or IRF-2 in an ESCC cell line resulted in either suppression or enhancement of cell growth, respectively. Also, proliferation- and apoptosis-related molecules (p21(WAF1/CIP1), cyclin-D1, Bcl-2, and histone H4) were regulated by IRF-1 and IRF-2. Additionally, high levels of IRF-2 blocked the function of IRF-1 by preventing the latter from translocating into the nucleus; in contrast, knock down of IRF-2 by small interfering RNA permitted nuclear localization and activity of IRF-1. In vivo assay using nude mice indicated that the tumorigenicity of ESCC cells was enhanced with IRF-2 overexpression but dramatically attenuated after forced expression of IRF-1. In conclusion, IRF-1 and IRF-2 are able to regulate tumorigenicity of ESCC cells as antioncoprotein and oncoprotein, respectively. Relative amounts of IRF-1 to IRF-2 are functionally very important for the development and progression of ESCCs, and reduction of the ratio of IRF-1/IRF-2 may lead to the enhancement of tumorigenicity of ESCC cells. Therefore, levels of IRF-1 and IRF-2 are useful indicators in diagnosis and prognosis for ESCCs, and these molecules are potential drug targets for ESCC therapy.


Subject(s)
Esophageal Neoplasms/metabolism , Interferon Regulatory Factor-1/biosynthesis , Interferon Regulatory Factor-2/biosynthesis , Animals , Apoptosis/physiology , Cell Growth Processes/physiology , Disease Progression , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophagus/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-2/genetics , Interferon Regulatory Factor-2/metabolism , Male , Mice , Mice, Nude , Middle Aged , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transfection , Transplantation, Heterologous
7.
J Immunol ; 178(6): 3602-11, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17339457

ABSTRACT

IFN regulatory factor (IRF)-2(-/-) mice are significantly more resistant to LPS challenge than wild-type littermates, and this was correlated with increased numbers of apoptotic Kupffer cells. To assess the generality of this observation, and to understand the role of IRF-2 in apoptosis, responses of peritoneal macrophages from IRF-2(+/+) and IRF-2(-/-) mice to apoptotic stimuli, including the fungal metabolite, gliotoxin, were compared. IRF-2(-/-) macrophages exhibited a consistently higher incidence of apoptosis that failed to correlate with caspase-3/7 activity. Using microarray gene expression profiling of liver RNA samples derived from IRF-2(+/+) and IRF-2(-/-) mice treated with saline or LPS, we identified >40 genes that were significantly down-regulated in IRF-2(-/-) mice, including Stat3, which has been reported to regulate apoptosis. Compared with IRF-2(+/+) macrophages, STAT3alpha mRNA was up-regulated constitutively or after gliotoxin treatment of IRF-2(-/-) macrophages, whereas STAT3beta mRNA was down-regulated. Phospho-Y705-STAT3, phospho-S727-STAT1, and phospho-p38 protein levels were also significantly higher in IRF-2(-/-) than control macrophages. Activation of the STAT signaling pathway has been shown to elicit expression of CASP1 and apoptosis. IRF-2(-/-) macrophages exhibited increased basal and gliotoxin-induced caspase-1 mRNA expression and enhanced caspase-1 activity. Pharmacologic inhibition of STAT3 and caspase-1 abolished gliotoxin-induced apoptosis in IRF-2(-/-) macrophages. A novel IFN-stimulated response element, identified within the murine promoter of Casp1, was determined to be functional by EMSA and supershift analysis. Collectively, these data support the hypothesis that IRF-2 acts as a transcriptional repressor of Casp1, and that the absence of IRF-2 renders macrophages more sensitive to apoptotic stimuli in a caspase-1-dependent process.


Subject(s)
Apoptosis/immunology , Caspase 3/immunology , Caspase 7/immunology , Interferon Regulatory Factor-2/immunology , Kupffer Cells/immunology , Macrophages, Peritoneal/immunology , Repressor Proteins/immunology , STAT1 Transcription Factor/immunology , STAT3 Transcription Factor/immunology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Caspase 3/biosynthesis , Caspase 7/biosynthesis , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/genetics , Down-Regulation/immunology , Gliotoxin/pharmacology , Immunosuppressive Agents/pharmacology , Interferon Regulatory Factor-2/biosynthesis , Interferon Regulatory Factor-2/deficiency , Kupffer Cells/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/metabolism , Mice , Mice, Knockout , Promoter Regions, Genetic/immunology , Repressor Proteins/biosynthesis , STAT1 Transcription Factor/biosynthesis , STAT3 Transcription Factor/biosynthesis
8.
J Interferon Cytokine Res ; 25(10): 587-94, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16241857

ABSTRACT

Interferon-gamma (IFN-gamma) is a pleiotropic cytokine with potent antitumor effects, both in vitro and in vivo. The antitumor activity of IFN-gamma is mediated in part through IFN regulatory factor-1 (IRF-1) and may be blocked by IRF-2. To test our hypothesis that some tumors escape the antitumor effects of IFN-gamma by cellular changes reflected in IRF-1 and IRF-2 expression, we examined IRF-1 and IRF-2 expression in tissue microarrays (TMA) containing 187 specimens of clinically defined invasive breast carcinoma. TMAs (Cooperative Breast Cancer Tissue Resource [CBCTR], National Cancer Institute [NCI]) were stained and then scored by three evaluators blinded to the patients' clinical status. After final scoring, the CBCTR provided the available clinical data for each patient. Whether sorted by carcinoma type or for all data together, statistical analysis showed a significant positive correlation between IRF-1 and IRF-2 expression (p = 0.01) and a negative correlation between IRF-1 expression and tumor grade (p = 0.005). IRF-1 expression is consistent with its role as a tumor suppressor; high-grade breast carcinomas were less likely to maintain expression of IRF-1, a finding consistent with a role for IRF-1 as a tumor suppressor. Further, tumors maintained expression of IRF-2 if there was coincident expression of IRF-1. These data support a model in which alterations of the expression of intracellular effectors of IFN-gamma signaling may diminish the immune-mediated tumor control mechanisms of IFN-gamma.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma/metabolism , Gene Expression Regulation, Neoplastic , Interferon Regulatory Factor-1/biosynthesis , Interferon Regulatory Factor-2/biosynthesis , Tissue Array Analysis , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Carcinoma/immunology , Carcinoma/pathology , Female , Gene Expression Regulation, Neoplastic/immunology , Humans , Interferon Regulatory Factor-1/immunology , Interferon Regulatory Factor-2/immunology , Interferon-gamma/immunology , Signal Transduction/immunology , Tissue Array Analysis/methods , Tumor Escape/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...