Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
2.
Oncogene ; 25(35): 4840-7, 2006 Aug 10.
Article in English | MEDLINE | ID: mdl-16547490

ABSTRACT

The transcription factor hypoxia inducible factor 1 (HIF1), an HIF1alpha-aryl hydrocarbon receptor nuclear translocator (ARNT) dimeric factor, is essential to the cellular response to hypoxia. We described a t(1;12)(q21;p13) chromosomal translocation in human acute myeloblastic leukemia that involves the translocated Ets leukemia (TEL/ETV6) and the ARNT genes and results in the expression of a TEL-ARNT fusion protein. Functional studies show that TEL-ARNT interacts with HIF1alpha and the complex binds to consensus hypoxia response element. In low oxygen tension conditions, the HIF1alpha/TEL-ARNT complex does not activate transcription but exerts a dominant-negative effect on normal HIF1 activity. Differentiation of normal human CD34+ progenitors cells along all the erythrocytic, megakaryocytic and granulocytic pathways was accelerated in low versus high oxygen tension conditions. Murine 32Dcl3 myeloid cells also show accelerated granulocytic differentiation in low oxygen tension in response to granulocyte colony-stimulating factor. Interestingly, stable expression of the TEL-ARNT in 32Dcl3 subclones resulted in impaired HIF1-mediated transcriptional response and inhibition of differentiation enhancement in hypoxic conditions. Taken together, our results underscore the role of oxygen tension in the modulation of normal hematopoietic differentiation, whose targeting can participate in human malignancies.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/physiology , Hematopoiesis/genetics , Oxygen/blood , Proto-Oncogene Proteins c-ets/physiology , Recombinant Fusion Proteins/physiology , Repressor Proteins/physiology , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Cell Differentiation/genetics , Cell Line , HeLa Cells , Humans , Mice , Proto-Oncogene Proteins c-ets/genetics , Recombinant Fusion Proteins/genetics , Repressor Proteins/genetics , ETS Translocation Variant 6 Protein
3.
Exp Cell Res ; 271(2): 277-85, 2001 Dec 10.
Article in English | MEDLINE | ID: mdl-11716540

ABSTRACT

Megakaryocytes (Mks) are unique cells in the human body in that they carry a single and polyploid nucleus. It is therefore of interest to understand their nuclear ultrastructure. PML oncogenic domains (PODs) were described in several types of eukaryotic cells using human autoantibodies which recognize nuclear antigens with a specific speckled pattern (dots) in indirect immunofluorescence (IF). Two main antigens, PML and Sp 100, usually colocalize and concentrate in these nuclear subdomains. We investigated the presence of PODs using IF and immunoelectron microscopy (IEM) in cells from megakaryocytic lineage: the HEL cell line and human cultured Mks. Antibodies against PML, Sp100, and anti-nuclear dots were used in single and double labeling. PODs were identified in HEL cells and in human Mks, and their ultrastructure was characterized. We then used IF to quantify PODs within Mks and showed that their number increased proportionally to nuclear lobularity. In summary, we report the identification of PODs in human Mks at an ultrastructural level and an increase in PODs number in parallel with Mk ploidy. We show that endomitosis not only leads to DNA increase but also to the multiplication of at least one of the associated nuclear structures.


Subject(s)
Antigens, Nuclear , Autoantigens/metabolism , Cell Compartmentation/genetics , Cell Nucleus/ultrastructure , Megakaryocytes/ultrastructure , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Oncogenes/genetics , Transcription Factors/metabolism , Autoantibodies , Autoantigens/genetics , Cell Nucleus/metabolism , Cells, Cultured/metabolism , Cells, Cultured/ultrastructure , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Megakaryocytes/metabolism , Microscopy, Electron , Mitosis/physiology , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Polyploidy , Promyelocytic Leukemia Protein , Protein Structure, Tertiary/genetics , Spindle Apparatus/metabolism , Spindle Apparatus/ultrastructure , Transcription Factors/genetics , Tumor Suppressor Proteins
4.
Oncogene ; 20(18): 2197-204, 2001 Apr 26.
Article in English | MEDLINE | ID: mdl-11402314

