Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Oncogene ; 25(9): 1420-3, 2006 Mar 02.
Article in English | MEDLINE | ID: mdl-16247441

ABSTRACT

The erythroleukemia developed by spi-1/PU.1 transgenic mice is a multistep process. At disease onset, preleukemic cells are arrested in differentiation at the proerythroblast stage (HS1 stage) and their survival and growth are under the tight control of erythropoietin (Epo). During disease progression, malignant proerythroblasts characterized by Epo autonomous growth and in vivo tumorigenicity can be isolated (HS2 stage). During analysis of transcriptional profiling representive of discrete stages of leukemic progression, we found that the phosphatidylinositol 4-phosphatase type II gene was turned off in malignant cells. PI-4-phosphatase II is an enzyme that hydrolyses the 4-phosphate position of phosphatidylinositol-3-4-bisphosphate (PtdIns(3,4)P(2)) to form PtdIns(3)P. Using malignant cells engineered to stably express PI-4-phosphatase II, we showed that PI-4-phosphatase II reduced Akt activation level. Moreover, stimulation of malignant cells with Epo-induced PI-4-phosphatase II transcription pointing this gene as an Epo-responsive gene. This study provides first insight for a physiological role of PI-4-phosphatase II in the proerythroblast by controlling Epo responsiveness through a negative regulation of the PI3K/Akt pathway.


Subject(s)
Erythropoietin/physiology , Gene Expression Regulation, Neoplastic , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/physiopathology , Phosphoric Monoester Hydrolases/biosynthesis , Phosphoric Monoester Hydrolases/genetics , Animals , Blotting, Northern , Cell Differentiation , Cell Survival , Cell Transformation, Neoplastic , Erythroblasts , Gene Expression Profiling , Mice , Mice, Transgenic , Oncogene Protein v-akt/physiology , Phosphatidylinositol 3-Kinases/physiology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transcription, Genetic , Tumor Cells, Cultured
2.
Blood ; 98(8): 2372-81, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11588033

ABSTRACT

During the cell transformation processes leading to erythroleukemia, erythroid progenitors often become erythropoietin (Epo)-independent for their proliferation. The biochemical events that could lead an erythroleukemic cell to growth factor-independence were investigated using spi-1 transgenic poerythroblasts. Spi-1/PU.1 is a myeloid and B-cell transcription factor of the ETS family and is activated by insertional mutagenesis during Friend erythroleukemia. Its overexpression in proerythroblasts induces their differentiation arrest without altering their erythropoietin requirement for proliferation (HS1 cells). At a later step, genetic alterations most probably occur allowing spi-1 transgenic poerythroblasts to proliferate in the absence of erythropoietin (HS2 cells). The signaling transduction pathways in HS1 and HS2 proerythroblasts were analyzed. The authors have previously shown that the Jak/STAT pathway was not activated in Epo-independent cells, but remained sensitive to Epo stimulation. In the present study, it is shown that the Epo-independent proliferation of HS2 cells requires active phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways. In these cells, PI3K was constitutively associated with the molecular adapters Grb2 and Gab1, and with the phosphatases SHP-2 and SHIP. Moreover, PI3K activity was correlated with the constitutive phosphorylation of serine-threonine protein kinase (AKT) in HS2 cells. Lastly, a constitutive activation of the MAPKs extracellular signal-regulated kinases (ERK1/2) in HS2 cells was observed that occurs in a PI3K-independent manner, but depends strictly on the activity of the protein kinase C (PKC). These results suggest that constitutive activations of PI3K/AKT and PKC/MAPK pathways can act in synergy to lead a proerythroblast to proliferate without Epo.


Subject(s)
Adaptor Proteins, Signal Transducing , Erythroblasts/physiology , Erythropoietin/pharmacology , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/blood , Phosphatidylinositol 3-Kinases/blood , Receptors, Erythropoietin/genetics , Animals , Base Sequence , Cell Division/drug effects , Cells, Cultured , DNA Primers , DNA, Complementary , Erythroblasts/drug effects , GRB2 Adaptor Protein , Kinetics , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Kinase C/metabolism , Protein Subunits , Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
3.
Oncogene ; 20(39): 5484-92, 2001 Sep 06.
Article in English | MEDLINE | ID: mdl-11571646

