scholarly journals Downregulation of c-Jun Expression by Transcription Factor C/EBPα Is Critical for Granulocytic Lineage Commitment

2002 ◽  
Vol 22 (24) ◽  
pp. 8681-8694 ◽  
Author(s):  
Janki Rangatia ◽  
Rajani Kanth Vangala ◽  
Nicolai Treiber ◽  
Pu Zhang ◽  
Hanna Radomska ◽  
...  

ABSTRACT The transcription factor C/EBPα is crucial for the differentiation of granulocytes. Conditional expression of C/EBPα triggers neutrophilic differentiation, and C/EBPα can block 12-O-tetradecanoylphorbol-13-acetate-induced monocytic differentiation of bipotential myeloid cells. In C/EBPα knockout mice, no mature granulocytes are present. A dramatic increase of c-Jun mRNA in C/EBPα knockout mouse fetal liver was observed. c-Jun, a component of the AP-1 transcription factor complex and a coactivator of the transcription factor PU.1, is important for monocytic differentiation. Here we report that C/EBPα downregulates c-Jun expression to drive granulocytic differentiation. An ectopic increase of C/EBPα expression decreases the c-Jun mRNA level, and the human c-Jun promoter activity is downregulated eightfold in the presence of C/EBPα. C/EBPα and c-Jun interact through their leucine zipper domains, and this interaction prevents c-Jun from binding to DNA. This results in downregulation of c-Jun's capacity to autoregulate its own promoter through the proximal AP-1 site. Overexpression of c-Jun prevents C/EBPα-induced granulocytic differentiation. Thus, we propose a model in which C/EBPα needs to downregulate c-Jun expression and transactivation capacity for promoting granulocytic differentiation.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 646-646
Author(s):  
Katharina Wagner ◽  
Pu Zhang ◽  
Frank Rosenbauer ◽  
Bettina Drescher ◽  
Susumu Kobayashi ◽  
...  

Abstract The lineage-determining transcription factor C/EBPα is required for myeloid differentiation. Decreased function or expression of C/EBPα is often found in human acute myeloid leukemia. However, the precise impact of C/EBPα deficiency on the maturation arrest in leukemogenesis is not well understood. To address this question, we used a murine transplantation model of a bcr/abl induced myeloproliferative disease. The expression of bcr/abl in C/EBPα+/+ and C/EBPα+/− fetal liver cells lead to a chronic myeloid leukemia-like disease. Surprisingly, bcr/abl expressing C/EBPα−/− fetal liver cells fail to induce a myeloid disease in transplanted mice, but instead cause a fatal, transplantable erythroleukemia. Accordingly, increased expression of SCL and GATA-1 in hematopoietic precursor cells of C/EBPα−/− fetal livers was found. The mechanism for the lineage shift from myeloid to erythroid leukemia was studied in a bcr/abl positive cell line. Consistent with findings of the transplant model, expression of C/EBPα and GATA-1 was inversely correlated. Id1, an inhibitor of erythroid differentiation, was upregulated upon C/EBPα expression. Chromatin immunoprecipitation was done and C/EBPα binding to a 3 prime enhancer of the Id1 gene was observed. Downregulation of Id1 by RNA interference impaired C/EBPα induced granulocytic differentiation. Thus, Id1 is a direct and critical target of C/EBPα. Taken together, our study provides the first evidence that myeloid lineage identity of malignant hematopoietic progenitor cells requires the residual expression of C/EBPα.


1993 ◽  
Vol 294 (1) ◽  
pp. 137-144 ◽  
Author(s):  
F Mollinedo ◽  
C Gajate ◽  
A Tugores ◽  
I Flores ◽  
J R Naranjo

