Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
J Immunol ; 166(8): 5058-67, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11290787

ABSTRACT

Members of the HOX family of homeobox transcription factors play a role in pattern formation in diverse developmental systems. The clearly documented role of HOX genes in the proliferation and differentiation of primary hematopoietic cells and cell lines provides a convenient system to pursue a biochemical analysis of HOX gene function in mammalian cells. To explore the role of HOXB7 in myeloid hematopoiesis, a number of mutations and deletions in the gene were constructed that targeted sequences with known functions or in regions that had not been examined previously. The wild-type and mutant B7 constructs were introduced into the murine myelomonocytic cell line, 32D, and assayed for their effects on G-CSF-induced myeloid differentiation. Wild-type HOXB7 inhibited the differentiation of 32D cells, whereas mutations in the Pbx-binding pentapeptide motif or the DNA-binding homeodomain, as well as internal deletions of the N-terminal unique region, blocked this effect. Interestingly, mutations eliminating two target sites for casein kinase II, the glutamate-rich C terminus, or the first 14 amino acids of HOXB7, led to enhanced 32D differentiation. A model proposing a role for these regions of HOXB7 is presented.


Subject(s)
Homeodomain Proteins/physiology , 3T3 Cells , Animals , Casein Kinase II , Cell Differentiation/genetics , Cell Line , Clone Cells , DNA, Complementary/genetics , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocytes/cytology , Granulocytes/enzymology , Growth Inhibitors/genetics , Growth Inhibitors/metabolism , Growth Inhibitors/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , K562 Cells , Mice , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/physiology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Transfection
2.
Cancer Res ; 60(23): 6744-9, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11118061

ABSTRACT

The majority of ovarian tumors arise from the transformation of the ovarian surface epithelial cells, a single layer of cells surrounding the ovary. To identify genes that may contribute to the malignant phenotype of ovarian cancers, cDNA representational difference analysis was used to compare expressed genes in primary cultures of normal human ovarian surface epithelium (HOSE) and ovarian tumor-derived epithelial cells from the Cedars-Sinai Ovarian Cancer (CSOC) repository. A total of 255 differentially expressed genes were identified, of which 160 and 95 were specifically expressed in HOSE and CSOC cells, respectively. Using cDNA array hybridization, the expression profiles of the genes identified by cDNA-representational difference analysis were examined in an additional 5 HOSE and 10 CSOC lines. The comparison of average signal of each gene revealed 44 HOSE-specific and 16 CSOC-specific genes that exhibited at least a 2.5-fold difference in expression. A large number of genes identified in this study encode membrane-associated or secreted proteins and, hence, may be useful as targets in the development of serum-based diagnostic markers for ovarian cancer. Very few genes associated with protein synthesis or metabolism were identified in this study, reflecting the lack of observable differences in phenotypic or growth characteristics between HOSE and CSOC cells. Northern blot analysis on a subset of these genes demonstrated comparable levels of gene expression as observed in the cDNA array hybridization.


Subject(s)
Gene Expression Profiling , Ovarian Neoplasms/genetics , Ovary/physiology , Blotting, Northern , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Epithelial Cells/physiology , Female , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , Humans , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis/methods , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovary/metabolism , Tumor Cells, Cultured
3.
J Immunol ; 165(8): 4428-36, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11035081

ABSTRACT

The Notch transmembrane receptors play important roles in precursor survival and cell fate specification during hematopoiesis. To investigate the function of Notch and the signaling events activated by Notch in myeloid development, we expressed truncated forms of Notch1 or Notch2 proteins that either can or cannot activate the core binding factor 1 (CBF1) in 32D (clone 3) myeloblasts. 32D cells proliferate as blasts in the presence of the cytokines, GM-CSF or IL-3, but they initiate differentiation and undergo granulopoiesis in the presence of granulocyte CSF (G-CSF). 32D cells expressing constitutively active forms of Notch1 or Notch2 proteins that signal through the CBF1 pathway maintained significantly higher numbers of viable cells and exhibited less cell death during G-CSF induction compared with controls. They also displayed enhanced entry into granulopoiesis, and inhibited postmitotic terminal differentiation. In contrast, Notch1 constructs that either lacked sequences necessary for CBF1 binding or that failed to localize to the nucleus had little effect. Elevated numbers of viable cells during G-CSF treatment were also observed in 32D cells overexpressing the basic helix-loop-helix protein (bHLH), HES1, consistent with activation of the CBF1 pathway. Taken together, our data suggest that Notch signaling enhances 32D cell survival, promotes entry into granulopoiesis, and inhibits postmitotic differentiation through a CBF1-dependent pathway.