ABSTRACT

Activation of the spi-1/PU.1 proto-oncogene and loss of p53 function are genetic alterations associated with the emergence of Friend malignant erythroleukemic cells. To address the role of p53 during erythroleukemogenesis, spi-1 transgenic mice (spi-1-Tg) which develop erythroleukemia were bred with p53-deficient mice. Three classes of spi-1 transgenic mice differing in their p53 functional status (p53(+/+), p53(+/-) and p53(-/-)) were generated. These mice developed a unique pattern of erythroleukemia. In wild-type p53 spi-1-Tg mice, none of the primary erythroleukemic spleen cells displayed autonomous growth in vitro and in vivo. In contrast, in p53(+/-) spi-1-Tg mice, erythroleukemic cells gave rise to growth factor-independent cell lines and generated tumors in vivo. Malignancy was associated with loss of the wild-type p53 allele. The p53(-/-) spi-1-Tg mice developed erythroleukemia with a total incidence and a reduced latency compared to the two other genotypes. Unexpectedly, 50% of p53(-/-) spi-1-Tg erythroleukemic spleens generated cell lines that were strictly dependent upon erythropoietin (Epo) for proliferation, whereas the remainder proliferated independently of cytokines. Moreover, only 70% of these spleen cells were tumorigenic. These findings indicate that p53 germ-line deletion did not confer malignancy to spi-1-transgenic proerythroblasts. Moreover Epo independence and tumorigenicity appear as separable phenotypic characteristics revealing that the spi-1-Tg proerythroblasts progress towards malignancy through multiple oncogenic events.


Subject(s)
Erythroid Precursor Cells/pathology , Genes, p53 , Leukemia, Erythroblastic, Acute/etiology , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Tumor Suppressor Protein p53/physiology , Animals , Animals, Newborn , Blood Cells/cytology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , Disease Progression , Erythroid Precursor Cells/metabolism , Female , Gene Expression Regulation, Neoplastic , Germ-Line Mutation , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/pathology , Male , Mice , Mice, Knockout , Mice, Transgenic , RNA, Neoplasm/biosynthesis , Spleen/cytology , Spleen/metabolism , Survival Rate , Tumor Cells, Cultured
4.
Oncogene ; 16(23): 2989-95, 1998 Jun 11.
Article in English | MEDLINE | ID: mdl-9662331

ABSTRACT

Spi-1 transcriptional activation and wild-type p53 extinction are two oncogenic alterations involved in the malignant transformation of erythroblasts during the Friend acute erythroleukemia. To dissect the contribution of these alterations in the deregulation of the differentiation and proliferation of erythroblasts, we generated spi-1 transgenic mice. Analysis of these animals revealed that Spi-1 overexpression was directly involved in the block of proerythroblast differentiation. However, the erythroleukemia that develops in these animals evolved in two steps. During the early step (HS1 step), non tumorigenic proerythroblasts remained strictly dependent upon erythropoietin (Epo) for their survival and proliferation. Later on, Epo-independent and tumorigenic proerythroblasts emerged (HS2 step) suggesting that other oncogenes cooperate with Spi-1 to lead to a fully malignant phenotype. By provirus tagging, we demonstrate that the HS1 step was clonal indicating that a cell selection must occur in vivo. Analysis of the nature of p53 in both the in vivo HS1 and HS2 proerythroblasts and in cultured erythroblastic cell lines showed that--p53 was normal in the HS1 primary tissues but was mutated in the HS1 cultured cell lines--p53 was frequently altered in HS2 primary tissues but was found normal in some mice. These data indicate that (i) the blockage of the erythroblast differentiation by Spi-1 occurs independently of p53 alteration (ii) p53 alteration is not necessary to confer Epo independence and tumorigenicity to spi-1 transgenic proerythroblasts.


Subject(s)
Leukemia, Erythroblastic, Acute/genetics , Mutation , Proto-Oncogene Proteins/physiology , Trans-Activators/physiology , Tumor Suppressor Protein p53/genetics , Animals , Clone Cells , Mice , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics
5.
J Biol Chem ; 273(9): 4838-42, 1998 Feb 27.
Article in English | MEDLINE | ID: mdl-9478924

ABSTRACT

Spi-1/PU.1 is an Ets protein deregulated by insertional mutagenesis during the murine Friend erythroleukemia. The overexpression of the normal protein in a proerythroblastic cell prevents its terminal differentiation. In normal hematopoiesis Spi-1/PU.1 is a transcription factor that plays a key role in normal myeloid and B lymphoid differentiation. Moreover, Spi-1/PU.1 binds RNA and interferes in vitro with the splicing process. Here we report that Spi-1 interacts in vivo with TLS (translocated in liposarcoma), a RNA-binding protein involved in human tumor-specific chromosomal translocations. This interaction appears functionally relevant, since TLS is capable of reducing the abilities of Spi-1/PU.1 to bind DNA and to transactivate the expression of a reporter gene. In addition, we observe that TLS is potentially a splicing factor. It promotes the use of the distal 5' splice site during the E1A pre-mRNA splicing. This effect is counterpoised in vivo by Spi-1. These data suggest that alteration of pre-mRNA alternative splicing by Spi-1 could be involved in the transformation of an erythroblastic cell.


Subject(s)
Alternative Splicing , Proto-Oncogene Proteins/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoproteins/metabolism , Trans-Activators/metabolism , Animals , Heterogeneous-Nuclear Ribonucleoproteins , Leukemia, Erythroblastic, Acute , Mice , Protein Binding , RNA-Binding Protein FUS , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...