Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Oncogene ; 34(5): 631-8, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-24469042

ABSTRACT

Constitutional dominant loss-of-function mutations in the SPRED1 gene cause a rare phenotype referred as neurofibromatosis type 1 (NF1)-like syndrome or Legius syndrome, consisted of multiple café-au-lait macules, axillary freckling, learning disabilities and macrocephaly. SPRED1 is a negative regulator of the RAS MAPK pathway and can interact with neurofibromin, the NF1 gene product. Individuals with NF1 have a higher risk of haematological malignancies. SPRED1 is highly expressed in haematopoietic cells and negatively regulates haematopoiesis. SPRED1 seemed to be a good candidate for leukaemia predisposition or transformation. We performed SPRED1 mutation screening and expression status in 230 paediatric lymphoblastic and acute myeloblastic leukaemias (AMLs). We found a loss-of-function frameshift SPRED1 mutation in a patient with Legius syndrome. In this patient, the leukaemia blasts karyotype showed a SPRED1 loss of heterozygosity, confirming SPRED1 as a tumour suppressor. Our observation confirmed that acute leukaemias are rare complications of the Legius syndrome. Moreover, SPRED1 was significantly decreased at RNA and protein levels in the majority of AMLs at diagnosis compared with normal or paired complete remission bone marrows. SPRED1 decreased expression correlated with genetic features of AML. Our study reveals a new mechanism which contributes to deregulate RAS MAPK pathway in the vast majority of paediatric AMLs.


Subject(s)
Cafe-au-Lait Spots/genetics , Genes, ras/genetics , Intracellular Signaling Peptides and Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Membrane Proteins/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adaptor Proteins, Signal Transducing , Adolescent , Cafe-au-Lait Spots/complications , Cafe-au-Lait Spots/pathology , Child , Child, Preschool , Female , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Infant , Infant, Newborn , Intracellular Signaling Peptides and Proteins/biosynthesis , Leukemia, Myeloid, Acute/complications , Leukemia, Myeloid, Acute/pathology , Loss of Heterozygosity/genetics , Male , Membrane Proteins/biosynthesis , Mutation , Neurofibromin 1/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology
2.
Cell Death Differ ; 17(11): 1739-50, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20467439

ABSTRACT

The early-response gene product IEX-1 (also known as IER3) was recently found to interact with the anti-apoptotic Bcl-2 family member, myeloid cell leukemia-1 (Mcl-1). In this study we show that this interaction specifically and timely controls the accumulation of Mcl-1 in the nucleus in response to DNA damage. The IEX-1 protein is rapidly induced by γ-irradiation, genotoxic agents or replication inhibitors, in a way dependent on ataxia telangiectasia mutated (ATM) activity and is necessary for Mcl-1 nuclear translocation. Conversely, IEX-1 protein proteasomal degradation triggers the return of Mcl-1 to the cytosol. IEX-1 and Mcl-1 are integral components of the DNA damage response. Loss of IEX-1 or Mcl-1 leads to genomic instability and increased sensitivity to genotoxic and replicative stresses. The two proteins cooperate to maintain Chk1 activation and G2 checkpoint arrest. Mcl-1 nuclear translocation may foster checkpoint and improve the tumor resistance to DNA damage-based cancer therapies. Deciphering the pathways involved in IEX-1 degradation should lead to the discovery of new therapeutic targets to increase sensitivity of tumor cells to chemotherapy.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Checkpoint Kinase 1 , Checkpoint Kinase 2 , Genes, bcl-2 , Genomic Instability , Humans , Immediate-Early Proteins/deficiency , Membrane Proteins/metabolism , Mice , Mitochondria/metabolism , Mitosis , Myeloid Cell Leukemia Sequence 1 Protein , Phosphorylation , Protein Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/deficiency
5.
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
6.
Mol Cell Biol ; 21(8): 2659-70, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11283246