Subject(s)
Membrane Proteins/physiology , Myeloid Progenitor Cells/physiology , Receptors, Cell Surface/physiology , Signal Transduction/physiology , Transcription Factors , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation/physiology , Cell Line , Cell Survival/genetics , Cell Survival/physiology , Gene Deletion , Genetic Vectors/biosynthesis , Genetic Vectors/chemical synthesis , Granulocyte Colony-Stimulating Factor/pharmacology , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Intracellular Fluid/physiology , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Phenotype , Receptor, Notch1 , Receptor, Notch2 , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Receptors, Notch , Subcellular Fractions/metabolism , Transcription Factor HES-1
4.
Blood ; 94(2): 519-28, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10397719

ABSTRACT

The role of the homeobox gene HOXA5 in normal human hematopoiesis was studied by constitutively expressing the HOXA5 cDNA in CD34(+) and CD34(+)CD38(-) cells from bone marrow and cord blood. By using retroviral vectors that contained both HOXA5 and a cell surface marker gene, pure populations of progenitors that expressed the transgene were obtained for analysis of differentiation patterns. Based on both immunophenotypic and morphological analysis of cultures from transduced CD34(+) cells, HOXA5 expression caused a significant shift toward myeloid differentiation and away from erythroid differentiation in comparison to CD34(+) cells transduced with Control vectors (P =.001, n = 15 for immunophenotypic analysis; and P <.0001, n = 19 for morphological analysis). Transduction of more primitive progenitors (CD34(+)CD38(-) cells) resulted in a significantly greater effect on differentiation than did transduction of the largely committed CD34(+) population (P =.006 for difference between HOXA5 effect on CD34(+) v CD34(+)CD38(-) cells). Erythroid progenitors (burst-forming unit-erythroid [BFU-E]) were significantly decreased in frequency among progenitors transduced with the HOXA5 vector (P =.016, n = 7), with no reduction in total CFU numbers. Clonal analysis of single cells transduced with HOXA5 or control vectors (cultured in erythroid culture conditions) showed that HOXA5 expression prevented erythroid differentiation and produced clones with a preponderance of undifferentiated blasts. These studies show that constitutive expression of HOXA5 inhibits human erythropoiesis and promotes myelopoiesis. The reciprocal inhibition of erythropoiesis and promotion of myelopoiesis in the absence of any demonstrable effect on proliferation suggests that HOXA5 diverts differentiation at a mulitpotent progenitor stage away from the erythroid toward the myeloid pathway.


Subject(s)
Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/physiology , Phosphoproteins/physiology , Cells, Cultured , Colony-Forming Units Assay , DNA, Complementary/genetics , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Erythropoiesis/genetics , Genetic Vectors/genetics , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/genetics , Humans , Moloney murine leukemia virus/genetics , Phosphoproteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology
5.
Stem Cells ; 17(3): 162-71, 1999.
Article in English | MEDLINE | ID: mdl-10342559

ABSTRACT

The cell surface receptor Notch1 is expressed on CD34+ hematopoietic precursors, whereas one of its ligands, Jagged1, is expressed on bone marrow stromal cells. To examine the role of Notch signaling in early hematopoiesis, human CD34+ cells were cultured in the presence or absence of exogenous cytokines on feeder layers that either did or did not express Jagged1. In the absence of recombinant growth factors, Jagged1 decreased myeloid colony formation by CD34+ cells, as well as 3H-thymidine incorporation and entry into S phase. In the presence of a strong cytokine signal to proliferate and mature, (interleukin 3 [IL-3] and IL-6, stem cell factor [SCF], and G-CSF), Jagged1 did not significantly alter either the fold expansion or the types of colonies formed by CD34+ cells. However, in the presence of SCF alone, Jagged1 increased erythroid colony formation twofold. These results demonstrate that Notch can modulate a growth factor signal, and that in the absence of growth factor stimulation, the Jagged1-Notch pathway preserves CD34+ cells in an immature state.


