Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 98(13): 7487-91, 2001 Jun 19.
Article in English | MEDLINE | ID: mdl-11416218

ABSTRACT

A major therapeutic target in the search for a cure to the devastating Alzheimer's disease is gamma-secretase. This activity resides in a multiprotein enzyme complex responsible for the generation of Abeta42 peptides, precipitates of which are thought to cause the disease. Gamma-secretase is also a critical component of the Notch signal transduction pathway; Notch signals regulate development and differentiation of adult self-renewing cells. This has led to the hypothesis that therapeutic inhibition of gamma-secretase may interfere with Notch-related processes in adults, most alarmingly in hematopoiesis. Here, we show that application of gamma-secretase inhibitors to fetal thymus organ cultures interferes with T cell development in a manner consistent with loss or reduction of Notch1 function. Progression from an immature CD4-/CD8- state to an intermediate CD4+/CD8+ double-positive state was repressed. Furthermore, treatment beginning later at the double-positive stage specifically inhibited CD8+ single-positive maturation but did not affect CD4+ single-positive cells. These results demonstrate that pharmacological gamma-secretase inhibition recapitulates Notch1 loss in a vertebrate tissue and present a system in which rapid evaluation of gamma-secretase-targeted pharmaceuticals for their ability to inhibit Notch activity can be performed in a relevant context.


Subject(s)
Endopeptidases/metabolism , Protease Inhibitors/pharmacology , Receptors, Cell Surface , T-Lymphocytes/physiology , Transcription Factors , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Cell Line , Cells, Cultured , Humans , Kidney , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mutagenesis , Organ Culture Techniques , Receptor, Notch1 , Recombinant Proteins/metabolism , Sequence Deletion , Thymus Gland/embryology , Thymus Gland/immunology , Transfection
2.
Mol Biol Cell ; 11(10): 3299-313, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11029037

ABSTRACT

Cellular adhesive events affect cell proliferation and differentiation decisions. How cell surface events mediating adhesion transduce signals to the nucleus is not well understood. After cell-cell or cell-substratum contact, cytosolic proteins are recruited to clustered adhesion receptor complexes. One such family of cytosolic proteins found at sites of cell adhesion is the Zyxin family of LIM proteins. Here we demonstrate that the family member Ajuba was recruited to the cell surface of embryonal cells, upon aggregate formation, at sites of cell-cell contact. Ajuba contained a functional nuclear export signal and shuttled into the nucleus. Importantly, accumulation of the LIM domains of Ajuba in the nucleus of P19 embryonal cells resulted in growth inhibition and spontaneous endodermal differentiation. The differentiating effect of Ajuba mapped to the third LIM domain, whereas regulation of proliferation mapped to the first and second LIM domains. Ajuba-induced endodermal differentiation of these cells correlated with the capacity to activate c-Jun kinase and required c-Jun kinase activation. These results suggest that the cytosolic LIM protein Ajuba may provide a new mechanism to transduce signals from sites of cell adhesion to the nucleus, regulating cell growth and differentiation decisions during early development.


Subject(s)
Cell Division/physiology , Cell Nucleus/physiology , Endoderm/cytology , Homeodomain Proteins/physiology , 3T3 Cells , Animals , Cell Adhesion , Cloning, Molecular , Cytosol/physiology , Embryo, Mammalian , Endoderm/physiology , LIM Domain Proteins , Mice , Recombinant Proteins/metabolism , Signal Transduction , Teratoma , Transfection , Tumor Cells, Cultured , Zinc Fingers
3.
Int J Hematol ; 72(2): 178-85, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11039666

ABSTRACT

Erythropoiesis is severely impaired in mice with inactivating mutations in the Steel factor (SF) gene (Sl/Sl mice) or in c-kit, in the SF receptor gene (W/W mice), and in mice with null mutations in the genes for either erythropoietin (EPO) or the erythropoietin receptor (EPO-R). Previous studies indicated that EPO is sufficient for colony development from colony-forming units-erythroid (CFU-E). However, recent studies have shown that there is a physical association between these 2 receptors and that c-kit can phosphorylate EPO-R. To examine the role SF and EPO play in regulating erythropoiesis, we examined the effect of SF and EPO on colony development from cells of the embryonic aorta-gonad-mesonephros (AGM) region, yolk sac, and liver of fetal wild-type and W/W mice. The maturation of CFU-E from these sites did not require the addition of SF to clonal cultures, whereas the efficient development of erythroid bursts required both EPO and SE The number of erythroid colony-forming cells was reduced in both the AGM region and liver of fetal W/W mice. The residual CFU-E present in W/W mice were dependent on EPO and independent of SF. These results indicate that EPO/EPO-R can function to support colony formation in the absence of an SF signal.