ABSTRACT

Thrombopoietin (TPO) regulates growth and differentiation of megakaryocytes. We previously showed that extracellular signal-regulated kinases (ERKs) are required for TPO-mediated full megakaryocytic maturation in both normal progenitors and a megakaryoblastic cell line (UT7) expressing the TPO receptor (Mpl). In these cells, intensity and duration of TPO-induced ERK signal are controlled by several regions of the cytoplasmic domain of Mpl. In this study, we explored the signaling pathways involved in this control. We show that the small GTPases Ras and Rap1 contribute together to TPO-induced ERK activation in UT7-Mpl cells and that they do so by activating different Raf kinases as downstream effectors: a Ras-Raf-1 pathway is required to initiate ERK activation while Rap1 sustains this signal through B-Raf. Indeed, (i) in cells expressing wild-type or mutant Mpl, TPO-induced Ras and Rap1 activation correlates with early and sustained phases of ERK signal, respectively; (ii) interfering mutants of Ras and Rap1 both inhibit ERK kinase activity and ERK-dependent Elk1 transcriptional activation in response to TPO; (iii) the kinetics of activation of Raf-1 and B-Raf by TPO follow those of Ras and Rap1, respectively; (iv) RasV12-mediated Elk1 activation was modulated by the wild type or interfering mutants of Raf-1 but not those of B-Raf; (v) Elk1 activation mediated by a constitutively active mutant of Rap1 (Rap1V12) is potentiated by B-Raf and inhibited by an interfering mutant of this kinase. UT7-Mpl cells represent the second cellular model in which Ras and Rap1 act in concert to modulate the duration of ERK signal in response to a growth factor and thereby the differentiation program. This is also, to our knowledge, the first evidence suggesting that Rap1 may play an active role in megakaryocytic maturation.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Neoplasm Proteins , Proto-Oncogene Proteins c-raf/metabolism , Receptors, Cytokine , Thrombopoietin/pharmacology , rap1 GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Animals , Cell Line , Enzyme Activation/drug effects , Humans , Mice , Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-raf/genetics , Receptors, Thrombopoietin , Signal Transduction , rap1 GTP-Binding Proteins/genetics
7.
Oncogene ; 19(16): 2033-42, 2000 Apr 13.
Article in English | MEDLINE | ID: mdl-10803464

ABSTRACT

Mpl is the receptor for thrombopoietin, the primary regulator of platelet production by megakaryocytes. Upon stimulation by its ligand, Mpl receptor induces proliferation and differentiation of hematopoietic cell lines of various origins. In this paper, we show that Mpl is also able to transform FRE rat fibroblasts in the presence of MGDF (pegylated Megakaryocyte Growth and Development Factor), a modified form of its ligand. We also demonstrate that upon MGDF stimulation Mpl receptor activates the classical transduction pathways described for hematopoietic cell lines in FRE cells. Introduction of Mpl deletion mutants in FRE cells allowed us to demonstrate that the C-terminal region of the Mpl intracytoplasmic domain, which is involved in hematopoietic differentiation, is necessary for the transformation process. Within that region, site-directed mutagenesis showed that the Y112 residue, which is required for Shc phosphorylation, is essential for rat fibroblast transformation by Mpl/MGDF, suggesting the involvement of Shc in Mpl-mediated transformation. Interestingly, we showed that transformation correlated with strong and sustained MAPK activation. Neither Jak2, Stat3 nor Stat5 phosphorylation was sufficient to induce the transformation process. Taken altogether, our results suggest the oncogenicity of Mpl in fibroblastic cells in the presence of its ligand.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Cell Transformation, Neoplastic , Milk Proteins , Neoplasm Proteins , Proto-Oncogene Proteins/metabolism , Receptors, Cytokine , 3T3 Cells/metabolism , 3T3 Cells/virology , Animals , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Genetic Vectors/genetics , Janus Kinase 2 , Leukemia Virus, Murine/genetics , Ligands , MAP Kinase Signaling System , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Mutation , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Proto-Oncogene Proteins/genetics , Rats , Rats, Inbred Strains , Receptors, Thrombopoietin , STAT3 Transcription Factor , STAT5 Transcription Factor , Shc Signaling Adaptor Proteins , Signal Transduction , Src Homology 2 Domain-Containing, Transforming Protein 1 , Thrombopoietin/metabolism , Thrombopoietin/pharmacology , Trans-Activators/metabolism
8.
Exp Hematol ; 28(12): 1363-72, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11146158