ABSTRACT

In this study, we show that upon thrombopoietin (Tpo) stimulation the two adapter proteins Gab1 and Gab2 are strongly tyrosine phosphorylated and associated with Shc, SHP2, PI 3-kinase and Grb2 in mpl-expressing UT7 cells. Although Gab1 and Gab2 seem to mediate overlapping biological signals in many cells, only Gab1 is expressed and phosphorylated in response to Tpo in primary human megakaryocytic progenitors; furthermore, it associates with the same proteins. Although a low level of tyrosine phosphorylated IRS-2 protein is also detected in PI 3-kinase immunoprecipitates, Gab proteins are the essential proteins associated with PI 3-kinase after Tpo stimulation. We demonstrate that, albeit no association is detected between the Tpo receptor mpl and Gab proteins, Y112 located in the C-terminal cytoplasmic domain of mpl is required for Gab1/2 tyrosine phosphorylation. Gab proteins are not tyrosine phosphorylated after Tpo stimulation of UT-7 and Ba/F3 cells expressing a mpl mutant lacking Y112. Moreover, no activation of the PI 3-kinase/Akt pathway is observed in cells expressing this mpl mutant. Finally, we show that this mutant does not allow cell proliferation, thereby confirming that PI 3-kinase activation is required for Tpo-induced cell proliferation.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/physiology , Protein Serine-Threonine Kinases , Thrombopoietin/pharmacology , Adaptor Proteins, Signal Transducing , Animals , Cell Division/drug effects , Cell Division/physiology , Enzyme Activation/drug effects , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Megakaryocytes/cytology , Megakaryocytes/drug effects , Megakaryocytes/metabolism , Mice , Phosphoproteins/metabolism , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rabbits , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Thrombopoietin/genetics , Tyrosine/metabolism
5.
Blood ; 97(7): 1968-74, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11264160

ABSTRACT

In adult bone marrow, mature erythroblasts are produced within structures called erythroblastic islands and then cross the endothelial barrier to reach circulation. Erythroblastic islands are composed of a central macrophage surrounded by maturing erythroblasts. In this study, it is shown that erythroid cells, but not the other mature hematopoietic cells, coexpress 2 angiogenic factors, vascular endothelial growth factor A (VEGF-A) and placenta growth factor (PlGF). Secretion of both VEGF-A and PlGF increases during in vitro erythroid differentiation. Erythroblast-conditioned medium can induce both migration of monocytes and endothelial cells and the permeability of endothelial cells. These effects are inhibited by anti-PlGF and/or anti-VEGF antibodies. Finally, it is shown that VEGF-A and PlGF proteins are expressed by bone marrow erythroblasts in vivo. Angiogenic factors secreted by erythroblasts may promote interactions either with macrophages in erythroblastic islands or with endothelial cells that would facilitate the passage of erythroid cells through the endothelial barrier. (Blood. 2001;97:1968-1974)


Subject(s)
Endothelial Growth Factors/biosynthesis , Erythroid Precursor Cells/metabolism , Pregnancy Proteins/biosynthesis , Animals , Bone Marrow/pathology , Cattle , Cell Differentiation/drug effects , Cell Membrane Permeability/drug effects , Cell Movement/drug effects , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Culture Media, Conditioned/pharmacology , Culture Media, Serum-Free , Endothelial Growth Factors/genetics , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/cytology , Erythroid Precursor Cells/drug effects , Erythropoiesis/genetics , Gene Expression Regulation, Developmental , Humans , Placenta Growth Factor , Pregnancy Proteins/genetics , Pregnancy Proteins/pharmacology , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Stem Cell Factor/pharmacology , Vascular Endothelial Growth Factor A
6.
J Biol Chem ; 276(3): 2007-14, 2001 Jan 19.
Article in English | MEDLINE | ID: mdl-11053408

ABSTRACT

To identify new proteins involved in erythropoietin (Epo) signal transduction, we purified the entire set of proteins reactive with anti-phosphotyrosine antibodies from Epo-stimulated UT7 cells. Antisera generated against these proteins were used to screen a lambdaEXlox expression library. One of the isolated cDNAs encodes Gbeta2, the beta2 subunit of heterotrimeric GTP-binding proteins. Gbeta and Galpha(i) coprecipitated with the Epo receptor (EpoR) in extracts from human and murine cell lines and from normal human erythroid progenitor cells. In addition, in vitro Gbeta associated with a fusion protein containing the intracellular domain of the EpoR. Using EpoR mutants, we found that the distal part of the EpoR (between amino acids 459-479) was required for Gi binding. Epo activation of these cells induced the release of the Gi protein from the EpoR. Moreover in isolated cell membranes, Epo treatment inhibited ADP-ribosylation of Gi and increased the binding of GTP. Our results show that heterotrimeric Gi proteins associate with the C-terminal end of the EpoR. Receptor activation leads to the activation and dissociation of Gi from the receptor, suggesting a functional role of Gi protein in Epo signal transduction.