Subject(s)
Erythroid Precursor Cells/drug effects , Erythropoietin/pharmacology , Animals , Cell Culture Techniques , Embryo, Mammalian/metabolism , Embryo, Mammalian/physiology , Erythropoiesis/drug effects , Erythropoietin/physiology , Female , Mice , Mice, Mutant Strains , Pregnancy , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/pharmacology , Proto-Oncogene Proteins c-kit/physiology , Signal Transduction/drug effects , Stem Cell Factor/genetics , Stem Cell Factor/pharmacology , Stem Cell Factor/physiology
4.
J Biol Chem ; 275(34): 26566-75, 2000 Aug 25.
Article in English | MEDLINE | ID: mdl-10858439

ABSTRACT

Hematopoietic cell development and function is dependent on cytokines and on intercellular interactions with the microenvironment. Although the intracellular signaling pathways stimulated by cytokine receptors are well described, little is known about the mechanisms through which these pathways modulate hematopoietic cell adhesion events in the microenvironment. Here we show that cytokine-activated Stat3 stimulates the expression and function of cell surface adhesion molecules in the myeloid progenitor cell line 32D. We generated an erythropoietin receptor (EpoR) isoform (ER343/401-S3) that activates Stat3 rather than Stat5 by substituting the Stat3 binding/activation sequence motif from gp130 for the sequences surrounding tyrosines 343 and 401 in the receptor cytoplasmic region. Activation of Stat3 leads to homotypic cell aggregation, increased expression of intercellular adhesion molecule 1 (ICAM-1), CD18, and CD11b, and activation of signaling through CD18-containing integrins. Unlike the wild type EpoR, ER343/401-S3 is unable to support long term Epo-dependent proliferation in 32D cells. Instead, Epo-treated ER343/401-S3 cells undergo G(1) arrest and express elevated levels of the cyclin-dependent kinase inhibitor p27(Kip1). Sustained activation of Stat3 in these cells is required for their altered morphology and growth properties since constitutive SOCS3 expression abrogates homotypic cell aggregation, signaling through CD18-containing integrins, G(1) arrest, and accumulation of p27(Kip1). Collectively, our results demonstrate that cytokine-activated Stat3 stimulates the expression and function of cell surface adhesion molecules, indicating that a role for Stat3 is to regulate intercellular contacts in myeloid cells.


Subject(s)
Cell Cycle Proteins , Cytokines/physiology , DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/cytology , Integrins/metabolism , Trans-Activators/metabolism , Tumor Suppressor Proteins , Animals , CD18 Antigens/biosynthesis , Cell Adhesion , Cyclin-Dependent Kinase Inhibitor p27 , Enzyme Activation , G1 Phase , Granulocyte Colony-Stimulating Factor/pharmacology , Intercellular Adhesion Molecule-1/biosynthesis , Macrophage-1 Antigen/biosynthesis , Mice , Microtubule-Associated Proteins/metabolism , Receptors, Erythropoietin/metabolism , STAT3 Transcription Factor , Signal Transduction
5.
J Interferon Cytokine Res ; 20(12): 1065-70, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11152572

ABSTRACT

Erythropoietin (Epo) is essential for formation of mature red blood cells (RBC). However, the function of Epo receptor (EpoR)-dependent signaling pathways in the regulation of erythropoiesis remains unclear. To determine whether specific Stat signals are required for RBC development, we changed the Stat signaling specificity of the EpoR. The wild-type EpoR activates only Stat5. Thus, we substituted the major Stat5 binding sites (residues 343 and 401) in the EpoR cytoplasmic region with the Stat3 binding/activation motif from gp130. We demonstrated that activated EpoRs containing a single substitution stimulate Stat5 and Stat3, whereas an EpoR with both substitutions stimulates Stat3 but not Stat5. We then determined the ability of these receptors to support fetal liver and adult erythropoiesis. Our results show that erythropoiesis is stimulated by EpoRs that activate Stat5, both Stat5 and Stat3, or Stat3 in place of Stat5. These findings demonstrate that the specificity of EpoR Stat signaling is not essential for RBC development.