Subject(s)
Hematopoietic Stem Cells/metabolism , Membrane Proteins/physiology , Proteins/physiology , 3T3 Cells , Animals , Antigens, CD34/metabolism , Calcium-Binding Proteins , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , In Vitro Techniques , Intercellular Signaling Peptides and Proteins , Interleukin-3/pharmacology , Interleukin-6/pharmacology , Jagged-1 Protein , Mice , Receptors, Notch , Serrate-Jagged Proteins , Stem Cell Factor/physiology , Transfection
6.
Blood ; 93(10): 3391-400, 1999 May 15.
Article in English | MEDLINE | ID: mdl-10233891

ABSTRACT

During the process of normal hematopoiesis, proliferation is tightly linked to maturation. The molecular mechanisms that lead to production of mature effector cells with a variety of phenotypes and functions from a single multipotent progenitor are only beginning to be elucidated. It is important to determine how these maturation events are regulated at the molecular level, because this will provide significant insights into the process of normal hematopoiesis as well as leukemogenesis. Transcription factors containing the highly conserved homeobox motif show considerable promise as potential regulators of hematopoietic maturation events. In this study, we focused on identification and characterization of homeobox genes of the HOX family that are important in regulating normal human myeloid differentiation induced by the hematopoietic growth factor, granulocyte-macrophage colony-stimulating factor (GM-CSF). We have identified three homeobox genes, HOX A5, HOX B6, and HOX B7, which are expressed during early myelopoiesis. Treating bone marrow cells with antisense oligodeoxynucleotides to HOX A5 resulted in inhibition of granulocytic/monocytic hematopoiesis and increased the generation of erythroid progenitors. Also, overexpression of HOX A5 inhibited erythroid differentiation of the K562 cell line. Based on these observations, we propose that HOX A5 functions as an important regulator of hematopoietic lineage determination and maturation.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Erythroid Precursor Cells/cytology , Genes, Homeobox , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Homeodomain Proteins/genetics , Leukopoiesis/physiology , Phosphoproteins/genetics , Cell Differentiation , Cloning, Molecular , Colony-Forming Units Assay , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/physiology , Gene Expression Regulation/drug effects , Hematopoiesis/drug effects , Homeodomain Proteins/biosynthesis , Humans , K562 Cells , Leukopoiesis/genetics , Multigene Family , Oligodeoxyribonucleotides, Antisense/pharmacology , Phosphoproteins/biosynthesis , Recombinant Proteins/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Transfection
7.
Blood ; 92(3): 867-76, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9680354

ABSTRACT

The high-affinity human granulocyte-macrophage colony-stimulating factor (GM-CSF) receptor (GMR) consists of an alpha (GMRalpha) and a common beta (betac) subunit. The intracellular domain of betac has been extensively characterized and has been shown to be critical for the activation of both the JAK/STAT and MAP kinase pathways. The function of the intracellular domain of GMRalpha, however, is not as well characterized. To determine the role of this domain in GMR signaling, an extensive structure-function analysis was performed. Truncation mutants alpha362, alpha371, and alpha375 were generated, as well as the site-directed mutants alphaVQVQ and alphaVVVV. Although alpha375beta, alphaVQNQbeta, and alphaVVVVbeta stimulated proliferation in response to human GM-CSF, the truncation mutants alpha362beta and alpha371beta were incapable of transducing a proliferative signal. In addition, both alpha371 and alphaVVVV were expressed at markedly reduced levels, indicating the importance of residues 372 to 374 for proper protein expression. More importantly, we show that GMRalpha plays a direct role in the activation of the JAK/STAT pathway, and electrophoretic mobility shift assays (EMSA) indicate that both GMRalpha and betac play a role in determining the STAT5 DNA binding complex activated by the GMR. Thus, the intracellular domain of the human GMRalpha is important for activation of the JAK/STAT pathway and protein stabilization.