Subject(s)
Heterotrimeric GTP-Binding Proteins/metabolism , Receptors, Erythropoietin/metabolism , Animals , Cytoplasm/metabolism , Erythropoietin/pharmacology , Humans , Mice , Protein Binding , Receptors, Erythropoietin/drug effects , Recombinant Proteins/metabolism , Tumor Cells, Cultured
7.
Cell Growth Differ ; 11(11): 573-80, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11095246

ABSTRACT

In the present study, we demonstrate that erythropoietin (Epo) induces the expression and the release of tissue inhibitors of metalloproteinase-1 (TIMP-1) in a time- and dose-dependent manner in Epo-dependent cell line UT-7 cells and in normal human erythroid progenitor cells from cord blood (CD36+) and required de novo protein synthesis. TIMP-1 was not expressed in the absence of Epo. Inhibition of the mitogen-activated protein kinase pathway by the specific inhibitors PD98059 and U0126 and of phosphatidylinositol 3-kinase by LY294002, strongly inhibited Epo-induced TIMP-1 expression and secretion. In the absence of Epo, both latent and active forms of matrix metalloproteinase-9 (MMP-9) were secreted into media. Upon Epo stimulation, MMP-9 and pro-MMP-9 secretion was inhibited in a dose-dependent manner parallel to TIMP-1 induction. The addition of PD98059, U0126, and LY294002 in the presence of Epo restored MMP-9 production in UT-7 and CD36+ cells. Our findings strongly suggest an inversely coordinated regulation of the TIMP-1 gene and MMP-9 production by Epo via mitogen-activated protein kinase and phosphatidylinositol 3-kinase pathways.


Subject(s)
Erythropoietin/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Butadienes/pharmacology , CD36 Antigens/metabolism , Chromones/pharmacology , Cycloheximide/pharmacology , Dose-Response Relationship, Drug , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Erythropoietin/antagonists & inhibitors , Flavonoids/pharmacology , Gene Expression Regulation/drug effects , Humans , MAP Kinase Kinase 1 , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/metabolism , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Morpholines/pharmacology , Nitriles/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tissue Inhibitor of Metalloproteinase-1/genetics , Tumor Cells, Cultured
8.
Exp Hematol ; 28(8): 885-94, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10989189

ABSTRACT

Erythropoiesis is positively regulated by stem cell factor, interleukin 3, and erythropoietin, which synergize to allow the production of hemoglobinized red blood cells from erythroid progenitors. In contrast, interferon gamma, tumor necrosis factor alpha, and transforming growth factor B(1), (TGF-beta(1)) are powerful inhibitors of erythropoiesis. Interferon gamma and alpha act principally by inducing apoptosis. The aim of this study was to elucidate the mechanisms by which TGF-beta(1) inhibits erythropoiesis. We used an in vitro serum-free system of human red blood cell production. From a virtually pure population of CD36(+) erythroid progenitors, stem cell factor, interleukin 3, and erythropoietin allowed massive proliferation (x300) and promoted terminal red blood cell differentiation. We show here that TGF-beta(1) (2 ng/mL) inhibited the growth of CD36(+) cells by 15-fold. TGF-beta(1) markedly accelerated and increased erythroid differentiation as assessed by hemoglobin and glycophorin expression. Furthermore, May-Grünwald-Giemsa staining and ultrastructural analysis revealed that TGF-beta(1) induced full differentiation toward normal enucleated red cells even in the absence of macrophages. This acceleration of erythroid differentiation did not modify the pattern of hemoglobin chains expression from adult or fetal erythroid progenitors. Analysis of apoptosis, cell cycle and Ki-67 expression showed that TGF-beta(1) inhibited cell proliferation by decreasing the cycle of immature erythroid cells and accelerating maturation toward orthochromatic normoblasts that are not in cycle. We showed that TGF-beta(1) is a paradoxical inhibitor of erythropoiesis that acts by blocking proliferation and accelerating differentiation of erythroid progenitors.