Subject(s)
DNA-Binding Proteins/metabolism , Erythropoiesis/physiology , Liver/metabolism , Milk Proteins , Receptors, Erythropoietin/physiology , Trans-Activators/metabolism , Animals , Cell Line , Colony-Forming Units Assay , Fetus/metabolism , Liver/physiology , Mice , STAT3 Transcription Factor , STAT5 Transcription Factor , Signal Transduction
6.
Blood ; 94(8): 2667-75, 1999 Oct 15.
Article in English | MEDLINE | ID: mdl-10515870

ABSTRACT

Erythropoietin (EPO) is required for the survival and expansion of red blood cell progenitor cells and supports continued differentiation of these committed progenitors to mature red blood cells. After binding to its cognate receptor, EPO promotes receptor homodimerization, activation of receptor-associated JAK2, subsequent receptor tyrosine phosphorylation, and transduction of signal. EPO is also internalized and degraded in lysosomes. The contribution of EPO-induced receptor internalization to modulation of EPO signals has not been determined. To examine this question, we generated a panel of hematopoietic cell lines containing progressively truncated isoforms of the erythropoietin receptor (EPO-R) and determined the rate and extent of EPO internalization and receptor downregulation. We demonstrated that a membrane-proximal domain of the cytoplasmic tail of the EPO-R was the minimal region required for EPO-induced receptor internalization. This cytoplasmic domain is also the minimal domain required for activation of JAK2, a cytosolic tyrosine kinase essential for the function of the EPO-R. However, neither EPO activation of cytosolic JAK2 tyrosine kinase activity nor tyrosine phosphorylation of the EPO-R cytoplasmic tail was required for EPO-induced receptor downregulation. Both functional and nonfunctional cell surface receptor isoforms were internalized equally. These results suggest that, for downregulation of cell surface ligand occupied EPO-R and possibly for signaling receptors of the cytokine receptor superfamily in general, internalization of cell surface ligand occupied receptors may follow a pathway distinct from signaling receptors of the receptor tyrosine kinase (RTK) family.


Subject(s)
Endocytosis/physiology , Erythropoietin/metabolism , Protein Processing, Post-Translational , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins , Receptors, Erythropoietin/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Dimerization , Enzyme Activation , Hematopoietic Stem Cells/metabolism , Janus Kinase 2 , Mice , Multigene Family , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/genetics , Sequence Deletion , Structure-Activity Relationship
7.
Ann Med ; 31(3): 208-16, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10442676

ABSTRACT

The production of erythroid cells is a dynamic and exquisitely regulated process. The mature red cell is only the final phase of a complex but orderly series of genetic events that are initiated at the time a multipotent stem cell becomes committed to expressing the erythroid programme. Aberrations either in the intrinsic generation and/or amplification of functional erythroid cells or in the regulatory influences of microenvironment or cytokines form the basis for a number of blood diseases. In this review we focus upon abnormalities in red blood cell production and discuss how alterations in cytokine regulation of red blood cell production may contribute to these disease processes. We discuss clinical states in which blood red cell numbers are altered, including primary familial and congenital polycythaemia, the myeloproliferative disorder polycythaemia vera, erythroleukaemia, and Diamond-Blackfan anaemia. These disorders are briefly described and evidence supporting a potential role of specific cytokine receptor signalling defects as contributing to these phenotypes is discussed.


Subject(s)
Hematologic Diseases/genetics , Hematopoiesis/physiology , Receptors, Cytokine/physiology , Erythropoietin/genetics , Erythropoietin/physiology , Fanconi Anemia/genetics , Fanconi Anemia/physiopathology , Hematologic Diseases/blood , Hematologic Diseases/physiopathology , Hematopoiesis/genetics , Humans , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/physiopathology , Mutation , Polycythemia/blood , Polycythemia/congenital , Polycythemia/genetics , Polycythemia/physiopathology , Polycythemia Vera/blood , Polycythemia Vera/genetics , Polycythemia Vera/physiopathology , Receptors, Cytokine/genetics , Receptors, Erythropoietin/physiology , Signal Transduction
8.
Blood ; 94(1): 74-86, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10381500

ABSTRACT

Erythropoietin (EPO) and its receptor (EPOR) are required for the development of mature erythrocytes. After binding of ligand, the EPOR activates a variety of signaling pathways that ultimately control cellular proliferation, survival, and specific gene expression. Although erythroid progenitors appear to be the principal EPO-responsive cell type in vivo due to the restricted expression of the EPOR, many growth factor-dependent cell lines expressing the EPOR can respond to EPO by activating many or all of these pathways. In the present study, we have identified a cellular context (the interleukin-2 [IL-2]-dependent HT-2 line) in which the EPO stimulation of the EPOR fails to support cellular proliferation, STAT-5 induction, or MAPK activation, despite efficient phosphorylation of the EPOR and JAK2 and inhibition of apoptosis after withdrawal of IL-2. Interestingly, when we fused HT-2 cells expressing the EPOR with Ba/F3 cells in a complementation assay, the resulting hybridomas proliferated and potently activated STAT-5 and MAPK in response to EPO. These data indicate that an unidentified cellular factor is needed to mediate signaling by the EPOR. Moreover, Ba/F3 cells apparently express this factor(s) and somatic fusions can, therefore, confer EPO-responsiveness to HT-2 cells that lack this factor.