ABSTRACT

To examine whether the in vitro model of embryonic stem (ES) cell hematopoietic differentiation is suitable to study the function of intracytoplasmic regions of cytokine receptors, we used the thrombopoietin receptor Mpl as a typical cytokine receptor.ES cells deficient in c-mpl (mpl(-/)-) were transfected with genes encoding the full-length or two mutated forms of the intracytoplasmic domain of Mpl using the pEF-BOS expression vector. The mutated forms lack box1 or box2.pEF-BOS was able to maintain protein production during ES cell differentiation. Reintroduction of full-length-c-mpl into mpl(-/)- ES cells restored the response of megakaryocyte progenitors to a truncated form of human Mpl-ligand conjugated to polyethylene glycol (PEG-rhuMGDF) and the formation of platelets, for which mpl(-/)- ES cells are defective. In addition, enforced expression of Mpl resulted in the development of all myeloid progenitors and mature cells in the presence of PEG-rhuMGDF. Blast colony-forming cells, the in vitro equivalent of the hemangioblast, also generated blast cell colonies with a hematopoietic potential equivalent to that of the wild type in the presence of PEG-rhuMGDF, although its growth is normally dependent on vascular endothelial cell growth factor (VEGF). Thus, Mpl acts as a substitute for other cytokine receptors and for a tyrosine kinase receptor, Flk-1, indicating that Mpl has no instructive role in hematopoietic cell commitment and differentiation. The Mpl mutant forms lacking box1 or box2 prevented response of ES cell-derived blast colony-forming cells or progenitors to PEG-rhuMGDF. Therefore, these two regions, essential for signaling by cytokine receptors, are required for the responses of ES cell-derived hematopoietic cells to PEG-rhuMGDF.These results show that the in vitro hematopoietic differentiation of ES cells is suitable for studying the role of various intracytoplasmic regions of cytokine receptors.


Subject(s)
Cell Differentiation , Embryo, Mammalian , Hematopoietic Stem Cells/cytology , Neoplasm Proteins , Proto-Oncogene Proteins/physiology , Receptors, Cytokine , Animals , Cell Line , Cytoplasm/chemistry , DNA, Complementary/genetics , Flow Cytometry , Gene Expression , Genetic Vectors , Growth Substances/pharmacology , Humans , Megakaryocytes/cytology , Mice , Mutagenesis, Site-Directed , Polyethylene Glycols , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Receptors, Thrombopoietin , Recombinant Proteins/pharmacology , Structure-Activity Relationship , Transfection
9.
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
10.
Mol Cell Biol ; 17(9): 4991-5000, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9271377

ABSTRACT

Thrombopoietin (TPO) is the major regulator of both growth and differentiation of megakaryocytes. We previously showed that both functions can be generated by TPO in the megakaryoblastic cell line UT7, in which murine Mpl was introduced, and are independently controlled by distinct regions of the cytoplasmic domain of Mpl. Particularly, residues 71 to 94 of this domain (deleted in the mutant mpl delta3) were found to be required for megakaryocytic maturation but dispensable for proliferation. We show here that TPO-induced differentiation in UT7 cells is tightly dependent on a strong, long-lasting activation of the mitogen-activated protein kinase (MAPK) pathway. Indeed, (i) in UT7-mpl cells, TPO induced a strong activation of extracellular signal-regulated kinases (ERK) which was persistent until at least 4 days in TPO-containing medium; (ii) a specific MAPK kinase (MEK) inhibitor inhibited TPO-induced megakaryocytic gene expression; (iii) the Mpl mutant mpl delta3, which displayed no maturation activity, transduced only a weak and transient ERK activation in UT7 cells; and (iv) TPO-induced megakaryocytic differentiation in UT7-mpl delta3 cells was partially restored by expression of a constitutively activated mutant of MEK. The capacity of TPO to trigger a strong and prolonged MAPK signal depended on the cell in which Mpl was introduced. In BAF3-mpl cells, TPO triggered a weak and transient ERK activation, similar to that induced in UT7-mpl delta3 cells. In these cells, no difference in MAPK activation was found between normal Mpl and mpl delta3. Thus, depending on the cellular context, several distinct regions of the cytoplasmic domain of Mpl and signaling pathways may contribute to generate quantitative variations in MAPK activation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Megakaryocytes/cytology , Neoplasm Proteins , Proto-Oncogene Proteins/metabolism , Receptors, Cytokine/metabolism , Receptors, Immunologic/metabolism , Thrombopoietin/pharmacology , Animals , Cell Differentiation/drug effects , Cell Line , Enzyme Activation , Megakaryocytes/drug effects , Mice , Mutagenesis, Site-Directed , Phosphorylation , Proto-Oncogene Proteins/genetics , Receptors, Cytokine/genetics , Receptors, Immunologic/genetics , Receptors, Thrombopoietin , Signal Transduction , src Homology Domains
11.
J Biol Chem ; 272(39): 24300-7, 1997 Sep 26.
Article in English | MEDLINE | ID: mdl-9305885