Subject(s)
DNA-Binding Proteins/metabolism , Milk Proteins , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Trans-Activators/metabolism , Animals , Cell Division , Cell Line , Enzyme Activation , Gene Expression Regulation/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Janus Kinase 2 , Mice , Mutagenesis, Site-Directed , Phosphorylation , Protein Conformation , Protein Processing, Post-Translational , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , STAT5 Transcription Factor , Sequence Deletion , Signal Transduction/physiology , Structure-Activity Relationship , Substrate Specificity
8.
Stem Cells ; 14(6): 714-24, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8948028

ABSTRACT

Fes is a nonreceptor protein tyrosine kinase that has been implicated in a variety of cytokine signal transduction pathways, as well as differentiation of myeloid cells. To address the role of Fes in these processes, we overexpressed a kinase-defective Fes protein in the factor-dependent cell-lines, TF-1 and 32D. Proliferative responses to GM-CSF and interleukin 3, and the induction of differentiation by G-CSF were not altered by expression of the kinase mutant Fes protein, indicating that Fes kinase activity is not critical for these biological events in these cell lines.


Subject(s)
Bone Marrow Cells , Membrane Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Cell Cycle , Cell Differentiation , Cell Division , Electrophoresis, Polyacrylamide Gel , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Interleukin-3/pharmacology , Polymerase Chain Reaction , Proto-Oncogene Proteins c-fes
9.
Stem Cells ; 14(1): 117-23, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8820957

ABSTRACT

The study of oncogenes has provided numerous insights, not only into the mechanisms by which growth regulation becomes uncontrolled in cancer cells, but also into signal transduction processes which regulate the orderly proliferation and maturation of cells. c-fes/fps is a cellular oncogene which has been transduced frequently by mammalian and avian retroviruses. There are several features about Fes which suggest it may play a unique role in myeloid cell growth and differentiation. While it contains a tyrosine kinase and SH2 domain, there is no SH3 domain or carboxy terminal regulatory phosphotyrosine such as found in the Src family of kinases. Fes has a unique N-terminal domain of over 400 amino acids of unknown function. It has been implicated in signaling by a variety of hematopoietic growth factors, and is predominantly a nuclear protein.


Subject(s)
Cell Transformation, Neoplastic , Hematopoiesis/physiology , Protein-Tyrosine Kinases , Proto-Oncogene Proteins/physiology , Animals , Bone Marrow Cells , Humans , Proto-Oncogene Proteins c-fes , Signal Transduction
10.
J Immunol ; 154(7): 3444-53, 1995 Apr 01.
Article in English | MEDLINE | ID: mdl-7897225

ABSTRACT

A superfamily of growth factor and cytokine receptors has recently been identified, which is characterized by four spatially conserved cysteine residues and a tryptophan-serine motif (WSXWS) in the extracellular domain and proline-rich cytoplasmic domain. The high-affinity human granulocyte-macrophage colony-stimulating factor (GM-CSF) receptor, hGM-CSFR, consists of two subunits, alpha (hGM-CSFR alpha), which is required for ligand binding, and beta (hGM-CSFR beta), which is required for signal transduction. Both the alpha and beta subunits are members of the cytokine receptor superfamily. In this study, we analyzed mutations in the conserved amino acids of the alpha subunit to determine their function in signal transduction, as assayed by tyrosine phosphorylation and proliferation. Disruption of either of the conserved disulfide bonds in the extracellular domain abolishes low-affinity binding but not binding to a preformed heterodimeric complex with the beta-chain. Cells expressing receptors with mutations in cysteines 2 or 3 grew as well as cells expressing wild-type receptors in human GM-CSF (hGM-CSF) and phosphorylated the same proteins on tyrosine residues, although the level of phosphorylation may be attenuated; cysteine 3 appears to be required for generation of the true high-affinity binding site. The WSXWS motif and the cytoplasmic domain are required for function of the human GM-CSF receptor, as stable cell lines expressing receptors with these mutations were unable to proliferate continuously in hGM-CSF. Surprisingly, no function for the conserved proline-rich region of the cytoplasmic domain could be ascertained from these studies; cells expressing these receptors were indistinguishable from wild-type in both binding and functional assays.