Subject(s)
Cell Differentiation , Cell Division , Erythroid Precursor Cells/cytology , Erythropoiesis , Transforming Growth Factor beta/pharmacology , Apoptosis , CD36 Antigens/analysis , Cell Cycle , Erythroblasts/ultrastructure , Erythropoietin/pharmacology , Glycophorins/biosynthesis , Hemoglobins/biosynthesis , Humans , Interleukin-3/pharmacology , Stem Cell Factor/pharmacology
10.
Br J Haematol ; 106(4): 912-22, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10519992

ABSTRACT

In humans, studies of the erythroid cell lineage are hampered by difficulties in obtaining sufficient numbers of erythroid progenitors. In fact, these progenitors in bone marrow or peripheral blood are scarce and no specific antibodies are available. We describe a new method which allows proliferation in liquid culture of large numbers of pure normal human erythroid progenitors. CD34+ cells were cultured for 7 d in serum-free conditions with the cytokine mixture interleukin (IL)-3/IL-6/stem cell factor (SCF). This resulted in cell expansion and the appearance of a high proportion of CD36+ cells which were purified on day 7. Methylcellulose clones from these cells were composed of 96.6% late BFU-E and 3.4% CFU-GM. These CD36+ cells could be recultured with the same cytokine mixture plus or minus erythropoietin (Epo) for a further 2-7 d. In both conditions further amplification of CD36+ cells was observed, but Epo induced a more dramatic cell expansion. Glycophorin-positive mature cells appeared only in the presence of Epo, and terminal red cell differentiation was observed after 7 d of secondary culture. Cells obtained from adult CD34+ progenitors mostly contained adult haemoglobin, whereas cord blood-derived cells contained equal proportions of adult and fetal haemoglobin. Activation of STAT5 and tyrosine phosphorylation of the Epo receptor and JAK2 were observed after Epo stimulation of these cells. This new method represents a straightforward alternative to the procedures previously described for the purification of normal erythroid progenitors and is useful in the study of erythropoietic regulation.


Subject(s)
Erythroid Precursor Cells/cytology , Antigens, CD34/metabolism , CD36 Antigens/metabolism , Cell Differentiation , Cell Division , Cell Separation/methods , Cells, Cultured , Cytokines/physiology , Erythropoietin/physiology , Flow Cytometry/methods , Hemoglobins/analysis , Humans
11.
Blood ; 94(5): 1601-13, 1999 Sep 01.
Article in English | MEDLINE | ID: mdl-10477685

ABSTRACT

In several erythroleukemia cell lines, activation of mitogen-activated protein kinases (MAPK) by phorbol esters or megakaryocyte growth and development factor (MGDF) is required for induction of megakaryocytic phenotype and growth arrest. To support this model, we have examined the effect of a specific inhibitor of this pathway (PD98059) on human CD34(+) hematopoietic progenitors isolated from cord blood (CB), induced to differentiate along the megakaryocytic lineage in liquid cultures supplemented with rhuMGDF. RhuMGDF induced a sustained activation of MAPK in megakaryocytes and this activation was completely inhibited in the presence of low concentrations of PD98059 (6 to 10 micromol/L). At this concentration, PD98059 induced an increase in cell proliferation, resulting in accumulation of viable cells and a prolongation of the life time of the cultures. This increase correlated with an increase in DNA synthesis rather than with a reduction in apoptosis. This effect was combined with developmental changes indicative of delayed megakaryocytic differentiation: (1) PD98059-treated cells tended to retain markers of immature progenitors as shown by the increased proportion of both CD34(+) and CD41(+)CD34(+) cells. (2) PD98059-treated cultures were greatly enriched in immature blasts cells. (3) PD98059 increased megakaryocytic progenitors able to form colonies in semisolid assays. Thus, the MAPK pathway, although not required for megakaryocyte formation, seems to be involved in the transition from proliferation to maturation in megakaryocytes. Inhibition of MAPK activation also led to an increase in the number and size of erythroid colonies without affecting granulocyte/macrophage progenitor numbers suggesting that, in addition to the megakaryocytic lineage, the MAPK pathway could play a role in erythroid lineage differentiation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/physiology , Hematopoietic Stem Cells/physiology , Signal Transduction/drug effects , Thrombopoietin/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Fetal Blood/cytology , Flavonoids/pharmacology , Hematopoietic Stem Cells/cytology , Humans
12.
J Virol ; 73(10): 8762-70, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10482630