Commitment of HL-60 cells to macrophage or granulocytic differentiation was achieved by incubation with 4 beta-phorbol 12-myristate 13-acetate (PMA) for 30-60 min or with dimethyl sulphoxide (DMSO) for 24 h respectively. The commitment stage towards PMA-induced macrophage differentiation was associated with increases in jun B and c-fos mRNA levels, as well as with an increase in the binding activity of transcription factor AP-1. Nevertheless, gel retardation analysis indicated that the AP-1 activity detected in untreated cells was drastically reduced during the commitment stage of DMSO-induced HL-60 differentiation towards granulocytes. When HL-60 cells were treated with sodium butyrate, which induced monocytic differentiation, a remarkable increase in AP-1 binding activity was detected. Treatment of HL-60 cells with 1 alpha,25-dihydroxyvitamin D3, another monocytic differentiation agent, induced a weak, but appreciable, increase in AP-1 activity. Furthermore, addition of sodium butyrate or 1 alpha,25-dihydroxyvitamin D3 to HL-60 cells induced the expression of c-fos, c-jun, jun B and jun D proto-oncogenes. In contrast, when HL-60 cells were treated with retinoic acid, a granulocytic differentiation inducer, no enhanced AP-1 binding activity was observed, and only a weak increase in jun D mRNA level was detected. These data indicate that formation of AP-1 is not required for the induction of HL-60 differentiation towards granulocytes, whereas induction of monocytic differentiation is correlated with an increase in AP-1 activity. The differential expression of AP-1 activity may be critical in the differentiation of HL-60 cells towards monocytic or granulocytic lineages.


2006 ◽  
Vol 26 (3) ◽  
pp. 1109-1123 ◽  
Author(s):  
Daniela S. Bassères ◽  
Elena Levantini ◽  
Hongbin Ji ◽  
Stefano Monti ◽  
Shannon Elf ◽  
...  

ABSTRACT The leucine zipper family transcription factor CCAAT enhancer binding protein alpha (C/EBPα) inhibits proliferation and promotes differentiation in various cell types. In this study, we show, using a lung-specific conditional mouse model of C/EBPα deletion, that loss of C/EBPα in the respiratory epithelium leads to respiratory failure at birth due to an arrest in the type II alveolar cell differentiation program. This differentiation arrest results in the lack of type I alveolar cells and differentiated surfactant-secreting type II alveolar cells. In addition to showing a block in type II cell differentiation, the neonatal lungs display increased numbers of proliferating cells and decreased numbers of apoptotic cells, leading to epithelial expansion and loss of airspace. Consistent with the phenotype observed, genes associated with alveolar maturation, survival, and proliferation were differentially expressed. Taken together, these results identify C/EBPα as a master regulator of airway epithelial maturation and suggest that the loss of C/EBPα could also be an important event in the multistep process of lung tumorigenesis. Furthermore, this study indicates that exploring the C/EBPα pathway might have therapeutic benefits for patients with respiratory distress syndromes.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2401-2401
Author(s):  
Jesus Duque-Afonso ◽  
Leticia Solari ◽  
Michael Luebbert

Abstract LAT2 (NTAL/LAB/WBSCR5) is a 28 KDa membrane protein which acts as adaptor molecule in the signalling pathways of FcεR I, c-Kit, B cell and T cell receptor. Bone marrow-derived mast cells from knock-out (KO) mice are hyperresponsive to stimulation via FcεR I. Although LAT2 is highly expressed in B cells, no major changes were found in function or development of B cells from LAT2 KO mice. An autoimmunity syndrome in LAT2 KO mice is caused, at least in part, by hyperreactivity and higher proliferation of T cells. Previously, we showed that LAT2 mRNA is repressed in vivo by AML1/ETO which was confirmed by others in several large series of primary AML blasts. We wished to elucidate the possible role of LAT2 during the myelopoiesis. AML1/ETO was induced by Ponasterone A in an ecdysone-inducible system in U937 cells (9/14/18 cell line). AML bone marrow samples from 43 patients (pts) were analyzed for LAT2 expression. Several myeloid cell lines were treated either with ATRA, DMSO or PMA for 3 days. Normal CD34+ cells were differentiated ex vivo by G-CSF towards granulocytes and by GM-CSF plus IL-4 towards monocytes and dendritic cells. LAT2 expression was determined by Northern and Western blot. LAT2 protein was repressed not only in AML1/ETO positive primary AML blasts (6/6), but also in blasts from patients with deletions of chromosome 7 (3/4) and the t(15;17) (4/4); expression was moderate to high in AML blasts with normal karyotype (14/15). LAT2 was expressed in normal monocytes and even higher in alveolar macrophages but not in granulocytes of healthy donors. It was downregulated after ATRA-induced granulocytic differentiation of NB4, HL60 and U937 cells but upregulated after DMSO-induced granulocytic differentiation of HL60 cells and PMA-induced monocytic-macrophage differentiation of HL60, U937 and Kasumi-1 cells. In normal CD34+ cells, LAT2 was strongly induced 7 days after the addition of G-CSF and GM-CSF+IL4 respectively, but after 14 days it was downregulated (0.7 +/− 0.4-fold) by G-CSF-induced granulocytic differentiation and upregulated (5.8 +/− 2.8-fold) by GM-CSF+IL4-induced monocytic-DC differentiation. Conditional expression of AML1/ETO in 9/14/18-U937 cells partially inhibited the PMA- and vitamin D3-induced monocytic differentiation of these cells, as determined by FACS for CD11b and CD11c. In conclusion, LAT2 protein is strongly repressed by AML1/ETO in primary leukemias and is upregulated during the monocytic differentiation in several cell lines and normal CD34+ cells. Further studies in a LAT2 knock-down by shRNAs in U937 cells are warranted to functionally address its possible role in monocytic differentiation.