ABSTRACT

Thrombopoietin (TPO) is the major regulator of proliferation and differentiation of megakaryocytes and their progenitors. These actions can be reproduced in the human megakaryoblastic cell line UT7 into which the murine TPO receptor, c-Mpl, was introduced. In these cells, TPO enhanced the expression of the specific megakaryocytic marker integrin glycoprotein (GP) IIb-IIIa while decreasing the expression of erythroid genes (Porteu, F., Rouyez, M. -C., Cocault, L., Benit, L., Charon, M., Picard, F., Gisselbrecht, S. , Souyri, M., and Dusanter-Fourt, I. (1996) Mol. Cell. Biol. 16, 2473-2482). We have now analyzed the effect of TPO on the transcriptional activity of the GPIIb promoter in these cells. Using transient transfection assays of a series of human GPIIb promoter fragments, we delineated a TPO-responsive element within the previously reported enhancer region of the promoter. Although this enhancer included GATA- and Ets-binding sites (EBSs), we found that only EBS -514 was important for TPO response. We identified PU. 1/Spi-1 as the endogenous Ets transcription factor that strongly and preferentially interacted with this enhancer EBS. This factor did not interact with other proximal EBSs in the GPIIb promoter. We next showed that TPO induced a strong and selective increase of PU. 1/Spi-1 expression and DNA binding activity in UT7-Mpl cells. In contrast, TPO did not affect the expression of Ets-1/2 while weakly increasing the levels of Fli-1. Overexpression of PU.1/Spi-1 was further shown to enhance GPIIb promoter activity in the absence and presence of TPO. Overall, our data indicated that, in UT7-Mpl cells, TPO increased the transcriptional activity of a GPIIb gene in part due to an enhanced expression of an unexpected transcription factor, the Ets family PU.1/Spi-1 factor. To our knowledge, this is the first evidence of a role for the PU.1/Spi-1 factor in the regulation of megakaryocytic genes.


Subject(s)
Gene Expression Regulation/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Proto-Oncogene Proteins/metabolism , Thrombopoietin/physiology , Trans-Activators/metabolism , Binding Sites , Cell Line , Humans , Promoter Regions, Genetic , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Proteins c-ets , Thrombopoietin/metabolism , Transcription Factors/metabolism
12.
Mol Cell Biol ; 16(5): 2473-82, 1996 May.
Article in English | MEDLINE | ID: mdl-8628315

ABSTRACT

Thrombopoietin (TPO) is the major regulator of growth and differentiation of megakaryocytes. To identify functionally important regions in the cytoplasmic domain of the TPO receptor, mpl, we introduced wild-type mpl and deletion mutants of murine mpl into the granulocyte-macrophage colony-stimulating factor (GM-CSF)- or erythropoietin (EPO)-dependent human cell line UT7. TPO induced differentiation of UT7-Wtmpl cells, not parental UT7 cells, along the megakaryocytic lineage, as evidenced by decreased proliferation, changes in cell morphology, and increased surface expression and mRNA levels of megakaryocytic markers CD41, CD61, and CD42b. When UT7-mpl cells were cultured long-term in EPO instead of GM-CSF, the TPO effect was dominant over that of EPO. Moreover, the differentiation induced by TPO was more pronounced for cells shifted from EPO to TPO than for cells shifted from GM-CSF to TPO, as shown by the appearance of polyploid cells. Mutational analysis of the cytoplasmic domain of mpl showed that proliferation and maturation functions of mpl can be uncoupled. Two functional regions were identified: (i) the first 69 amino acids comprising the cytokine receptor motifs, box I and box 2, which are necessary for both TPO-induced mitogenesis and maturation; and (ii) amino acids 71 to 94, which are dispensable for proliferation but required for differentiation. Surprisingly, however, EPO could complement this latter domain for TPO-induced differentiation, suggesting a close relationship between EPO and TPO signaling.