Subject(s)
Erythrocytes/physiology , Erythropoietin/pharmacology , Milk Proteins , Receptors, Erythropoietin/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cell Line , DNA-Binding Proteins/physiology , Enzyme Activation , Erythropoietin/physiology , Humans , STAT5 Transcription Factor , Trans-Activators/physiology
9.
Mol Cell Biol ; 19(6): 4379-89, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10330178

ABSTRACT

LIM domain-containing proteins contribute to cell fate determination, the regulation of cell proliferation and differentiation, and remodeling of the cell cytoskeleton. These proteins can be found in the cell nucleus, cytoplasm, or both. Whether and how cytoplasmic LIM proteins contribute to the cellular response to extracellular stimuli is an area of active investigation. We have identified and characterized a new LIM protein, Ajuba. Although predominantly a cytosolic protein, in contrast to other like proteins, it did not localize to sites of cellular adhesion to extracellular matrix or interact with the actin cytoskeleton. Removal of the pre-LIM domain of Ajuba, including a putative nuclear export signal, led to an accumulation of the LIM domains in the cell nucleus. The pre-LIM domain contains two putative proline-rich SH3 recognition motifs. Ajuba specifically associated with Grb2 in vitro and in vivo. The interaction between these proteins was mediated by either SH3 domain of Grb2 and the N-terminal proline-rich pre-LIM domain of Ajuba. In fibroblasts expressing Ajuba mitogen-activated protein kinase activity persisted despite serum starvation and upon serum stimulation generated levels fivefold higher than that seen in control cells. Finally, when Ajuba was expressed in fully developed Xenopus oocytes, it promoted meiotic maturation in a Grb2- and Ras-dependent manner.


Subject(s)
Adaptor Proteins, Signal Transducing , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Oocytes/metabolism , Proteins/physiology , Xenopus/embryology , ras Proteins/physiology , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cloning, Molecular , Cytosol/metabolism , Fibroblasts/metabolism , Fluorescent Antibody Technique , GRB2 Adaptor Protein , Homeodomain Proteins/chemistry , Immunoblotting , LIM Domain Proteins , Meiosis , Mice , Microinjections , Molecular Sequence Data , Precipitin Tests , Proto-Oncogene Proteins c-myc/metabolism , Recombinant Fusion Proteins , Sequence Analysis, DNA , Stem Cells , Time Factors , Tissue Distribution , src Homology Domains/physiology
10.
J Biol Chem ; 274(9): 5415-21, 1999 Feb 26.
Article in English | MEDLINE | ID: mdl-10026152

ABSTRACT

Signal transduction by the erythropoietin receptor (EPOR) is activated by ligand-mediated receptor homodimerization. However, the relationship between extracellular and intracellular domain oligomerization remains poorly understood. To assess the requirements for dimerization of receptor cytoplasmic sequences for signaling, we overexpressed mutant EPORs in combination with wild-type (WT) EPOR to drive formation of heterodimeric (i.e. WT-mutant) receptor complexes. Dimerization of the membrane-proximal portion of the EPOR cytoplasmic region was found to be critical for the initiation of mitogenic signaling. However, dimerization of the entire EPOR cytoplasmic region was not required. To examine this process more closely, we generated chimeras between the intracellular and transmembrane portions of the EPOR and the extracellular domains of the interleukin-2 receptor beta and gammac chains. These chimeras allowed us to assess more precisely the signaling role of each receptor chain because only heterodimers of WT and mutant receptor chimeras form in the presence of interleukin-2. Coexpression studies demonstrated that a functional receptor complex requires the membrane-proximal region of each receptor subunit in the oligomer to permit activation of JAK2 but only one membrane-distal tail to activate STAT5 and to support cell proliferation. Thus, this study defines key relationships involved in the assembly and activation of the EPOR signal transduction complex which may be applicable to other homodimeric cytokine receptors.