ABSTRACT

Human erythroid progenitor cells are the main target cells of the human parvovirus B19 (B19), and B19 infection induces a transient erythroid aplastic crisis. Several authors have reported that the nonstructural protein 1 (NS-1) encoded by this virus has a cytotoxic effect, but the underlying mechanism of NS-1-induced primary erythroid cell death is still not clear. In human erythroid progenitor cells, we investigated the molecular mechanisms leading to apoptosis after natural infection of these cells by the B19 virus. The cytotoxicity of NS-1 was concomitantly evaluated in transfected erythroid cells. B19 infection and NS-1 expression induced DNA fragmentation characteristic of apoptosis, and the commitment of erythroid cells to undergo apoptosis was combined with their accumulation in the G(2) phase of the cell cycle. Since B19- and NS-1-induced apoptosis was inhibited by caspase 3, 6, and 8 inhibitors, and substantial caspase 3, 6, and 8 activities were induced by NS-1 expression, there may have been interactions between NS-1 and the apoptotic pathways of the death receptors tumor necrosis factor receptor 1 and Fas. Our results suggest that Fas-FasL interaction was not involved in NS-1- or B19-induced apoptosis in erythroid cells. In contrast, these cells were sensitized to tumor necrosis factor alpha (TNF-alpha)-induced apoptosis. Moreover, the ceramide level was enhanced by B19 infection and NS-1 expression. Therefore, our results suggest that there may be a connection between the respective apoptotic pathways activated by TNF-alpha and NS-1 in human erythroid cells.


Subject(s)
Apoptosis , Erythrocytes/pathology , Erythrocytes/virology , Parvoviridae Infections/pathology , Parvoviridae Infections/virology , Parvovirus B19, Human , Tumor Necrosis Factor-alpha/metabolism , Viral Nonstructural Proteins/metabolism , Erythrocytes/metabolism , Humans , Parvoviridae Infections/metabolism , Signal Transduction
13.
Blood ; 93(8): 2578-85, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10194437

ABSTRACT

Five tyrosine-phosphorylated proteins with molecular masses of 180, 145, 116, 100, and 70 kD are associated with phosphatidylinositol 3-kinase (PI 3-kinase) in erythropoietin (Epo)-stimulated UT-7 cells. The 180- and 70-kD proteins have been previously shown to be IRS2 and the Epo receptor. In this report, we show that the 116-kD protein is the IRS2-related molecular adapter, GAB1. Indeed, Epo induced the transient tyrosine phosphorylation of GAB1 in UT-7 cells. Both kinetics and Epo dose-response experiments showed that GAB1 tyrosine phosphorylation was a direct consequence of Epo receptor activation. After tyrosine phosphorylation, GAB1 associated with the PI 3-kinase, the phosphotyrosine phosphatase SHP2, the phosphatidylinositol 3,4,5 trisphosphate 5-phosphatase SHIP, and the molecular adapter SHC. GAB1 was also associated with the molecular adapter GRB2 in unstimulated cells, and this association dramatically increased after Epo stimulation. Thus, GAB1 could be a scaffold protein able to couple the Epo receptor activation with the stimulation of several intracellular signaling pathways. Epo-induced tyrosine phosphorylation of GAB1 was also observed in normal human erythroid progenitors isolated from cord blood. Granulocyte-macrophage colony-stimulating factor (GM-CSF) and thrombopoietin (TPO) also induced the tyrosine phosphorylation of GAB1 in UT-7 cells, indicating that this molecule participates in the signal transduction of several cytokine receptors.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Erythropoietin/pharmacology , Hematopoietic Stem Cells/physiology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphoric Monoester Hydrolases/metabolism , Protein Tyrosine Phosphatases/metabolism , Proteins/metabolism , Amino Acid Sequence , Fetal Blood , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Intracellular Signaling Peptides and Proteins , Leukemia , Molecular Weight , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphorylation , Phosphotyrosine/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Recombinant Proteins/metabolism , SH2 Domain-Containing Protein Tyrosine Phosphatases , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Transfection , Tumor Cells, Cultured , src Homology Domains
14.
Biochem Biophys Res Commun ; 256(3): 685-91, 1999 Mar 24.
Article in English | MEDLINE | ID: mdl-10080960