Subject(s)
Cell Differentiation , Erythropoietin/pharmacology , Neoplasm Proteins , Proto-Oncogene Proteins/physiology , Receptors, Cytokine , Thrombopoietin/pharmacology , Animals , Biomarkers , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Kinetics , Megakaryocytes/cytology , Megakaryocytes/drug effects , Mice , Mutagenesis , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/chemistry , Receptors, Thrombopoietin , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Deletion , Transfection
13.
Clin Exp Immunol ; 101(1): 25-32, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7621589

ABSTRACT

In vitro analysis of polymorphonuclear neutrophils (PMN) has allowed various stages of cell activation to be distinguished, characterized by the expression level of specific membrane markers and of functional receptors. Among those, TNF-alpha receptors (TNF-R) are modulated by various PMN activators, a mechanism which may be important to control cell responses to TNF in inflammatory reactions such as rheumatoid arthritis (RA). PMN, isolated from the blood of 36 RA patients and from the synovial fluid of 23 of them, were analysed for membrane expression of the two TNF-R (p55 and p75). Soluble p55 and p75 (sTNF-R) and TNF concentrations were measured in the plasma and synovial fluid by specific ELISA assays. Our results show that PMN from the blood of RA patients bear a normal number of TNF-R, with a normal p55/p75 ratio, compared with PMN from normal controls. Soluble TNF-R levels were similar in patients and normal plasma. In spite of high endogenous TNF concentration, patients' circulating PMN were not activated, as shown by a CD11b/CD18 expression similar to that of control resting cells. In contrast with blood neutrophils, PMN from RA patients' synovial fluids had an activated phenotype, characterized by increased expression of CD11b, decreased expression of leukosialin, CD43, and the appearance on the plasma membrane of an azurophil granule protein, CD63. High levels of soluble TNF-R were measured in RA synovial fluids. Nevertheless, membrane TNF-R levels and p55 and p75 proportions were similar to those of PMN from normal blood. These results suggest the existence of regulatory mechanisms which maintain a stable neutrophil expression of TNF-R as well as a balance between both types of receptors in inflammatory situations where neutrophils are strongly activated.


Subject(s)
Antigens, CD/biosynthesis , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/metabolism , Neutrophils/metabolism , Receptors, Tumor Necrosis Factor/biosynthesis , Antigens, CD/blood , Humans , Leukosialin , Macrophage-1 Antigen/biosynthesis , Macrophage-1 Antigen/blood , Neutrophil Activation/physiology , Platelet Membrane Glycoproteins/biosynthesis , Platelet Membrane Glycoproteins/blood , Receptors, Tumor Necrosis Factor/analysis , Sialoglycoproteins/biosynthesis , Sialoglycoproteins/blood , Synovial Fluid/chemistry , Tetraspanin 30
14.
Blood ; 86(2): 598-606, 1995 Jul 15.
Article in English | MEDLINE | ID: mdl-7541671