Subject(s)
Cell Division/physiology , Protein-Tyrosine Kinases/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Amino Acid Sequence , Cell Line , Flow Cytometry , Humans , Immunoblotting , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphoproteins/analysis , Protein Binding/physiology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Recombinant Proteins/chemistry , Signal Transduction
11.
Oncogene ; 10(6): 1239-42, 1995 Mar 16.
Article in English | MEDLINE | ID: mdl-7700650

ABSTRACT

FES is a non-receptor protein tyrosine kinase expressed in hematopoietic progenitors and differentiated myeloid cells. It has recently been implicated in granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-3 (IL-3) and erythropoietin signal transduction. To better understand the role played by FES in normal and neoplastic hematopoiesis, we used cell fractionation techniques to examine the subcellular localization of FES in myeloid cells and cell lines. FES was observed in the nuclear, granular and plasma membrane fractions of primary human neutrophils and the myeloid leukemia cell line, HL-60. The nuclear localization was confirmed by immunocytochemistry of neutrophils.


Subject(s)
Cell Nucleus/enzymology , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Animals , Cell Line , Cloning, Molecular , Humans , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-fes , Subcellular Fractions/enzymology , Tumor Cells, Cultured
12.
Blood ; 85(3): 692-7, 1995 Feb 01.
Article in English | MEDLINE | ID: mdl-7530503

ABSTRACT

Homeobox genes encode transcription factors known to be important morphogenic regulators during embryogenesis. An increasing body of work implies a role for homeobox genes in both hematopoiesis and oncogenesis. We have analyzed the role of the homeobox gene, HOX B7, in the program of differentiation of the biphenotypic myeloid cell line, HL60. Induction of monocytic differentiation in HL-60 cells by vitamin D3 resulted in rapid expression of HOX B7 mRNA, but stimulation with phorbol ester or dimethyl sulfoxide (DMSO) did not. Constitutive overexpression of HOX B7 in the HL60 cell line inhibited the granulocytic differentiation associated with stimulation with DMSO or retinoic acid, but had no effect on the monocytic differentiation induced by vitamin D3. Normal human monocytes do not constitutively express HOX B7, nor are they able to be induced to do so by stimulation with colony-stimulating factor 1 (CSF-1) and gamma interferon (IFN gamma), or with vitamin D3 and lipopolysaccharide. Human bone marrow (BM) cells were found to express HOX B7 in response to granulocyte-macrophage CSF (GM-CSF) and antisense oligonucleotides directed against HOX B7 inhibited the formation of colonies derived from GM-CSF-stimulated BM. These data suggest a critical role for HOX B7 in myelomonocytic differentiation.


Subject(s)
Genes, Homeobox , Base Sequence , Calcitriol/pharmacology , Cell Differentiation , Cell Line , Cloning, Molecular , DNA Primers , Humans , Leukemia, Promyelocytic, Acute , Molecular Sequence Data , Oligonucleotides, Antisense , Polymerase Chain Reaction , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , RNA-Directed DNA Polymerase , Tumor Cells, Cultured
13.
Curr Top Microbiol Immunol ; 193: 25-49, 1995.
Article in English | MEDLINE | ID: mdl-7648877

ABSTRACT

We have focused this chapter on interactions with two of the best characterized transregulatory genes, tax for HTLV-I/II and Tat for HIV-1. Both genes illustrate the complex interplay between retroviral regulatory genes and cellular gene regulation. In both instances a viral gene of relatively straightforward function in the viral context appears to cause extensive dysregulation of cellular genes, either directly or as a consequence of altered cellular differentiation. Understanding this viral/cellular gene cross-talk may elucidate mechanisms leading to malignant transformation autoimmune disease and to neurologic and paraneoplastic complications such as hypercalcemia for HTLV-I/II, as well as the pathogenesis of immune dysfunction and opportunistic malignancy in HIV-I/II-infected individuals. An understanding of functional mechanisms of these transregulatory viral genes will undoubtedly afford better explanations for the myriad manifestations of retroviral infection.


