17 research outputs found

    Absence of the transcription factor CCAAT enhancer binding protein Ī± results in loss of myeloid identity in bcr/abl-induced malignancy

    No full text
    The lineage-determining transcription factor CCAAT enhancer binding protein Ī± (C/EBPĪ±) is required for myeloid differentiation. Decreased function or expression of C/EBPĪ± is often found in human acute myeloid leukemia. However, the precise impact of C/EBPĪ± 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/EBPĪ±(pos) fetal liver cells led to a chronic myeloid leukemia-like disease. Surprisingly, bcr/abl-expressing C/EBPĪ±(āˆ’/āˆ’) fetal liver cells failed to induce a myeloid disease in transplanted mice, but caused a fatal, transplantable erythroleukemia instead. Accordingly, increased expression of the transcription factors SCL and GATA-1 in hematopoietic precursor cells of C/EBPĪ±(āˆ’/āˆ’) 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/EBPĪ± and GATA-1 was inversely correlated. Id1, an inhibitor of erythroid differentiation, was identified as a critical direct target of C/EBPĪ±. Down-regulation of Id1 by RNA interference impaired C/EBPĪ±-induced granulocytic differentiation. Taken together, our study provides evidence that myeloid lineage identity of malignant hematopoietic progenitor cells requires the residual expression of C/EBPĪ±

    Contingent gene regulatory networks and B cell fate specification

    No full text
    The B cell developmental pathway represents a leading system for the analysis of regulatory circuits that orchestrate cell fate specification and commitment. Considerable progress has been achieved within the past decade in the identification and genetic analysis of various regulatory components. These components include the transcription factors PU.1, Ikaros, Bcl11a, E2A, EBF, and Pax-5, as well as the cytokine receptors Flk2 and IL-7R. Experimental evidence of connectivity among the regulatory components is used to assemble sequentially acting and contingent gene regulatory networks. Transient signaling inputs, self-sustaining positive feedback loops, and crossantagonism among alternate cell fate determinants are key features of the proposed networks that instruct the development of B lymphocyte precursors from hematopoietic stem cells

    CpG methylation in exon 1 of transcription factor 4 increases with age in normal gastric mucosa and is associated with gene silencing in intestinal-type gastric cancers

    No full text
    Transcriptional factor 4 (TCF4), encoding a basic helix-loop-helix transcriptional factor, has recently been demonstrated as a causative gene for Pitt-Hopkins syndrome, a neurodevelopmental disease. Examination of gastric cancers using the restriction landmark genomic scanning technique revealed methylation at a NotI enzyme site in TCF4 intron 8 and further identified CpG dinucleotide hypermethylation in TCF4 exon 1, strongly associated with gene silencing in gastric cancer cell lines. Treatment with 5-aza-2ā€²-deoxycytidine and/or trichostatin A restored TCF4 expression in TCF4-silenced gastric cancer cell lines. Real-time reverse transcriptionā€“polymerase chain reaction analysis of 77 paired primary gastric tumor samples revealed that 38% of analyzed tumors had a >2-fold decrease in TCF4 expression compared with adjacent normal-appearing tissue, and the decrease significantly correlated with increased CpG methylation in TCF4 exon 1. Clinicopathologic data showed that decreased TCF4 expression occurred significantly more frequently in intestinal-type (22/37, 59%) than in diffuse-type (7/37, 19%) gastric cancers (P = 0.0004) and likewise more frequently in early (12/18, 67%) than in advanced (17/59, 29%) gastric cancers (P = 0.004). CpG methylation markedly increased with patient age among normal-appearing tissues, suggesting that CpG methylation in gastric mucosa may be one of the earliest events in carcinogenesis of intestinal-type gastric cancers. Furthermore, ectopic expression of TCF4 decreased cell growth in a gastric cancer cell line, and the knock down of TCF4 using small interfering RNA increased cell migration. Based on these results, we propose that the observed frequent epigenetic-mediated TCF4 silencing plays a role in tumor formation and progression
    corecore