ABSTRACT

The erythropoietin (Epo) receptor belongs to the cytokine receptor superfamily. Although the cytokine receptors do not possess a tyrosine kinase consensus sequence in the intracellular domain, rapid stimulation of a tyrosine kinase activity occurs after activation by the ligand. We and others have shown that Epo induces the tyrosine phosphorylation of its cognate receptor as well as phosphorylation of other proteins. In this report, we examined the role of the receptor tyrosine residues in signal transduction. Eight tyrosine residues are located within the intracellular domain of the murine Epo receptor. A single tyrosine residue is present in the region previously shown to be sufficient for proliferative signal transduction. This tyrosine (Tyr 343) was mutated to phenylalanine. Moreover, mutant receptors were also generated with either a tyrosine residue or a phenylalanine residue at position 343 and with a COOH terminal truncation that removed the 7 other tyrosine residues. Expression vectors carrying these mutated receptors were transfected into the interleukin-3-dependent murine cell line Ba/F3. Epo-induced growth was sustained efficiently by all these receptors, although receptors without any tyrosine residues conferred a significantly reduced mitogenic activity. Moreover, all receptors were able to mediate Epo-dependant accumulation of beta-globin mRNA. The mutated receptors all induced the tyrosine phosphorylation of several cellular proteins after Epo stimulation. However, the truncated receptors induced the phosphorylation of a reduced number of proteins, suggesting that phosphorylated tyrosines of the receptor could have a role in the recruitment either of a tyrosine kinase or of tyrosine kinase substrate proteins. The receptors were all able to mediate Epo-induced activation of phosphatidylinositol 3-kinase, although truncated receptors no longer bound phosphatidylinositol 3-kinase.


Subject(s)
Erythropoiesis/physiology , Erythropoietin/physiology , Protein Processing, Post-Translational , Protein-Tyrosine Kinases/physiology , Receptors, Erythropoietin/metabolism , Signal Transduction/physiology , Tyrosine/metabolism , Cell Differentiation/drug effects , Cell Line , Enzyme Activation/drug effects , Erythropoiesis/drug effects , Gene Expression Regulation/drug effects , Globins/biosynthesis , Globins/genetics , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Interleukin-3/pharmacology , Mutagenesis, Site-Directed , Phosphatidylinositol 3-Kinases , Phosphorylation/drug effects , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotyrosine , Protein Processing, Post-Translational/drug effects , Receptors, Erythropoietin/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Signal Transduction/drug effects , Transfection , Tyrosine/analogs & derivatives
15.
J Biol Chem ; 269(4): 2834-40, 1994 Jan 28.
Article in English | MEDLINE | ID: mdl-8300617

ABSTRACT

The effect of tumor necrosis factor alpha (TNF) on the expression of its specific receptors (p55 TNF-R and p75 TNF-R) on the surface of human neutrophils (PMN) and mononuclear cells (MNC) was investigated and compared to the effect of various agonists. PMN and MNC express both p55 and p75 TNF-R on their membranes. Within minutes of incubation with chemotactic factors or calcium ionophore A23187, both types of TNF-R were down-regulated from the surface on both cell populations. At the same time, soluble forms of these TNF-R appeared in supernatants, in amounts proportional to the extent of down-regulation induced by each stimulus, suggesting that shedding is the major mechanism leading to loss of p55 and p75 TNF-R upon activation with these agonists. Likewise, TNF induced 60-80% and 73-90% decreases in PMN surface p55 TNF-R and p75 TNF-R, respectively. However, modulation of the two types of TNF-R by TNF proceeded through different mechanisms. TNF induced a selective shedding of the p75 TNF-R since, by both enzyme-linked immunosorbent assay and Western blot analysis, only the p75 TNF-R was detected in supernatants of cells stimulated with TNF. Down-modulation of surface p55 TNF-R most probably resulted from TNF-induced receptor internalization, since 125I-TNF bound to PMN p55 TNF-R was rapidly internalized with a t1/2 = 5 min and preincubation of PMN with TNF inhibited by 68 +/- 6% the release of p55 TNF-R triggered upon subsequent treatment with A23187. The apparently unique property of TNF to induce a differential modulation of the two types of TNF-R at the surface of PMN and MNC might play an important role in the control of peripheral blood cell responses to TNF.


Subject(s)
Neutrophils/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Adult , Calcimycin/pharmacology , Complement C5a/pharmacology , Down-Regulation , Humans , In Vitro Techniques , Kinetics , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Molecular Weight , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Receptors, Tumor Necrosis Factor/drug effects , Receptors, Tumor Necrosis Factor/isolation & purification , Recombinant Proteins/pharmacology
16.
Eur J Immunol ; 22(11): 3021-6, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1358626

