scholarly journals Fusion with E2A converts the Pbx1 homeodomain protein into a constitutive transcriptional activator in human leukemias carrying the t(1;19) translocation.

1994 ◽  
Vol 14 (6) ◽  
pp. 3938-3948 ◽  
Author(s):  
Q Lu ◽  
D D Wright ◽  
M P Kamps

E2A-PBX1 is a chimeric gene formed by the t(1;19)(q23;p13.3) chromosomal translocation of pediatric pre-B-cell leukemia. The E2A-Pbx1 fusion protein contains sequences encoding the transactivation domain of E2A joined to a majority of the Pbx1 protein, which contains a novel homeodomain. Earlier, we found that expression of E2A-Pbx1 causes malignant transformation of NIH 3T3 fibroblasts and induces myeloid leukemia in mice. Here we demonstrate that the homeodomains encoded by PBX1, as well as by the highly related PBX2 and PBX3 genes, bind the DNA sequence ATCAATCAA. E2A-Pbx1 strongly activates transcription in vivo through this motif, while Pbx1 does not. This finding suggests that E2A-Pbx1 transforms cells by constitutively activating transcription of genes regulated by Pbx1 or by other members of the Pbx protein family.

1994 ◽  
Vol 14 (6) ◽  
pp. 3938-3948
Author(s):  
Q Lu ◽  
D D Wright ◽  
M P Kamps

E2A-PBX1 is a chimeric gene formed by the t(1;19)(q23;p13.3) chromosomal translocation of pediatric pre-B-cell leukemia. The E2A-Pbx1 fusion protein contains sequences encoding the transactivation domain of E2A joined to a majority of the Pbx1 protein, which contains a novel homeodomain. Earlier, we found that expression of E2A-Pbx1 causes malignant transformation of NIH 3T3 fibroblasts and induces myeloid leukemia in mice. Here we demonstrate that the homeodomains encoded by PBX1, as well as by the highly related PBX2 and PBX3 genes, bind the DNA sequence ATCAATCAA. E2A-Pbx1 strongly activates transcription in vivo through this motif, while Pbx1 does not. This finding suggests that E2A-Pbx1 transforms cells by constitutively activating transcription of genes regulated by Pbx1 or by other members of the Pbx protein family.


2020 ◽  
Vol 6 (1) ◽  
pp. eaau5670 ◽  
Author(s):  
K. Hennig ◽  
I. Wang ◽  
P. Moreau ◽  
L. Valon ◽  
S. DeBeco ◽  
...  

Directional cell motility relies on the ability of single cells to establish a front-rear polarity and can occur in the absence of external cues. The initiation of migration has often been attributed to the spontaneous polarization of cytoskeleton components, while the spatiotemporal evolution of cell-substrate interaction forces has yet to be resolved. Here, we establish a one-dimensional microfabricated migration assay that mimics the complex in vivo fibrillar environment while being compatible with high-resolution force measurements, quantitative microscopy, and optogenetics. Quantification of morphometric and mechanical parameters of NIH-3T3 fibroblasts and RPE1 epithelial cells reveals a generic stick-slip behavior initiated by contractility-dependent stochastic detachment of adhesive contacts at one side of the cell, which is sufficient to trigger cell motility in 1D in the absence of pre-established polarity. A theoretical model validates the crucial role of adhesion dynamics, proposing that front-rear polarity can emerge independently of a complex self-polarizing system.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2570-2570
Author(s):  
Ning Wang ◽  
Mei Wan ◽  
Yi Tang ◽  
Hyung G. Kim ◽  
Claudia Cotta ◽  
...  