Subject(s)
Cytoplasm/metabolism , Receptors, Erythropoietin/metabolism , Base Sequence , Biopolymers , Cell Division , Cell Line , Oligodeoxyribonucleotides , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/genetics , Signal Transduction
11.
Proc Natl Acad Sci U S A ; 95(12): 7006-11, 1998 Jun 09.
Article in English | MEDLINE | ID: mdl-9618529

ABSTRACT

Erythropoietin (EPO) is required for red blood cell development, but whether EPO-specific signals directly instruct erythroid differentiation is unknown. We used a dominant system in which constitutively active variants of the EPO receptor were introduced into erythroid progenitors in mice. Chimeric receptors were constructed by replacing the cytoplasmic tail of constitutively active variants of the EPO receptor with tails of diverse cytokine receptors. Receptors linked to granulocyte or platelet production supported complete erythroid development in vitro and in vivo, as did the growth hormone receptor, a nonhematopoietic receptor. Therefore, EPOR-specific signals are not required for terminal differentiation of erythrocytes. Furthermore, we found that cellular context can influence cytokine receptor signaling.


Subject(s)
Erythrocytes/physiology , Erythropoiesis/physiology , Erythropoietin/physiology , Receptors, Erythropoietin/physiology , 3T3 Cells , Animals , Cell Differentiation/physiology , Erythrocytes/cytology , Mice , Recombinant Fusion Proteins/physiology , Signal Transduction , Transfection
12.
J Immunol ; 160(7): 3502-12, 1998 Apr 01.
Article in English | MEDLINE | ID: mdl-9531312

ABSTRACT

Cytokines play an essential role in the regulation of lymphocyte survival and growth. We have analyzed the pathways activated by IE-2 that lead to protection from apoptosis and cell proliferation. IL-2 can act as a long-term growth factor in 32D cells expressing the wild-type human (hu)IL-2R beta. By contrast, cells expressing a truncated form of the huIL-2R beta, which is able to induce Bcl-2 and c-myc expression but not STAT5 activation, were not protected from apoptosis by IL-2; consequently, they could not be grown long term in the presence of IL-2. However, IL-2 promoted cell cycle progression in cells bearing the truncated huIL-2R beta with percentages of viable cells in the G0/G1, S, and G2/M phases similar to cells expressing the wild-type huIL-2R beta. Transplantation of a region from the erythropoietin receptor, which contains a docking site for STAT5 (Y343) to the truncated huIL-2R beta, restored the ability of IL-2 to signal both activation of STAT5 and protection from apoptosis. By contrast, transplantation of a region from the huIL-4R alpha containing STAT6 docking sites did not confer protection from apoptosis. These results indicate that the IL-2-induced cell cycle progression can be clearly distinguished from protection from apoptosis and that STAT5 participates in the regulation of apoptosis.


Subject(s)
Apoptosis/immunology , Cell Cycle/immunology , Interleukin-2/physiology , Protein Serine-Threonine Kinases/physiology , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Division/drug effects , Cell Division/genetics , Cell Division/immunology , Humans , Mice , Protein Serine-Threonine Kinases/metabolism , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/physiology , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Cells, Cultured
13.
Blood ; 91(3): 870-8, 1998 Feb 01.
Article in English | MEDLINE | ID: mdl-9446647

ABSTRACT

Cytokine receptors have been shown in cell culture systems to use phosphotyrosine residues as docking sites for certain signal transduction intermediates. Studies using various cellular backgrounds have yielded conflicting information about the importance of such residues. The present studies were undertaken to determine whether or not tyrosine residues within the erythropoietin receptor (EPOR) are essential for biologic activity during hematopoiesis in vivo. A variant of the EPOR was constructed that contains both a substitution (R129C) causing constitutive receptor activation as well as replacement of all eight cytoplasmic tyrosines by phenylalanines (cEPORYF). A comparison between animals exposed to recombinant retroviruses expressing cEPOR and cEPORYF showed that efficient red blood cell (RBC) development in vivo is dependent on the pressence of tyrosine residues in the cytoplasmic domain of the EPOR. In addition, an inefficient EPOR tyrosine independent pathway supporting RBC development was detected. Tyrosine add-back mutants showed that multiple individual tyrosines have the capacity to restore full erythropoietic potential to the EPOR as determined in whole animals. The analysis of primary erythroid progenitors transduced with the various cEPOR tyrosine mutants and tyrosine add-backs showed that only tyrosine 343 (Y1) and tyrosine 479 (Y8) were capable of supporting immature burst-forming unit-erythroid progenitor development. Thus, this receptor is characterized by striking functional redundancy of tyrosines in a biologically relevant context. However, selective tyrosine residues may be uniquely important for early signals supporting erythroid development.


