Autocrine GM-CSF transcription in the leukemic progenitor cell line KG1a is mediated by the transcription factor ETS1 and is negatively regulated through SECTM1 mediated ligation of CD7

2014 ◽  
Vol 1840 (3) ◽  
pp. 1004-1013 ◽  
Author(s):  
Christina Bade-Döding ◽  
Wiebke Göttmann ◽  
Anja Baigger ◽  
Matthew Farren ◽  
Kelvin P. Lee ◽  
...  
Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 721-721
Author(s):  
Charles G. Mullighan ◽  
Christopher B. Miller ◽  
Letha A. Phillips ◽  
James Dalton ◽  
Jing Ma ◽  
...  

Abstract Using high-resolution SNP arrays and genomic resequencing, we recently reported deletions, translocations, and mutations involving regulators of B cell development in 40% of pediatric B-progenitor ALL (Nature2007;446:758). The most frequently involved genes were PAX5, EBF1, IKZF1 (Ikaros), IKZF3 (Aiolos) and LEF1. Ikaros is a transcription factor required for normal lymphoid development and acts as a tumor suppressor in mice. Focal deletions of IKZF1 were observed in 7 B-progenitor ALL cases, suggesting that the previously reported expression of dominant-negative, non-DNA binding Ikaros isoforms may be caused by genomic IKZF1 abnormalities. We have now extended the analysis to 283 pediatric ALL cases, including B-progenitor ALL with high hyperdiploidy (N=39), hypodiploidy (N=10), rearrangement of MLL (N=23), TCF3-PBX1 (N=17), ETV6-RUNX1 (N=48) and BCR-ABL1 (N=21), as well as cases with low hyperdiploid, normal or miscellaneous cytogenetics (N=75), and T-lineage ALL (N=50). We also examined 22 adult BCR-ABL1 positive ALL cases, 37 acute leukemia cell lines and 49 samples from 23 chronic myeloid leukemia (CML) cases at various stages of disease, including 15 with matched blast crisis samples (12 myeloid, 3 lymphoid). All samples were examined with 500,000 feature Affymetrix SNP arrays (250k Sty and Nsp). The 50k Hind and Xba arrays were also used in 252 ALL cases. Sixty-two (20.3%) ALL cases harboured IKZF1 deletions, including 36 of 43 (83.7%) BCR-ABL1 positive B-ALL cases (76.2% of 21 childhood cases, and 90.9% of 22 adult cases). The deletions were limited to IKZF1 in 25 BCR-ABL1 ALL cases, and in 19 cases deleted an internal subset of IKZF1 exons, most commonly 3–6 (Δ3–6). Remarkably, chronic phase CML samples lacked evidence of IKZF1 deletion, whereas four of 15 matched CML blast crisis samples (66% of lymphoid and 17% of myeloid) had acquired an IKZF1 deletion. The IKZF1 Δ3–6 deletion was also detected in the BCR-ABL1 B-progenitor cell lines BV173, OP1 and SUP-B15, the Δ1–6 deletion in the MYC-IGH/BCL2-IGH B-progenitor cell line 380, and Δ1–7 in the BCR-ABL1 B-progenitor cell line TOM-1. RT-PCR analysis for IKZF1 transcripts demonstrated complete concordance between the extent of IKZF1 deletion and the expression of aberrant Ikaros transcripts lacking internal exons. Importantly, on quantitative RT-PCR analysis and western blotting, expression of the dominant-negative Ik6 transcript and protein, which lacks exons 3–6, was exclusively observed in those cases with IKZF1 Δ3–6, demonstrating that the Ik6 transcript is the result of a specific genetic lesions and not alternative splicing of wild-type IKZF1. Lastly, sequence analysis of the IKZF1 Δ3–6 breakpoints indicated that the deletions arise from aberrant activity of RAG-mediated V(D)J recombination. Taken together, these data demonstrate that deletion of IKZF1, resulting in either haploinsufficiency or the expression of a dominant negative form of the transcription factor, is a central event in the pathogenesis of both pediatric and adult BCR-ABL1 B-progenitor ALL.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2228-2228
Author(s):  
Zhijia Ye ◽  
Sherman M. Weissman