ABSTRACT

Leukosialin (CD43) is a sialic acid-rich molecule with a relative molecular mass (M(r)) of 140,000 highly represented on polymorphonuclear neutrophils (PMN) and on most leukocytes. One of its functions may be to prevent nonspecific cell-to-cell interactions through negative charge repulsions. As tested by immunofluorescence, neutrophil CD43 membrane expression was shown to decrease by up to 80% upon cell activation by phorbol myristate acetate (10 ng/ml) or by N-formyl-L-methionyl-L-leucyl-L-phenylalanine (FMLP; 10(-6) M) in the presence of cytochalasin B. The kinetic of this decrease paralleled that of CD11b up-regulation. FMLP alone, tumor necrosis factor (TNF-alpha), lipopolysaccharide and granulocyte macrophage colony-stimulating factor had moderate or insignificant effects, while inducing striking CD11b up-regulation. Cell priming with TNF-alpha followed by FMLP stimulation resulted in up to 40% decrease of CD43 expression. Anti-CD43 mAb immunoprecipitated three fragments of M(r) 130,000, 49,000 and 34,000 from the cell-free supernatant of activated neutrophils, suggesting that CD43 is released from the membrane by proteolysis. Indeed, the decrease in CD43 expression was inhibited by phenylmethanesulfonylfluoride (PMSF). Homotypic aggregation of activated PMN was also inhibited by PMSF and could result, at least in part, from the shedding of CD43. The shedding of such a strongly anionic and major membrane protein should drastically modify PMN surface charge and may allow previously hindered interactions by exposing new adhesion molecules.


Subject(s)
Neutrophils/physiology , Sialoglycoproteins/physiology , Animals , Antigens, CD/analysis , CD11 Antigens , CD18 Antigens , Cell Aggregation , Cell Communication , Cells, Cultured , Down-Regulation , Humans , Leukosialin , Mice , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Sialoglycoproteins/analysis , Tetradecanoylphorbol Acetate/pharmacology
18.
J Leukoc Biol ; 52(1): 122-4, 1992 Jul.
Article in English | MEDLINE | ID: mdl-1322441

ABSTRACT

The distribution of type I (p55) and type II (p75) tumor necrosis factor receptors (TNF-Rs) in human polymorphonuclear neutrophils (PMNs) was analyzed by Western blotting of subcellular fractions obtained by centrifugation of PMN cavitates on Percoll density gradients. In resting PMNs, the p55 receptor was associated with both gamma and beta fractions, enriched in plasma membranes and specific granules, respectively, whereas the p75 TNF-R was located only in the gamma fraction. Intracellular p55 TNF-R bound 125I-labeled TNF in a ligand blot assay and migrated as a diffuse band of 46-60 kd, similar to the plasma membrane receptor. Activation of PMNs with the chemoattractant N-formyl-methionyl-leucyl-phenylalanine (fMLP), in conditions that induced the selective release of the tertiary granule marker gelatinase, caused the shedding of most of membrane p55 TNF-R as a 28-kd soluble form but had little effect on the distribution of the receptors in beta fractions. By contrast, when specific granule secretion was induced in the presence of cytochalasin B, a marked decrease in reactivity of beta fractions with anti-p55 TNF-R antibody was observed. At the same time, the amount of soluble 28-kd fragment found in the supernatant was increased. Thus, the intracellular pool of p55 TNF-R is associated with functional secretory granules and is redistributed in response to PMN activation.


Subject(s)
Neutrophils/ultrastructure , Receptors, Cell Surface/analysis , Blotting, Western , Humans , Intracellular Fluid/chemistry , Receptors, Tumor Necrosis Factor
19.
J Biol Chem ; 266(28): 18846-53, 1991 Oct 05.
Article in English | MEDLINE | ID: mdl-1655765