Abstract Homeobox (Hox) genes play crucial roles in hematopoiesis. Alterations of Hox gene expressions are found in patients with hematological malignancy. Nup98-Hoxa9, a fusion protein combining the N-terminus of Nup98 and the C-terminus of Hoxa9 resulting from t (7; 11) chromosomal translocation, is the first evidence of Hox gene involvement in human leukemia. We have shown that BMP specific R-Smads and Smad4 interact with Hox proteins including Hoxa9, and the interaction blocks binding of Hox proteins to their DNA elements. Here, we show that Smad4, the common signal transducer of both TGF-β and BMP, interacts with the homeodomain of Nup98-Hoxa9 in a coimmunoprecipitation assay. Furthermore, this interaction abolished the DNA binding ability of Nup98-Hoxa9 to DNA in both EMSA using purified GST fusion protein and in in vivo chromatin immunoprecipitation assays. Subsequent mapping data revealed the MH1 domain of Smad4 mediated the interaction. These findings suggest that Smad4 could be used to inhibit Hox-induced leukemia. We then attempted to examine whether Smad4 could inhibit the downstream target gene by Nup98-Hoxa9. Although Nup98-Hoxa9 is believed to function as an aberrant DNA-binding transcriptional activator, downstream genes that contribute to leukemogenesis are largely unknown. It has been reported that Nup98-Hoxa9 induces Hoxa9 expression in murine primary bone marrow cells. Accumulating evidence suggests that Hoxa9 plays important roles in normal and malignant hematopoiesis. Enforced expression of Hoxa9 causes proliferative expansion of primitive hematopoietic stem/progenitor cells and subsequently inhibits their differentiation. Aberrant expression of HOXA9 due to retroviral integration induced acute myeloid leukemia (AML) in mice. By visually scanning the sequence of the Hoxa9 promoter, we found many TGAT and TTA (T/C) consensus sequences, which are preferentially recognized by the heterodimer of PBX protein and the proteins of the Abd-B-like gene family, including Hoxa9, respectively. To test whether Hoxa9 could be a direct transcriptional target for Nup98-Hoxa9, we used a Hoxa9 promoter/luciferase reporter construct along with the expression plasmid pcDNA-Nup98/Hoxa9 to transiently transfect NIH/3T3 fibroblast cells and Ba/F3 murine hematopoietic progenitor cells. Nup98-Hoxa9 showed substantial transcription activation on Hoxa9 promoter in these cell lines. This activation is further enhanced by cotransfection of PBX1a, which stabilizes the binding of Nup98-Hoxa9 to DNA. Nup98-Hoxa9 also bound to radiolabelled DNA oligo from the Hoxa9 promoter in vitro by EMSA assay. These data indicate that Nup98-Hoxa9 directly activates the Hoxa9 promoter. Interestingly, Smad4 removed the binding of Nup98-Hoxa9 to the Hoxa9 promoter. Co-overexpression of Smad4 inhibited the transactivation activity of Nup98-Hoxa9 on the Hoxa9 promoter regardless of the presence of PBX1a. In NIH/3T3 cells, Smad4 inhibited Nup98-Hoxa9-induced Hoxa9 expression both at the mRNA and protein levels. In summary, we show that Smad4 inhibited DNA binding activity of Nup98-Hoxa9 by interacting at Hox homeodomain. This interaction abolished the transcription activity of Nup98-Hoxa9 on its target gene, thus preventing Nup98-Hoxa9 from working as a transcription factor, which could lead to a new therapeutic target for the treatment of myeloid leukemia caused by Nup98-Hoxa9.


2008 ◽  
Vol 130 (6) ◽  
Author(s):  
Troy J. Eggum ◽  
Christopher J. Hunter

Various cell populations have been shown to respond to hydrostatic pressure; however, many of the culture systems suffer from shortcomings in design or methodology. Of particular interest to us is the potential role of pressure and other environmental factors in modulating stem cell behavior in intervertebral disk repair. A system was developed for the growth of cells and tissues under intermittent hydrostatic pressure. The system was validated with NIH 3T3 fibroblasts for sterilizability and cytotoxicity. Further experiments were conducted with canine mesenchymal stem cells under various levels of pressure, oxygen, glucose, and conditioned medium. The culture system showed no cytotoxicity and was able to demonstrate that the proliferation and metabolism of mesenchymal stem cells are sensitive to medium glucose and oxygen concentration and hydrostatic pressure. The cells exposed to hydrostatic pressure differed in their morphology from nonexposed cells. The system is capable of supporting long-term cell culture and examining the role of mechanical and environmental stimulation in vivo.


1999 ◽  
Vol 19 (1) ◽  
pp. 594-601 ◽  
Author(s):  
Paula Y. P. Lam ◽  
Jack E. Sublett ◽  
Andrew D. Hollenbach ◽  
Martine F. Roussel

ABSTRACT The chimeric transcription factor Pax3-FKHR, produced by the t(2;13)(q35;q14) chromosomal translocation in alveolar rhabdomyosarcoma, consists of the two Pax3 DNA binding domains (paired box and homeodomain) fused to the C-terminal forkhead (FKHR) sequences that contain a potent transcriptional activation domain. To determine which of these domains are required for cellular transformation, Pax3, Pax3-FKHR, and selected mutants were retrovirally expressed in NIH 3T3 cells and evaluated for their capacity to promote anchorage-independent cell growth. Mutational analysis revealed that both the third α-helix of the homeodomain and a small region of the FKHR transactivation domain are absolutely required for efficient transformation by the Pax3-FKHR fusion protein. Surprisingly, point mutations in the paired domain that abrogate sequence-specific DNA binding retained transformation potential equivalent to that of the wild-type protein. This finding suggests that DNA binding mediated through the Pax3 paired box is not required for transformation. Our results demonstrate that the integrity of the Pax3 homeodomain recognition helix and the FKHR transactivation domain is necessary for efficient cellular transformation by the Pax3-FKHR fusion protein.