Subject(s)
Erythropoiesis , Receptors, Erythropoietin/chemistry , Tyrosine/physiology , Animals , Cell Line , Erythropoietin/pharmacology , Gene Expression , Gene Transfer Techniques , Mice , Mutagenesis , Phenylalanine , Receptors, Erythropoietin/genetics , Recombinant Proteins , Retroviridae/genetics , Signal Transduction , Structure-Activity Relationship
14.
J Biol Chem ; 272(14): 9099-107, 1997 Apr 04.
Article in English | MEDLINE | ID: mdl-9083037

ABSTRACT

During erythroid development erythropoietin (EPO) binds specifically to a receptor primarily present on committed erythroid progenitors, stimulating mitogenic, survival, and differentiative growth response pathways. Other modes of erythropoietin receptor (EPO-R) activation, such as interaction with the env gene Friend virus envelope glycoprotein (F-gp55) of spleen focus-forming virus or specific mutations in the extracellular domain of the EPO-R, give rise to pathological consequences, in vivo and EPO-independent proliferation and differentiation of cultured cells. Activating extracellular receptor mutations result in covalently linked receptor homodimers. These observations and others have led to the proposal that EPO activates the EPO-R by inducing dimer formation on the cell surface. It has been assumed that F-gp55 also induces dimer formation of the EPO-R; however, clear evidence of this is lacking. In addition, EPO and F-gp55 stimulation of the EPO-R elicit different biological responses. To probe whether the cell surface EPO-R is structurally different with these activators, we contrasted the cell surface EPO-R complex formed following receptor activation by EPO, F-gp55, and mutations in the extracellular domain of the receptor. Our results indicate that cell surface forms of activated EPO-R differ, as judged by their differential association with F-gp55 and pattern of associated cell surface proteins. Interestingly, we find that the env gene of an anemic strain of Friend virus, Rauscher virus envelope glycoprotein, does not interact with the EPO-R at the cell surface. Thus, the mode of Rauscher virus envelope glycoprotein-induced erythroblastosis may be distinct from F-gp55-induced erythroblastosis and possibly not involve the EPO-R.


Subject(s)
Erythropoietin/metabolism , Receptors, Erythropoietin/metabolism , Viral Envelope Proteins/metabolism , Animals , Biotin/metabolism , Cell Line , Kinetics , Membrane Glycoproteins/metabolism , Molecular Weight , Point Mutation , Protein Conformation , Receptors, Erythropoietin/chemistry , Spleen Focus-Forming Viruses , Surface Properties
15.
Proc Natl Acad Sci U S A ; 93(1): 231-5, 1996 Jan 09.
Article in English | MEDLINE | ID: mdl-8552611

ABSTRACT

The specific signal transduction function of the gamma c subunit in the interleukin (IL) 2, IL-4, IL-7, IL-9, and IL-15 receptor complexes remains undefined. The present structure-function analyses demonstrated that the entire cytoplasmic tail of gamma c could be functionally replaced in the IL-2 receptor (IL-2R) signaling complex by a severely truncated erythropoietin receptor cytoplasmic domain lacking tyrosine residues. Heterodimerization of IL-2R beta with either gamma c or the truncated erythropoietin receptor chain led to an array of specific signals normally derived from the native IL-2R despite the substitution of Janus kinase JAK2 for JAK3 in the receptor complex. These findings thus suggest a model in which the gamma c subunit serves as a common and generic "trigger" chain by providing a nonspecific Janus kinase for signaling program initiation, while signal specificity is determined by the unique "driver" subunit in each of the gamma c- containing receptor complexes. Furthermore, these results may have important functional implications for the asymmetric design of many cytokine receptor complexes and the evolutionary design of receptor subfamilies that share common trigger or driver subunits.


Subject(s)
Lymphocyte Activation , Proto-Oncogene Proteins , Receptors, Cytokine/chemistry , Receptors, Interleukin-2/chemistry , Signal Transduction , Cells, Cultured , DNA-Binding Proteins/physiology , Gene Expression , Humans , Janus Kinase 2 , Janus Kinase 3 , Macromolecular Substances , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/genetics , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/metabolism , Receptors, Fc/genetics , Receptors, Interleukin-2/metabolism , STAT1 Transcription Factor , Structure-Activity Relationship , Trans-Activators/physiology
16.
J Biol Chem ; 270(37): 21729-37, 1995 Sep 15.
Article in English | MEDLINE | ID: mdl-7665592