Subject(s)
Gene Expression Regulation, Viral , HIV-1/genetics , Human T-lymphotropic virus 1/genetics , Human T-lymphotropic virus 2/genetics , Transcriptional Activation , Animals , Gene Products, tax/metabolism , Genes, tat , Humans
14.
Stem Cells ; 12(6): 626-37, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7533581

ABSTRACT

There is increasing clinical interest focused on ex vivo manipulation and expansion of hematopoietic cells. In this study, we demonstrate that a simple combination of growth factors can expand progenitors to yield functional myeloid cells. Furthermore, this system can produce mature, functionally competent cells in the absence of fetal bovine serum (FBS), which will enhance the clinical utility of this approach. Hematopoietic progenitor cells obtained from normal bone marrow and from leukapheresis products were studied. The mononuclear fraction was enriched for CD34 cells using the Ceprate CD34 biotin kit (CellPro #LC34-1 or LC34-2). The selected cells were expanded for two weeks in Iscove's medium supplemented with 20% FBS and various combinations of interleukin-3 (IL-3), granulocyte colony-stimulating factor (G-CSF), stem cell factor (SCF) and interleukin -6 (IL-6) added either simultaneously or sequentially. The optimal combination of these factors identified for myeloid expansion was simultaneous addition of IL-3, SCF and G-CSF (at 50 ng/ml each), resulting in an average 773 +/- 133-fold expansion of nucleated cells (n = 5). When corrected for the purity of CD34 cells in the starting population, the mean fold expansion with IL-3, SCF and G-CSF was 2,265 +/- 729. A mean of 74.7 +/- 10.5% (n = 3) of the expanded cells was positive for CD11b; 86-91% (n = 2) of the cells were promyelocytes or more mature granulocytes. Functional assays demonstrated normal phagocytosis and intracellular killing of Staphylococcus aureus (S. aureus) by the expanded cell population. Studies performed using cells expanded in defined serum-free media demonstrated that fold expansion was decreased and that the cells produced were less mature and functionally less competent than cells expanded with FBS. The decreased expansion could be partially reversed, and the functionality almost completely restored by the addition of autologous plasma.


Subject(s)
Hematopoietic Stem Cells/cytology , Antigens, CD/metabolism , Antigens, CD34 , Cell Separation , Colony-Forming Units Assay , Culture Media , Cytotoxicity, Immunologic , Flow Cytometry , Granulocyte Colony-Stimulating Factor/administration & dosage , Granulocytes/cytology , Granulocytes/drug effects , Granulocytes/physiology , Growth Substances/administration & dosage , Hematopoiesis/drug effects , Hematopoietic Cell Growth Factors/administration & dosage , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Humans , In Vitro Techniques , Interleukin-3/administration & dosage , Interleukin-6/administration & dosage , Phagocytosis , Stem Cell Factor
15.
Blood ; 84(8): 2523-30, 1994 Oct 15.
Article in English | MEDLINE | ID: mdl-7919370

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a hematopoietic growth factor that stimulates the proliferation, maturation, and functional activity of myeloid cells in peripheral blood and bone marrow. Expression of GM-CSF is tightly regulated and is limited to cells stimulated directly (T cells, macrophages) or indirectly (fibroblasts, endothelial cells) by immune challenge. Several studies of the transcriptional control of GM-CSF expression have elucidated a region of the GM-CSF promoter that mediates positive regulatory activity in a number of cell types. This region contains a direct repeat of the sequence CATTA/T that extends from nucleotides -37 to -48 upstream of the start of mRNA synthesis. Although specific DNA:protein interactions have been shown within this region, neither the nature nor the number of nuclear factors responsible for these interactions have been characterized. In this study, we use DNase I footprinting analysis to demonstrate that point mutations, which inactivate the GM-CSF promoter, disrupt DNA:protein interactions within this region. By combined electrophoretic mobility shift and ultraviolet cross-linking analysis, we have detected several protein species that bind specifically to the positive regulatory sequence.