Blood ◽  
1999 ◽  
Vol 94 (9) ◽  
pp. 3141-3150 ◽  
Author(s):  
Walter Verbeek ◽  
Julie Lekstrom-Himes ◽  
Dorothy J. Park ◽  
Pham My-Chan Dang ◽  
Peter T. Vuong ◽  
...  

Abstract Targeted mutation of the myeloid transcription factor C/EBPɛ in mice results in gram-negative septic death at 3 to 5 months of age. This study defines the underlying molecular defects in their terminal granulocytic differentiation. The mRNA for the precursor protein of the cathelin-related antimicrobial peptides was almost completely absent in the bone marrow cells of C/EBPɛ−/− mice. This finding may help explain their susceptibility to gram-negative sepsis, because both are bacteriocidal peptides with potent activity against gram-negative bacteria. Superoxide production was found to be reduced in both granulocytes and monocytes of C/EBPɛ−/− mice. While gp91 phox protein levels were normal, p47phox protein levels were considerably reduced in C/EBPɛ −/− granulocytes/monocytes, possibly limiting the assembly of the NADPH oxidase. In addition, expression of mRNA of the secondary and tertiary granule proteins, lactoferrin and gelatinase, were not detected, and levels of neutrophil collagenase mRNA were reduced in bone marrow cells of the knock-out mice. The murine lactoferrin promoter has a putative C/EBP site close to the transcription start site. C/EBPɛ bound to this site in electromobility shift assay studies and mutation of this site abrogated binding to it. A mutation in the C/EBP site reduced the activity of the promoter by 35%. Furthermore, overexpression of C/EBPɛ in U937 cells increased the activity of the wild-type lactoferrin promoter by 3-fold. In summary, our data implicate C/EBPɛ as a critical factor of host antimicrobial defense and suggests that it has a direct role as a positive regulator of expression of lactoferrin in vivo.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3651-3651
Author(s):  
Boris Bartholdy ◽  
Yukiya Yamamoto ◽  
Erica Evans ◽  
John Crispino ◽  
Daniel G. Tenen

Abstract Abstract 3651 Poster Board III-587 The Ets transcription factor PU.1 is a master regulator absolutely required for the differentiation of monocytes, macrophages, and B cells in the fetal liver and in the adult bone marrow. PU.1 drives hematopoietic differentiation partly through direct protein-protein interactions with other transcription factors, such as the AP-1 transcription factor c-Jun. We have shown that c-Jun can be recruited to promoters which do not include AP-1 binding sites, such as the MCSFR promoter, and act as a PU.1-dependent co-activator. To address the functional importance of this interaction, we identified and studied PU.1 point mutants that lost the capability to physically interact with c-Jun while retaining normal DNA binding affinity. These mutants failed to efficiently transactivate a PU.1 target reporter, and, more importantly, were unable to induce monocyte/macrophage differentiation of the PU.1-deficient immature myeloid 503 cell line. Subsequently, we have generated knock-in mouse models harboring these single point mutations by means of homologous recombination. The mutant mice phenotypically resemble PU.1-deficient mice, have an early block in hematopoiesis, and die perinatally. We show that the mutant PU.1 mRNA and protein is expressed in long-term and short-term hematopoietic stem cells, but that the maturation into lymphoid primed multipotent progenitor (LMPP) and later progenitor populations is severely blocked, leading to an almost complete loss of mature B, T and myeloid cells. Collectively, our data strongly suggest that the PU.1-c-Jun interaction is crucial for normal PU.1 function in vivo during murine fetal hematopoiesis. Disclosures: No relevant conflicts of interest to declare.


