Promoters of the genes encoding the transcription factors regulating the cytokine gene expression in macrophages contain putative binding sites for aryl hydrocarbon receptor

2009 ◽  
Vol 33 (6) ◽  
pp. 465-468 ◽  
Author(s):  
Dagmara P. Furman ◽  
Eugenia A. Oshchepkova ◽  
Dmitry Yu. Oshchepkov ◽  
Marina Yu. Shamanina ◽  
Viatcheslav A. Mordvinov
2002 ◽  
Vol 156 (5) ◽  
pp. 771-774 ◽  
Author(s):  
Valerie Horsley ◽  
Grace K. Pavlath

The nuclear factor of activated T cells (NFAT) proteins are a family of transcription factors whose activation is controlled by calcineurin, a Ca2+-dependent phosphatase. Originally identified in T cells as inducers of cytokine gene expression, NFAT proteins play varied roles in cells outside of the immune system. This review addresses the recent data implicating NFAT in the control of gene expression influencing the development and adaptation of numerous mammalian cell types.


1996 ◽  
Vol 16 (3) ◽  
pp. 1074-1084 ◽  
Author(s):  
M Rincon ◽  
R A Flavell

The ability of thymocytes to express cytokine genes changes during the different stages of thymic development. Although CD4- CD8- thymocytes are able to produce a wide spectrum of cytokines in response to a T-cell receptor (TcR)-independent stimulus, as they approach the double-positive (DP) CD4+ CD8+ stage, they lose the ability to produce cytokine. After the DP stage, thymocytes become single-positive CD4+ or CD8+ thymocytes which reacquire the ability to secrete cytokines. In an attempt to understand the molecular basis of this specific regulatin, we use AP-1-luciferase and newly generated NFAT-luciferase transgenic mice to analyze the transcriptional and DNA-binding activities of these two transcription factors that are involved in the regulation of cytokine gene expression. Here, we show that both AP-1 and NFAT transcriptional activities are not inducible in the majority of DP cells but that during the differentiation of DP cells to the mature single-positive stage, thymocytes regain this inducibility. Subpopulation analysis demonstrates that this inducibility is reacquired at the DP stage before the down-modulation of one of the coreceptors. Indeed AP-1 inducibility, just like the ability to express the interleukin-2 gene, is reacquired during the differentiation of DP TcRlow CD69low heat-stable antigen (HSA)high thymocytes to DP TcRhigh CD69high HSAhigh cells, which is considered to be the consequence of the first signal that initiates positive selection. We therefore propose that the inability of DP thymocytes to induce AP-1 and NFAT activities is one of the causes for the lack of cytokine gene expression at this stage and that this inducibility is reacquired at the latest stage of DP differentiation as a consequence of positive selection. This could be a mechanism to prevent the activation of DP thymocytes before selection has taken place.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3669-3669
Author(s):  
Teruhiko Kozuka ◽  
Alan M. Gewirtz ◽  
Yuji Nakata

Abstract Abstract 3669 Poster Board III-605 The c-myb proto-oncogene encodes a transcription factor, c-Myb, which regulates genes important for early myeloid, B, and T lymphoid cell development. Our group has recently shown that c-Myb binds a conserved site in the GATA-3 promoter, upregulating its expression and forming a complex that plays an important role in human Th2 cell development. We have also observed that silencing c-Myb in primary human peripheral blood CD4+ cells under Th2 cell promoting conditions decreases the expression of the Th2 cytokines, IL-4, IL-5 and IL-13. We wondered if this was a direct effect of c-Myb alone, or was also due to an effect of the c-Myb/GATA-3 complex? The human IL-4, IL-5 and IL-13 genes are located within a 150kb locus that is called the “Th2 cytokine gene” on Chromosome 5. The Th2 cytokine locus has numerous DNase I hypersensitive sites (DHSs) which act as regulatory elements for Th2 cytokine gene expression. Among these DHSs, the conserved GATA-3 response element (CGRE), the conserved noncoding sequence 1 (CNS-1), the intronic enhancer (IE) and the hypersensitive site V (HSV) are all known to be activated by GATA-3 directly. Interestingly, sequence analysis identified c-Myb binding sites in close proximity to GATA binding sites within all 4 DHSs. To determine if c-Myb, and GATA-3, occupied these putative binding sites, we performed ChIP assays and found that anti-c-Myb antibody, for example, enriched the CGRE locus 30-fold compared to control IgG. Similar experiments revealed that both c-Myb and GATA-3 bound to all 4 of the elements in human naïve CD4+ T-cells under Th2 promoting conditions. To determine the role of c-Myb and GATA-3 in Th2 cytokine gene expression, we performed reporter assays in 293T, Jurkat and human CD4+ T-cells using IL-4, IL-13 and IL-5 promoter constructs with, or without, the 4 DHSs. c-Myb weakly (2-2.5-fold), but significantly, activated both the IL-4 and IL-5 promoters in 293T cells in the absence of DHSs. In contrast, GATA-3 did not activate the IL-4 and IL-5 promoters without the enhancer elements, though it weakly activated the IL-4 promoter through CNS-1. GATA-3 activated the minimal IL-13 promoter 5-fold, and the addition of c-Myb increased promoter activation 10-fold. In marked contrast to these results, an IL-13 promoter construct containing the CGRE enhancer was activated 65-fold by GATA-3 alone, and 160-fold by the addition of c-Myb. The CGRE locus has only one canonical Myb binding site, but Myb's strong enhancer effect on IL-13 expression was completely lost when the CGRE element's Myb binding site was mutated. Mutating the GATA binding sites decreased IL-13 promoter activity by ∼50%. These results suggest that c-Myb and GATA-3 regulate IL-13 expression directly, and in a cooperative manner. These results lead us to hypothesize that the binding of a c-Myb and GATA-3 complex to the CGRE element changes the chromatin structure of the IL-13 locus which acts as a switch to promote IL-13 expression. Though the most dramatic effects of c-Myb were on IL-13 expression, because c-Myb binds to the other three enhancers, and activates both the IL-4 and IL-5 promoters as well, we hypothesize that c-Myb directly contributes to all Th2 cytokine gene expressions, including IL-13. Finally, a ChIP assay showed that MLL also bound to the locus of CGRE. Since we have already shown that c-Myb can bind to MLL through Menin those results suggest, in aggregate, the possibility of histone modification in Th2 cytokine locus by MLL, GATA-3 and c-Myb. This possibility is under active investigation in our laboratory. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document