Subject(s)
DNA/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid , Base Sequence , Cross-Linking Reagents , DNA/chemistry , Deoxyribonuclease I , Electrophoresis , Humans , Molecular Sequence Data , Oligonucleotide Probes , Phytohemagglutinins/pharmacology , Point Mutation , Repetitive Sequences, Nucleic Acid , T-Lymphocytes/ultrastructure , Ultraviolet Rays
16.
Mol Cell Biol ; 14(9): 5975-85, 1994 Sep.
Article in English | MEDLINE | ID: mdl-8065330

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) stimulates myeloid progenitor cell proliferation and enhances the function of terminally differentiated effector cells. Interleukin-3 (IL-3) stimulation results in the proliferation and maturation of early bone marrow progenitor cells. These activities are mediated by non-tyrosine kinase-containing receptors which consist of ligand-specific alpha subunits that complex with a common beta subunit required for signal transduction. Both GM-CSF and IL-3 rapidly and transiently induce expression of early growth response gene 1 (egr-1) in the human factor-dependent cell line TF-1. To define the mechanism of early response gene induction by GM-CSF and IL-3, growth factor- and serum-starved TF-1 cells transfected with recombinant constructs containing sequences of the human egr-1 promoter were stimulated with GM-CSF or IL-3. A 116-nucleotide (nt) region of the egr-1 promoter which contains sequences inducible by GM-CSF and IL-3 was defined. DNase I footprint analysis identified a 20-nt region, including nt -57 to -76, which contains a potential cyclic AMP (cAMP) response element (CRE). Electrophoretic mobility shift assays performed with CREB antibody confirmed the presence of CREB in the DNA-binding complex. Mutational analysis of the cytokine-responsive region of the egr-1 promoter revealed that both the cAMP response and serum response elements are required for induction by GM-CSF and IL-3. Nuclear extracts from GM-CSF- or IL-3-stimulated but not unstimulated TF-1 cells contain factors which specifically bind to the Egr-1-binding site in the nt -600 to -480 region of the promoter. Electrophoretic mobility shift assays were performed with antibodies against the Egr-1 protein to demonstrate the presence of the protein product in the shifted complex. Our studies suggest that the Egr-1 protein may further stimulate transcription of the egr-1 gene in response to GM-CSF as a secondary event.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Immediate-Early Proteins , Interleukin-3/physiology , Promoter Regions, Genetic , Transcription Factors/genetics , Base Sequence , Binding Sites , Cyclic AMP/physiology , Early Growth Response Protein 1 , Humans , In Vitro Techniques , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligodeoxyribonucleotides/chemistry , RNA, Messenger/genetics , Signal Transduction , Structure-Activity Relationship , Transcriptional Activation
17.
Mol Cell Biol ; 14(3): 2213-21, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8114751

ABSTRACT

Human granulocyte-macrophage colony-stimulating factor (GM-CSF) stimulates the proliferation and maturation of normal myeloid progenitor cells and can also stimulate the growth of acute myelogenous leukemia (AML) blasts. GM-CSF is not normally produced by resting cells but is expressed by a variety of activated cells including T lymphocytes, macrophages, and certain cytokine-stimulated fibroblasts and endothelial cells. Production of GM-CSF by cultured AML cells has been demonstrated, and GM-CSF expression by normal myeloid progenitors has been postulated to play a role in myelopoiesis. We have investigated the regulation of expression of GM-CSF in AML cell lines, and our results demonstrate the presence of a strong constitutive promoter element contained within 53 bp upstream of the cap site. We have also identified a negative regulatory element located immediately upstream of the positive regulatory element (within 69 bp of the cap site) that is active in AML cell lines but not T cells or K562 CML cells. Competition transfection and mobility shift studies demonstrate that this activity correlates with binding of a 45-kDa protein.


Subject(s)
Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Leukemia, Myeloid, Acute/genetics , Promoter Regions, Genetic , Base Sequence , Cell Line , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Humans , Molecular Sequence Data , Molecular Weight , Mutagenesis, Site-Directed , Oligodeoxyribonucleotides/chemistry , RNA, Messenger/genetics , Sequence Deletion , Structure-Activity Relationship , T-Lymphocytes/physiology , Transcription, Genetic
18.
Recept Channels ; 2(3): 175-81, 1994.
Article in English | MEDLINE | ID: mdl-7874444