Abstract Hematopoiesis is regulated by a number of growth factors and cytokines, among which stem cell factor (SCF) plays a critical role for normal hematopoiesis. An important feature of SCF is its strong capacity to synergize with the cytokines including interleukin-3(IL-3), granulocyte-marcrophage colony-stimulation factor (GM-CSF) and erythropoietin to regulate proliferation and differentiations of hematopoietic progenitor cells. The mouse hematopoietic progenitor cell line EML contains two populations of lineage negative cells, distinguished by the expression or absence of CD34. The two sub-populations express similar levels of the SCF receptor c-kit, but only the CD34+ population replicates in the presence of SCF alone, while the CD34 negative population replicates in the presence of IL3 but this replication is synergistically stimulated by the addition of SCF. Our previous studies indicated that synergistic proliferation of the mouse hematopoietic progenitor cell line EML induced by SCF in combination with IL-3 was probably mediated by the synergistic increases in tyrosine phosphorylation of c-Kit and β-chain of IL-3 receptor, which is shared with GM-CSF receptor and interleukin-5 receptor. Conversely, the decreased response of c-kit to SCF alone in the CD34 negative population may be a result of the increased expression of IL3 receptor in these cells. Trans-phosphorylation between c-Kit and the β-chain of IL-3 receptor was also observed in EML cells. In studies of the molecular mechanism behind the functional interactions between SCF and IL-3 we found that c-Kit and IL-3 receptor β-chain form a complex in EML cells. Antibody Ab-1 specific against the Ig-like domain 4 in the extracellular region of c-Kit blocked not only cell proliferation induced by SCF but also synergistic proliferation induced by SCF plus IL-3. Consistent with this finding, Ab-1 inhibited phosphorylation of c-Kit induced by SCF or IL-3, and synergistic phosphorylation of c-kit induced by SCF plus IL-3 respectively. SCF and IL-3 in combination synergistically activated the protein kinases Akt and Erks, the downstream mediators of c-Kit and IL-3R. Phosphorylation of Erks is independent on the phosphatidylinositol 3-kinase (PI3-kinase) pathway in EML cells. These data suggested the possibility that the synergistic growth of EML cells induced by SCF in combination with IL-3 was mediated by multiple mechanisms that could include trans- and synergistic phosphorylation of c-Kit and the β-chain of IL-3 receptor, PI3-kinase dependent synergistic phosphorylation of Akt and PI3-kinase independent synergistic phosphorylation of Erks. The trans- and synergistic phosphorylation of c-Kit and β-chain of IL-3 receptor was mediated by physical interaction of two receptors.


Blood ◽  
1999 ◽  
Vol 93 (2) ◽  
pp. 554-563 ◽  
Author(s):  
Christoph Heberlein ◽  
Jutta Friel ◽  
Christine Laker ◽  
Dorothee von Laer ◽  
Ulla Bergholz ◽  
...  

Abstract We show a dramatic downregulation of the stem cell factor (SCF) receptor in different hematopoietic cell lines by murine stroma. Growth of the human erythroid/macrophage progenitor cell line TF-1 is dependent on granulocyte-macrophage colony-stimulating factor (GM-CSF) or interleukin-3 (IL-3). However, TF-1 cells clone and proliferate equally well on stroma. Independent stroma-dependent TF-1 clones (TF-1S) were generated on MS-5 stroma. Growth of TF-1S and TF-1 cells on stroma still requires interaction between c-kit (SCF receptor) and its ligand SCF, because antibodies against c-kit inhibit growth to less than 2%. Surprisingly, c-kit receptor expression (RNA and protein) was downregulated by 2 to 3 orders of magnitude in TF-1S and TF-1 cells grown on stroma. This stroma-dependent regulation of the kit receptor in TF-1 was also observed on exposure to kit ligand-negative stroma, thus indicating the need for heterologous receptor ligand interaction. Removal of stroma induced upregulation by 2 to 4 orders of magnitude. Downregulation and upregulation of c-kit expression could also be shown for the megakaryocytic progenitor cell line M-07e and was comparable to that of TF-1, indicating that stroma-dependent regulation of c-kit is a general mechanism. Downregulation may be an economic way to compensate for the increased sensitivity of the c-kit/ligand interaction on stroma. The stroma-dependent c-kit regulation most likely occurs at the transcriptional level, because mechanisms, such as splicing, attenuation, differential promoter usage, or mRNA stability, could be excluded.


2005 ◽  
Vol 53 (2) ◽  
pp. 104-115 ◽  
Author(s):  
Makoto Horiuchi ◽  
Yasuhiro Tomooka

1988 ◽  
Vol 8 (6) ◽  
pp. 2335-2341
Author(s):  
R J Akhurst ◽  
N B Flavin ◽  
J Worden

A new variant rat myogenic cell line, ts485, was isolated by subcloning the cell line ts3b2 (H. T. Nguyen, R. M. Medford, and B. Nadal-Ginard, Cell 34:281-293, 1983). Unlike the progenitor cell line, ts485 was thermosensitive for differentiation. Experiments with conditioned medium suggested that diffusible extracellular factors were not involved in dictating the differential phenotypes of ts485 cells cultured at the permissive and nonpermissive temperatures. Temperature shift experiments performed on cultures of ts485 cells indicated that the temperature-sensitive lesion was in a factor active during the growth phase and required to trigger a cascade of events leading to terminal differentiation.


Sign in / Sign up

Export Citation Format

Share Document