ABSTRACT

To evaluate the possible role for receptor-based tyrosine phosphorylation in growth signaling induced by interleukin-2 (IL-2), a series of substitution tyrosine mutants of the IL-2 receptor beta and gamma c chains was prepared and analyzed. Concurrent mutation of all six of the cytoplasmic tyrosines present in the beta chain markedly inhibited IL-2-induced growth signaling in both pro-B and T cell lines. Growth signaling in a pro-B cell line was substantially reconstituted when either of the two distal tyrosines (Tyr-392, Tyr-510) was selectively restored in the tyrosine-negative beta mutant, whereas reconstitution of the proximal tyrosines (Tyr-338, Tyr-355, Tyr-358, Tyr-361) did not restore this signaling function. Furthermore, at least one of the two cytoplasmic tyrosines that is required for beta chain function was found to serve as a phosphate acceptor site upon induction with IL-2. Studies employing a chimeric receptor system revealed that tyrosine residues of the beta chain likewise were important for growth signaling in T cells. In contrast, although the gamma c subunits is a target for tyrosine phosphorylation in vivo, concurrent substitution of all four cytoplasmic tyrosines of this chain produced no significant effect on growth signaling by chimeric IL-2 receptors. However, deletion of either the Box 1, Box 2, or intervening (V-Box) regions of gamma c abrogated receptor function. Therefore, tyrosine residues of beta but not of gamma c appear to play a pivotal role in regulating growth signal transduction through the IL-2 receptor, either by influencing cytoplasmic domain folding or by serving as sites for phosphorylation and subsequent association with signaling intermediates. These findings thus highlight a fundamental difference in the structural requirements for IL-2R beta and gamma c in receptor-mediated signal transduction.


Subject(s)
Interleukin-2/pharmacology , Receptors, Interleukin-2/physiology , Signal Transduction , Tyrosine , Amino Acid Sequence , Animals , B-Lymphocytes , Cell Division , Cloning, Molecular , Humans , Interleukin-2/metabolism , Kinetics , Macromolecular Substances , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Receptors, Erythropoietin/chemistry , Receptors, Erythropoietin/metabolism , Receptors, Interleukin-2/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , T-Lymphocytes , Transfection
17.
Exp Hematol ; 23(7): 645-54, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7601257

ABSTRACT

The receptor for erythropoietin (EpoR) is normally restricted in its expression to the relatively mature cells of the erythroid and megakaryocytic lineages. Using retrovirus-mediated gene transfer, the wild-type EpoR and a constitutively activated mutant of the EpoR, EpoRR129C, were expressed in primary hematopoietic cells. Retroviral infection of day-12 murine fetal liver, followed by stimulation with Epo as a single stimulus, generated day-8 erythroid colonies resembling colonies derived from burst-forming units-erythroid (BFU-E). Similarly, murine post-5 fluorouracil (5-FU) bone marrow cells or fetal liver cells, induced to express EpoR and stimulated by Epo, displayed a significant enhancement of megakaryocyte colony formation, particularly of the BFU-megakaryocyte (BFU-Mk) colony type. Cultures of bone marrow cells transduced with the EpoR retrovirus and stimulated by Epo contained macrophage colonies but very few granulocyte colonies. Experiments to culture single clones demonstrated direct action of Epo on megakaryocyte and macrophage clones but failed to demonstrate a direct action on granulocyte precursors. A similar pattern of lineage-restricted effects was demonstrated in unstimulated cultures of cells infected with the EpoRR129C retrovirus. In summary, we have demonstrated Epo-induced recruitment of immature erythroid and megakaryocyte precursors induced to express the EpoR. Furthermore, we have also demonstrated lineage-restricted cell proliferation in response to Epo by normal myeloid hematopoietic cells transduced with the EpoR.


Subject(s)
Erythropoietin/metabolism , Hematopoietic Stem Cells/metabolism , Receptors, Erythropoietin/metabolism , Animals , Base Sequence , Cell Differentiation/drug effects , Colony-Forming Units Assay , Erythropoietin/genetics , Female , Gene Expression Regulation , Gene Transfer Techniques , Hematopoietic Stem Cells/cytology , Liver/embryology , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Molecular Sequence Data , Pregnancy , Receptors, Erythropoietin/genetics
18.
J Biol Chem ; 270(12): 6523-30, 1995 Mar 24.
Article in English | MEDLINE | ID: mdl-7896787

ABSTRACT

The JAK2 tyrosine kinase is known to associate with the receptors for growth hormone (GH) and erythropoietin (EPO) and with the interleukin-6 receptor signal transducing protein, gp130. Here we demonstrate that chimeric cytokine receptors which contain the cytoplasmic domain of the receptors for GH and EPO or for gp130 can form complexes with JAK2 when transiently co-expressed in HeLa cells. Mutational analyses of chimeras for the the GH and EPO receptors and gp130 demonstrated that box 1, a motif critical for cytokine receptor signal transduction, was required for the association of JAK2. Although JAK2 was capable of associating with all three of the chimeras, JAK1 co-precipitated only with the gp130 chimera. Association of JAK1 and JAK2 with cytokine receptor proteins, therefore, requires the highly conserved box 1 domain, but other sequences within the receptor proteins may influence the specificity of JAK binding. Mutational analysis of JAK2 revealed that multiple or complex protein sequences within JAK2 are required for association with cytokine receptors.