ABSTRACT

To study the role of the cytoplasmic domain and particularly the tyrosine residues of the erythropoietin receptor (EpoR) in erythroid differentiation of human primary stem cells, we infected cord blood-derived CD34+ cells with retroviruses encoding chimeric receptors containing the extracellular domain of the prolactin receptor (PRLR) and the cytoplasmic domain of either the normal EpoR or a truncated EpoR devoid of tyrosine residues. Erythroid differentiation of the infected progenitors could thus be studied after stimulation by PRL. The complete PRLR was used to assess its ability to substitute for EpoR in erythroid differentiation. Typical erythroid day-14 colonies were observed from CD34+ cells grown in PRL when infected with any of the three viral constructs. These results demonstrate that: (i) the activation of the virally transduced PRLR leads to erythroid colony formation showing that erythroid terminal differentiation can be induced by a non-erythroid receptor in human progenitors; (ii) a chimeric receptor PRLR/EpoR is able to transduce a signal leading to terminal erythroid differentiation of human CD34+ cells; (iii) in contrast to results previously reported in murine models, tyrosine residues of the EpoR are not required for growth and terminal differentiation of human erythroid progenitors.


Subject(s)
Antigens, CD34/analysis , Erythrocytes/cytology , Erythroid Precursor Cells/cytology , Receptors, Erythropoietin/metabolism , Tyrosine/metabolism , Cell Differentiation/drug effects , Cell Division/drug effects , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Fetal Blood/cytology , Genetic Vectors/genetics , Growth Substances/pharmacology , Humans , Phosphorylation/drug effects , Precipitin Tests , Prolactin/pharmacology , RNA, Viral/analysis , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/genetics , Receptors, Prolactin/chemistry , Receptors, Prolactin/genetics , Receptors, Prolactin/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retroviridae/genetics , Sequence Deletion , Signal Transduction/drug effects , Transduction, Genetic , Tumor Cells, Cultured , Tyrosine/genetics
16.
Leuk Res ; 22(6): 527-35, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9678719

ABSTRACT

Thrombocytopenia is a frequent feature of myelodysplastic syndromes (MDS) that could be improved by the use of recombinant human megakaryocyte growth and development factor (rHuMGDF). Using short-term liquid cultures and progenitor assays, we have found that rHuMGDF stimulated DNA synthesis and potentiated leukemic cluster growth of bone marrow mononuclear cells in 10/38 MDS cases (26%). Cytogenetically malignant colonies were detectable in rHuMGDF-stimulated cultures (n=3) by fluorescence in situ hybridization. rHuMGDF was able to stimulate CFU-MK formation in 45% of the samples tested. Finally, rHuMGDF-induced blast cell proliferation correlated with elevated expression of c-MPL, previously identified as a bad prognosis factor in MDS.


Subject(s)
Blast Crisis/pathology , Megakaryocytes/cytology , Myelodysplastic Syndromes/pathology , Neoplasm Proteins , Receptors, Cytokine , Thrombopoietin/pharmacology , Blotting, Northern , Cell Culture Techniques , Cell Division/drug effects , Cell Division/genetics , Clone Cells/cytology , Colony-Forming Units Assay , DNA/biosynthesis , Hematopoietic Stem Cells/cytology , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Methylcellulose , Myelodysplastic Syndromes/classification , Proto-Oncogene Proteins/genetics , RNA/analysis , Receptors, Immunologic/genetics , Receptors, Thrombopoietin , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Thrombopoietin/biosynthesis , Thrombopoietin/genetics
18.
Cancer Genet Cytogenet ; 101(1): 12-5, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9460494

ABSTRACT

Myelodysplastic syndromes (MDS) are clonal malignancies characterized by peripheral blood pancytopenia and signs of maturation disturbances of one or several cell lineages in bone marrow. MDS present as chimeras associating normal polyclonal and malignant monoclonal progenitors cells in various proportions. Numerous cytogenetic abnormalities have been reported in MDS and can be detected by fluorescence in situ hybridization (FISH) on interphase cells. We have used this technique on methylcellulose cultured hematopoietic progenitors obtained from three patients suffering from MDS and exhibiting informative karyotypic features. Hematopoietic cells were cultured for 14 days, and individual clones (BFU-E, CFU-GM) were picked up and then cytocentrifuged for FISH analysis. We used centromeric probes realized and labeled in our laboratory by PCR to detect aneuploidies for chromosomes 7 and 11 in two patients. Furthermore, we could detect a 5q partial deletion on interphase cells from the third patient using a 5q31 specific probe visualized with the HNPP Fluorescent Detection Set from Boehringer Mannheim. In conclusion, FISH is a helpful method to detect malignant clones in hematopoietic progenitor cultures and hence to study the relative growth of normal vs. leukemic cells in MDS.