ABSTRACT

Granulocyte-macrophage colony stimulating factor (GM-CSF) and Interleukin-3 (IL-3) are cytokines which stimulate myeloid bone marrow progenitor cell proliferation and maturation. GM-CSF also enhances the function of terminally differentiated effector cells including neutrophils, monocytes and eosinophils. Both growth factors exhibit similar biological activities on overlapping cell populations which are mediated by high affinity receptors. These receptors share a common beta subunit necessary for signal transduction. The receptors for GM-CSF and IL-3 are members of the hematopoietin receptor superfamily and consequently lack intrinsic tyrosine kinase activity. Several kinases, including JAK2 and raf-1, and other downstream molecules are likely to be responsible for the functional redundancy demonstrated by GM-CSF and IL-3 in factor-responsive cells. This review discusses recent findings which elucidate the signaling pathways activated by these two cytokines.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Hematopoietic Stem Cells/physiology , Interleukin-3/physiology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Receptors, Interleukin-3/physiology , Signal Transduction/physiology , Amino Acid Sequence , Animals , Enzyme Activation , Gene Expression Regulation/physiology , Humans , Molecular Sequence Data , Multigene Family , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/physiology
19.
J Biol Chem ; 269(1): 277-83, 1994 Jan 07.
Article in English | MEDLINE | ID: mdl-8276807

ABSTRACT

A superfamily of growth factor and cytokine receptors has recently been identified, which is characterized by four spatially conserved cysteine residues, a tryptophan-serine motif (WSXWS) in the extracellular domain, and a proline-rich cytoplasmic domain. The high affinity human granulocyte-macrophage colony-stimulating factor (GM-CSF) receptor (hGM-CSFR) consists of two subunits, alpha (hGM-CSFR alpha) and beta (hGM-CSFR beta), both of which are members of the receptor superfamily. In this study, we prepared mutations in conserved amino acids of the receptor subunit necessary for GM-CSF binding (hGM-CSFR alpha) and analyzed mutant receptors for low affinity binding, internalization, and high affinity binding when complexed with the beta subunit. Mutations in the cytoplasmic domain did not affect GM-CSF binding or receptor internalization. Mutation of a single conserved serine residue within the WSXWS motif diminishes cell surface receptor expression but not ligand binding. Mutation of either the second or third conserved cysteine residue of hGM-CSFR alpha resulted in complete loss of low affinity binding; however, co-expression of the cysteine 2 mutant with hGM-CSFR beta yielded a high affinity receptor complex. Since neither the cysteine 2 mutant nor the beta subunit can bind ligand alone, this result suggests that hGM-CSFR alpha and hGM-CSFR beta exist in a preformed heterodimeric protein complex on the plasma membrane.


Subject(s)
Conserved Sequence , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Amino Acid Sequence , Base Sequence , Cell Membrane/metabolism , Cells, Cultured , Cytosol/metabolism , DNA, Complementary , Humans , Ligands , Molecular Sequence Data , Mutagenesis , Proline/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
20.
Blood ; 82(11): 3279-82, 1993 Dec 01.
Article in English | MEDLINE | ID: mdl-8241500

ABSTRACT

We previously showed the presence of receptors for granulocyte-macrophage colony-stimulating factor (GM-CSF) on tumor tissues and tumor cell lines that are derived from the neural crest. To determine whether normal neural cells express functional GM-CSF receptors, we isolated and analyzed primary rat brain cells, including microglia, astrocytes, and oligodendrocytes. Scatchard analysis of equilibrium binding of 125I-GM-CSF to primary rat oligodendrocytes showed an average of 1,110 GM-CSF binding sites per cell, with a kd of 20 pmol/L. In six separate experiments, no specific binding was detectable on the astrocyte population. Microglia were used in competitive binding experiments with oligodendrocytes, and addition of microglia did not increase the specific binding of labeled ligand to oligodendrocytes. In dose-response assays, we measured 3H-thymidine uptake in rat oligodendrocytes, microglia and control murine 32D cells stimulated with various concentrations of GM-CSF. Over concentration ranges of 0.025 to 1000 pmol/L, cell proliferation and peak 3H-thymidine incorporation was observed at approximately 30 pmol/L for both the control cells and the oligodendrocytes. However, the microglial cells did not proliferate in response to GM-CSF. These data indicate the presence of a functional receptor for GM-CSF on primary rat oligodendrocytes, and suggest that hematopoietic growth factors such as GM-CSF may play a role in nerve cell development, function, or response to injury.


Subject(s)
Oligodendroglia/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/analysis , Animals , Cells, Cultured , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hematopoiesis , Rats , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...