Subject(s)
Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins , Receptors, Cytokine/metabolism , Animals , Base Sequence , HeLa Cells , Humans , Janus Kinase 1 , Janus Kinase 2 , Mice , Molecular Sequence Data , Receptors, Erythropoietin/metabolism , Receptors, Somatotropin/metabolism , Recombinant Fusion Proteins/metabolism , Signal Transduction , Structure-Activity Relationship
19.
Proc Natl Acad Sci U S A ; 91(16): 7482-6, 1994 Aug 02.
Article in English | MEDLINE | ID: mdl-8052607

ABSTRACT

Whether the presence of specific receptors on the surface of developing cells is the cause or consequence of lineage restriction is not known. If activation of specific receptors is the driving event in differentiation, the premature expression of specific receptors would promote differentiation along that pathway. In this study pluripotent progenitors, obtained from blast cell colonies (pooled or individual) of 5-fluorouracil-treated mice, were infected with retroviral vectors containing either an activated receptor for erythropoietin (EPO), an erythroid progenitor growth factor, or the receptor for colony-stimulating factor 1 (CSF-1), a macrophage growth factor. These receptors exhibit expression patterns restricted to committed progenitors. The developmental potential of infected pluripotent progenitors was not changed, although they expressed the exogenous genes, suggesting that in these cells activation of lineage-specific receptors does not induce differentiation. Acquisition of a constitutively activated EPO receptor allowed erythroid development in mixed colonies in the absence of EPO, as expected. Infection of progenitors with a virus containing the CSF-1 receptor promoted the development of granulocyte/macrophage (GM) colonies but did not alter the differentiation potential of either colony-forming unit (CFU)-GM or CFU-mix.


Subject(s)
Receptor, Macrophage Colony-Stimulating Factor/biosynthesis , Receptors, Erythropoietin/biosynthesis , Stem Cells/physiology , Animals , Base Sequence , Bone Marrow Cells , Cell Differentiation , Cell Division , Cells, Cultured , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptors, Erythropoietin/genetics , Recombinant Proteins/biosynthesis , Retroviridae/genetics , Spleen/cytology
20.
Mol Cell Biol ; 14(4): 2266-77, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8139532

ABSTRACT

If the env gene of spleen focus-forming virus (SFFV) is replaced by a cDNA encoding a constitutively active form of the erythropoietin receptor, EPO-R(R129C), the resultant recombinant virus, SFFVcEPO-R, induces transient thrombocytosis and erythrocytosis in infected mice. Clonogenic progenitor cell assays of cells from the bone marrow and spleens of these infected mice suggest that EPO-R(R129C) can stimulate proliferation of committed megakaryocytic and erythroid progenitors as well as nonerythroid multipotent progenitors. From the spleens of SFFVcEPO-R-infected mice, eight multiphenotypic immortal cell lines were isolated and characterized. These included primitive erythroid, lymphoid, and monocytic cells. Some expressed proteins characteristic of more than one lineage. All cell lines resulting from SFFVcEPO-R infection contained a mutant form of the p53 gene. However, in contrast to infection by SFFV, activation of PU.1 gene expression, by retroviral integration, was not observed. One cell line had integrated a provirus upstream of the fli-1 gene, in a location typically seen in erythroleukemic cells generated by Friend murine leukemia virus infection. This event led to increased expression of fli-1 in this cell line. Thus, infection by SFFVcEPO-R can induce proliferation and lead to transformation of nonerythroid as well as very immature erythroid progenitor cells. The sites of proviral integration in clonal cell lines are distinct from those in SFFV-derived lines.


Subject(s)
Blood Platelets/cytology , Cell Transformation, Viral , Erythrocytes/cytology , Genes, env , Hematopoietic Stem Cells/cytology , Receptors, Erythropoietin/physiology , Spleen Focus-Forming Viruses/genetics , Animals , Bone Marrow/physiology , Bone Marrow Cells , Cell Division , Cell Line , Erythrocyte Count , Female , Hematopoietic Stem Cells/physiology , Leukemia, Experimental/blood , Mice , Mice, Inbred BALB C , Mice, Inbred Strains , Platelet Count , Receptors, Erythropoietin/biosynthesis , Retroviridae Infections/blood , Spleen/cytology , Spleen/physiology , Tumor Virus Infections/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...