2007 ◽  
Vol 282 (38) ◽  
pp. 27685-27692 ◽  
Author(s):  
Inga Waldmann ◽  
Sarah Wälde ◽  
Ralph H. Kehlenbach

c-Jun and c-Fos are major components of the transcriptional complex AP-1. Here, we investigate the nuclear import pathway(s) of the transcription factor c-Jun. c-Jun bound specifically to the nuclear import receptors importin β, transportin, importin 5, importin 7, importin 9, and importin 13. In digitonin-permeabilized cells, importin β, transportin, importin 7, and importin 9 promoted efficient import of c-Jun into the nucleus. Importin α, by contrast, inhibited nuclear import of c-Jun in vitro. A single basic region preceding the leucine zipper of c-Jun functions as a nuclear localization signal (NLS) and was required for interaction with all tested import receptors. In vivo, nuclear import of a c-Jun reporter protein lacking the leucine zipper strictly depended on this NLS. In a leucine zipper-dependent manner, c-Jun with mutations in its NLS was still imported into the nucleus in a complex with endogenous leucine zipper proteins or, for example, with cotransfected c-Fos. Together, these results explain the highly efficient nuclear import of the transcription factor c-Jun.


2002 ◽  
Vol 169 (10) ◽  
pp. 5715-5725 ◽  
Author(s):  
Shanjin Cao ◽  
Jianguo Liu ◽  
Marta Chesi ◽  
Peter Leif Bergsagel ◽  
I-Cheng Ho ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2606-2606
Author(s):  
SunHwa Hong ◽  
Alan D. Friedman

Abstract Abstract 2606 C/EBP and AP-1 proteins represent two sub-families of the basic region-leucine zipper (BR-LZ or bZIP) family of transcription factors. C/EBPs, including C/EBPα, C/EBPβ, C/EBPδ, and C/EBPε, homodimerize or heterodimerize via their leucine zipper domains to bind DNA at palindromic DNA element 5'-ATTGCGCAAT or related sequences. The AP-1 family consists of several Jun and Fos proteins which heterodimerize to bind the palindromic DNA element 5'-TGACGTCA. We previously utilized directed dimerization via artificial acidic and basic zippers to show that C/EBPα:AP-1 heterodimers bind novel DNA elements and stimulate monocytic differentiation upon transduction into murine marrow cells (Cai et al 2008). We now demonstrate that intact C/EBPα and c-Jun have the capacity to heterodimerize and preferentially bind a hybrid DNA site, 5'-TGACGCAAT. Using in vitro translated, myc-tagged proteins to control input ratios based on myc Western blotting, a 2:1 ratio of c-Jun:C/EBPα was found to be sufficient for preferential binding to the hybrid element in a gel shift/super-shift assay. As a control, we demonstrate that C/EBPαGZ, containing the GCN4 leucine zipper in place of the C/EBPα zipper, does not heterodimerize with excess c-Jun to bind the hybrid element. We next sought to determine whether a 2:1 AP-1:C/EBP ratio might be achievable by proteins expressed endogenously in myeloid cells. The relative affinities of several C/EBP or AP-1 antisera were determined using myc-tagged proteins whose expression ratios could be normalized using myc antisera. Semi-quantitative Western blot analysis of 32Dcl3 or HF-1 cells undergoing granulocytic differentiation in response to G-CSF or of HL-60 or M1 cells undergoing monocytic differentiation in response to phorbol ester or IL-6, respectively, indicated that the summation of c-Jun, JunB, and c-Fos levels approximately equals the sum of C/EBPα and C/EBPβ, demonstrating the feasibility of endogenous C/EBP:AP-1 heterodimer formation. Myeloid nuclear extracts were then evaluated for C/EBP, C/EBP:AP-1, and AP-1 complexes using biotinylated oligonucleotides. Myeloid cell extracts were incubated with biotinylated DNA probes containing C/EBPα, AP-1, or hybrid cis elements, pulled down using streptavidin beads, washed, eluted, and subjected to Western blotting for bound proteins. Induction of AP-1 proteins during monopoiesis favored binding of C/EBP:AP-1 heterodimers, whereas C/EBPα homodimers were more evident during granulocytic differentiation. Global analysis of genes bound and activated by C/EBP:AP-1 complexes is in progress, and we demonstrate that the C/EBPα:c-Jun heterodimer binds the element 5'-TAGCGCAAG in the PU.1 promoter with increased affinity compared with C/EBPα homodimers. In summary, C/EBP:AP-1 leucine zipper complexes readily form when in vitro translated AP-1 proteins are in modest excess to C/EBPα, endogenous AP-1 proteins are expressed at concentrations similar to endogenous C/EBP proteins making heterodimerization feasible, C/EBP:AP-1 complexes are detected in myeloid nuclear extracts, and increased expression of Jun/Fos proteins in response to M-CSF or other external cues is predicted to reduce C/EBP homodimer and increase C/EBP:AP-1 heterodimer formation to favor monopoiesis over granulopoiesis via activation of genes harboring hybrid C/EBP:AP-1 cis elements. Formation of C/EBP:AP-1 complexes is likely also relevant to additional cell lineages and biologic processes. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3617-3617
Author(s):  
Wen-yue Zhuang ◽  
Zi-Xing Chen