Blood ◽  
2003 ◽  
Vol 102 (9) ◽  
pp. 3363-3370 ◽  
Author(s):  
Claudia S. Huettner ◽  
Steffen Koschmieder ◽  
Hiromi Iwasaki ◽  
Junko Iwasaki-Arai ◽  
Hanna S. Radomska ◽  
...  

AbstractThe BCR/ABL fusion protein is found in more than 90% of patients with chronic myeloid leukemia (CML) as well as in a subset of patients with acute B-cell leukemia. We have previously described a transgenic model for an inducible and reversible acute B-cell leukemia caused by p210 BCR/ABL. Here, we describe a new model of an inducible BCR/ABL disease by directing the expression of the oncogene to megakaryocytic progenitor cells within the murine bone marrow using the tetracycline-responsive expression system under the control of human CD34 regulatory elements. The predominant feature was the development of a chronic thrombocytosis. The condition progressed with the development of splenomegaly accompanied by lymphadenopathy in some mice. Affected animals demonstrated a dramatic increase in the number of megakaryocytes in the bone marrow and the spleen. Immunohistochemistry demonstrated that the reporter gene was expressed in hematopoietic stem cells (HSCs), common myeloid progenitor (CMP) cells, as well as in megakaryocytic/erythroid progenitor cells (MEPs). Although these mice did not display the increase in granulopoiesis commonly found in chronic myeloid leukemia (CML), the phenotype closely resembles a myeloproliferative disorder affecting the megakaryocytic lineage observed in some patients with the BCR/ABL P210 translocation.


1995 ◽  
Vol 15 (7) ◽  
pp. 3786-3795 ◽  
Author(s):  
Q Lu ◽  
P S Knoepfler ◽  
J Scheele ◽  
D D Wright ◽  
M P Kamps

E2A-PBX1 is the oncogene produced at the t(1;19) chromosomal breakpoint of pediatric pre-B-cell leukemia. Expression of E2A-Pbx1 induces fibroblast transformation and myeloid and T-cell leukemia in mice and arrests differentiation of granulocyte macrophage colony-stimulating factor-dependent myeloblasts in cultured marrow. Recently, the Drosophila melanogaster protein Exd, which is highly related to Pbx1, was shown to bind DNA cooperatively with the Drosophila homeodomain proteins Ubx and Abd-A. Here, we demonstrate that the normal Pbx1 homeodomain protein, as well as its oncogenic derivative, E2A-Pbx1, binds the DNA sequence ATCAATCAA cooperatively with the murine Hox-A5, Hox-B7, Hox-B8, and Hox-C8 homeodomain proteins, which are themselves known oncoproteins, as well as with the Hox-D4 homeodomain protein. Cooperative binding to ATCAATCAA required the homeodomain-dependent DNA-binding activities of both Pbx1 and the Hox partner. In cotransfection assays, Hox-B8 suppressed transactivation by E2A-Pbx1. These results suggest that (i) Pbx1 may participate in the normal regulation of Hox target gene transcription in vivo and therein contribute to aspects of anterior-posterior patterning and structural development in vertebrates, (ii) that E2A-Pbx1 could abrogate normal differentiation by altering the transcriptional regulation of Hox target genes in conjunction with Hox proteins, and (iii) that the oncogenic mechanism of certain Hox proteins may require their physical interaction with Pbx1 as a cooperating, DNA-binding partner.


Author(s):  
Erna Yang ◽  
Wei Guan ◽  
Desheng Gong ◽  
Jieying Li ◽  
Caixia Han ◽  
...  

AbstractThe formation of the RUNX1-RUNX1T1 fusion protein, resulting from the t(8;21) translocation, is considered to be one of the initiating events of t(8;21) acute myeloid leukemia (AML). However, the mechanisms of the oncogenic mechanism of RUNX1-RUNX1T1 remain unclear. In this study, we found that RUNX1-RUNX1T1 triggers the heterochromatic silencing of UBXN8 by recognizing the RUNX1-binding sites and recruiting chromatin-remodeling enzymes to the UBXN8 promoter region. Decitabine, a specific inhibitor of DNA methylation, upregulated the expression of UBXN8 in RUNX1-RUNX1T1+ AML cell lines. Overexpression of UBXN8 inhibited the proliferation and colony-forming ability of and promoted cell cycle arrest in t(8;21) AML cell lines. Enhancing UBXN8 levels can significantly inhibit tumor proliferation and promote the differentiation of RUNX1-RUNX1T1+ cells in vivo. In conclusion, our results indicated that epigenetic silencing of UBXN8 via methylation of its promoter region mediated by the RUNX1-RUNX1T1 fusion protein contributes to the leukemogenesis of t(8;21) AML and that UBXN8 targeting may be a potential therapeutic strategy for t(8;21) AML.


Sign in / Sign up

Export Citation Format

Share Document