ABSTRACT

To localize the protease(s) involved in shedding of tumor necrosis factor receptors (TNF-R) from activated neutrophils (PMN) (Porteu, F., and C. Nathan (1990) J. Exp. Med. 172, 599-607), we tested subcellular fractions from PMN for their ability to cause loss of TNF-R from intact cells. Exposure of PMN to sonicated azurophil granules at 37 degrees C resulted in inhibition of 125I-TNF binding; 50% inhibition ensued when PMN were treated for approximately 1 min with azurophil granules equivalent to 2-3 PMN per indicator cell. The TNF-R-degrading activity in azurophil granules were identified as elastase by its sensitivity to diisopropyl fluorophosphate (DFP), alpha 1-antitrypsin and N-methoxysuccinyl-Ala-Ala-Pro-Val chloromethyl ketone (MSAAPV-CK), and by the ability of purified elastase to reproduce the effect of azurophil granules. Elastase preferentially acted on the 75-kDa TNF-R, reducing by 85-96% the binding of 125I-TNF to mononuclear cells expressing predominantly this receptor, while having no effect on endothelial cells expressing almost exclusively the 55-kDa TNF-R. Elastase-treated PMN released a 32-kDa soluble fragment of p75 TNF-R that bound TNF and reacted with anti-TNF-R monoclonal antibodies. In contrast, fMet-Leu-Phe-activated PMN shed a 42-kDa fragment from p75 TNF-R, along with similar amounts of a 28-kDa fragment from p55 TNF-R. Shedding of both TNF-Rs by intact activated PMN was more extensive than shedding caused by elastase and was completely resistant to DFP and MSAAPV-CK. Thus, the TNF-R-releasing activity of azurophil granules is distinct from that operative in intact stimulated PMN and could provide an additional mechanism for the control of cellular responses to TNF at sites of inflammation.


Subject(s)
Neutrophils/enzymology , Pancreatic Elastase/metabolism , Receptors, Cell Surface/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adult , Cell Fractionation , Cytoplasmic Granules/metabolism , Down-Regulation , Endopeptidases , Humans , Kinetics , Ligands , Lymphocyte Activation , Neutrophils/immunology , Neutrophils/ultrastructure , Receptors, Tumor Necrosis Factor
20.
Eur J Immunol ; 20(12): 2643-50, 1990 Dec.
Article in English | MEDLINE | ID: mdl-1702720

ABSTRACT

Membrane regulatory molecules normally prevent complement activation by autologous cells, therefore we compared the membrane control system of human lymphoid cell lines which activate or not human complement through the alternative pathway (AP). Membrane expression of decay-accelerating factor (DAF), membrane cofactor protein (MCP), complement receptors (CR)1, CR2 and H was measured either by radioimmunoassay or enzyme-linked immunosorbent assay on cell lysates. Soluble extracts of isolated membranes were tested functionally for their ability to accelerate the decay of C3bBb C3-convertase and allow the cleavage of C3b by factor I. Both regulatory functions were detected in solubilized membranes of Ramos cells, which do not activate the AP, as well as on the potent AP activator, Raji. Raji cells were found to express CR2, DAF and MCP molecules, while MCP was the only known regulatory protein detected on Ramos cells which expressed neither CR1, nor CR2, H or DAF. The I-cofactor activity of both Raji and Ramos cells was immunoprecipitated by anti-MCP, but the decay-accelerating activity was not adsorbed by anti-DAF nor by any of the available antibodies. Two EBV genome-negative cell lines (BJAB, BL41) were tested before and after in vitro conversion by EBV. As previously shown, EBV-converted cell lines activate the AP more efficiently than EBV- cell lines. At the same time, EBV superinfection induces an increase of both AP regulatory functions of cell membranes and enhances the expression of DAF, MCP and CR2. The results of this study show that complement activation by lymphoid cell lines is not related to an impaired autologous control of these cells, but that the expression of regulatory molecules increases together with the appearance of activating structures on the cell surface. Our results also suggest the occurrence of a new factor involved in the decay-accelerating activity on BL lines.


Subject(s)
Antigens, CD , B-Lymphocytes/physiology , Complement Pathway, Alternative , Antigens, Differentiation, B-Lymphocyte/physiology , CD55 Antigens , Cell Line , Cell Membrane/physiology , Complement C3-C5 Convertases/metabolism , Complement C3b/metabolism , Complement C3b Inactivator Proteins/physiology , Complement Factor H , Complement Factor I , Herpesvirus 4, Human/immunology , Humans , In Vitro Techniques , Membrane Cofactor Protein , Membrane Glycoproteins/metabolism , Membrane Proteins/physiology , Receptors, Complement/physiology , Receptors, Complement 3b , Receptors, Complement 3d , Serine Endopeptidases/metabolism , Tumor Virus Infections/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...