Abstract Abstract 3617 The t(8; 21) is one of the most frequent chromosomal translocations associated with acute leukemia. The AML1(RUNX1)-ETO(MTG8) fusion transcription factor generated by the t (8; 21) translocation is believed to deregulate the expression of genes that are crucial for normal differentiation and proliferation of hematopoietic progenitors, resulting in acute myelogenous leukemia by recruiting co-repressor complexes to DNA. To investigate the role of AML1-ETO in leukemogenesis, we transfected the cloned AML1-ETO cDNA with plasmid and expressed the AML1-ETO protein in U937 myelomonocytic leukemia cells. Interestingly, we found dichotomous phenomena in these transfected leukemic cells, i.e. growth arrest versus differentiation block (detailed data?). The AML1-ETO transfected U937cells were growing significantly slower than that of empty vector-transfected cells and nontransfected cells (P<0.01). As the surface markers for myeloid differentiation, the expression of CD11b and CD14 measured by flow cytometry demonstrated that the percentage of CD11b+ cell was 4.1%-7.0% in U937-A/E1-4 cells, which was significantly lower(P<0.01) than those in U937-Mock cells (11.4%) and U937-WT cells (11.0%). Moreover, the expression of CD14 antigen was decreased by 1.5–2-fold as compared with the control cells. By focusing on the anti-apoptotic gene (Bcl-2), a key transcription factor (C/EBPA) which regulates granulocytic differentiation and a tumor suppressor gene (p14ARF), we found that AML1-ETO- expressing cell subclones displayed low levels of these three genes in comparison with the non-transfected U937 (P<0.001). In primary bone marrow cells of acute myeloid leukemia containing t(8;21)/AML1-ETO, levels of Bcl-2, C/EBPA and p14ARF mRNA were markedly lower (P<0.001) when compared with other acute myeloid leukemias lacking this translocation (n=10). Chromatin immunoprecipitation assays (ChIP) demonstrated that Bcl-2, C/EBPA and p14ARF were among the direct transcriptional regulating targets of AML1-ETO. The universal binding of AML1-ETO to genomic DNA resulted in MeCP2 recruitment (P<0.01), reduction of histone H3 (P<0.0 1) or histone H4(P<0.01) acetylation and increased tri-methylation on histone H3 lysine 9 (P<0.01)as well as histone H3 lysine 27 (P<0.01), indicating that AML1-ETO induced heterochromatic silencing of Bcl-2, C/EBPA and p14ARF. These results suggested that the aberrant transcription factor AML1-ETO epigenetically silences the function of Bcl-2, C/EBPA and p14ARF gene by inducing repressed chromatin configurations at their promoters through histone modification. Considering that apoptosis-enhancing effect of AML1-ETO would not be favorable to the leukemogenesis, it must be compensated by some other effects to permit its leukemogenic potential. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document