Subject(s)
Hematopoietic Stem Cells/pathology , In Situ Hybridization, Fluorescence/methods , Myelodysplastic Syndromes/diagnosis , Aged , Aneuploidy , Cells, Cultured , Centromere , Chromosome Aberrations/diagnosis , Chromosome Disorders , Female , Humans , Male , Methylcellulose , Middle Aged
19.
J Immunol ; 154(4): 1577-86, 1995 Feb 15.
Article in English | MEDLINE | ID: mdl-7836743

ABSTRACT

v-mpl is a constitutively activated, truncated form of a cytokine receptor that has been transduced in a murine retrovirus, the myeloproliferative leukemia virus (MPLV). Expression of this oncogene results in the factor-independent proliferation of myeloid, erythroid, megakaryocytic, and mast precursor cells, which retain the ability to differentiate. However, no lymphoid disease was ever reported. To determine whether MPLV could infect and transform very early B cells and their precursors (BCPs), lymphoid long-term bone marrow cultures were infected with a helper-free MPLV. Within 3 wk after infection, highly proliferating BCPs could be isolated. These cells were able to clone spontaneously in semi-solid cultures, grown in the absence of feeder cell layer or exogenous growth factor and rapidly produced tumors after s.c. injection into synegic irradiated mice. In addition, MPLV transformation of pre-B cells led to the induction of an autocrine activity. Immunophenotypic and molecular analysis indicated that MPLV transformed early pro-B, pro-B, and pre-B cells, according to the expression of HSA, CD43, B220, Thy1, s-IgM and BP1 Ags, and to the rearrangements of Ig genes. Interestingly, MPLV-transformed BCPs expressed Mac1 Ag without acquiring further characteristics of macrophagic differentiation. Although the v-mpl cytoplasmic domain is devoid of tyrosine kinase consensus sequence, MPLV-transformed pre-B cells contained a major approximately 105-kDa tyrosine-phosphorylated protein that was not detected in uninfected cells or in cells transformed by the Abelson viral oncogene (v-abl). These results demonstrate that, like v-abl, the truncated cytokine receptor v-mpl is able to transform BCPs in vitro and suggest that the oncogenic transformation of BCPs by either v-mpl or v-abl use different pathways.


Subject(s)
B-Lymphocytes/virology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Viral , Hematopoietic Stem Cells/virology , Leukemia Virus, Murine/physiology , Oncogenes , Receptors, Cytokine/physiology , Animals , Antigens, Differentiation/biosynthesis , Base Sequence , Cell Differentiation , Cell Division , Cell Line, Transformed/transplantation , Cells, Cultured , Culture Media, Conditioned , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Neoplasm Transplantation , Neoplasms, Experimental , Oncogene Proteins v-abl/physiology , Phosphoproteins/metabolism , Phosphorylation , Protein Processing, Post-Translational , Protein-Tyrosine Kinases/metabolism , Receptors, Cytokine/genetics
20.
Blood ; 83(5): 1348-54, 1994 Mar 01.
Article in English | MEDLINE | ID: mdl-8118036

ABSTRACT

Specific rearrangements involving 3q21 and 3q26 are well documented in acute myeloid leukemia (AML). Aberrant expression of the Ecotropic virus integration-1 (EVI1) gene, located at 3q26, has been reported in individuals with AML and translocations or inversions of chromosome 3 long arm. We have studied six individuals with AML and inv(3)(q21q26) for disruptions to the EVI1 locus by in situ hybridization and long-range mapping. EVI1 transcripts have been detected in the blast cells of the two individuals available for expression studies. We derived a YAC containing the EVI1 gene and showed that it crossed the 3q26 inversion breakpoints in three of four cases examined. Pulsed field analysis detected aberrant fragments 3' of the EVI1 gene in all six patients. The orientation of the gene was established and the locations of the breakpoints were refined by in situ hybridization using phage clones from this region.


Subject(s)
Chromosomes, Human, Pair 3 , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/genetics , Proto-Oncogenes , Transcription Factors , Base Sequence , Chromosome Aberrations/genetics , Chromosome Disorders , Chromosome Inversion , Chromosomes, Artificial, Yeast , Cloning, Molecular , DNA Primers/chemistry , Gene Rearrangement , Humans , In Situ Hybridization, Fluorescence , MDS1 and EVI1 Complex Locus Protein , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Restriction Mapping , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL
...