Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Anticancer Res ; 21(5): 3261-8, 2001.
Article in English | MEDLINE | ID: mdl-11848481

ABSTRACT

BACKGROUND: p27Kip1 is a potential tumor suppressor gene. As malignant gliomas express Fas at high levels, the relationship between Fas-mediated apoptosis and p27Kip1 expression may improve therapeutic approaches for treating gliomas. MATERIALS AND METHODS: In this study, we transduced U-373MG glioma cells with the Fas ligand or caspase-8 genes using adenovirus vectors after transduction of the p27Kip1 gene to induce cell cycle arrest in U-373MG cells, and evaluated the degree of apoptosis. RESULTS: The results demonstrate that expression of p27Kip1 enhanced Fas ligand- or caspase-8-mediated apoptosis in U-373MG cells. Expression of apoptosis-related genes such as Bax, Bcl-X(L), Bcl-2 or caspase-8 were reduced by p27Kip1 transduction compared with that of beta-actin, whereas p27Kip1 transduction did not affect the expression level of Fas or the Fas ligand. CONCLUSION: Combined transduction of p27Kip1 with Fas ligand or caspase-8 would overide the resistance mechanism to apoptosis in malignant gliomas.


Subject(s)
Apoptosis/genetics , Caspases/physiology , Cell Cycle Proteins/genetics , Glioma/pathology , Membrane Glycoproteins/physiology , Tumor Suppressor Proteins/genetics , Adenoviridae/genetics , Apoptosis/physiology , Caspase 8 , Caspase 9 , Caspases/biosynthesis , Caspases/genetics , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/physiology , Cyclin-Dependent Kinase Inhibitor p27 , Fas Ligand Protein , Genetic Vectors/genetics , Glioma/genetics , Glioma/metabolism , Humans , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Transduction, Genetic , Tumor Cells, Cultured , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/physiology
2.
Int Arch Allergy Immunol ; 106(4): 377-85, 1995 Apr.
Article in English | MEDLINE | ID: mdl-7536501

ABSTRACT

The c-kit protooncogene encodes a receptor tyrosine kinase that mediates signals required for differentiation, proliferation and survival of mast cells. We have already shown the constitutive activation of c-kit receptor tyrosine kinase (KIT) in a human mast cell leukemia line (HMC-1) and a murine mastocytoma cell line (P-815). We here examined whether such constitutive activation of KIT occurred in the rat tumor mast cell line RBL-2H3 as well, which is frequently used as a tool for studying functions of mast cells. In RBL-2H3 cells, KIT was constitutively phosphorylated on tyrosine and activated in the absence of autocrine production of its ligand, stem cell factor (SCF). Sequencing analysis revealed that one of c-kit genes of RBL-2H3 cells had a point mutation, resulting in amino acid substitution of Tyr for Asp in codon 817. When rat wild-type c-kit cDNA and mutant-type c-kit cDNA encoding KITTyr817 were transfected into cells of a human embryonic kidney cell line (293T), only mutant form KITTyr817 was constitutively phosphorylated on tyrosine and activated in the absence of SCF. Since mutations at the same Asp codon constitutively activated KIT in all the human HMC-1, murine P-815, and rat RBL-2H3 cell lines, and since the incorporation of antisense oligonucleotides of c-kit messenger RNA significantly suppressed the proliferation of RBL-2H3 cells, the activating mutations in the Asp codon of the c-kit gene appeared to be involved in neoplastic growth of mast cells.


Subject(s)
Aspartic Acid/pharmacology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Colony-Stimulating Factor/metabolism , Amino Acid Sequence , Animals , Base Sequence , DNA, Complementary/genetics , Humans , Mice , Molecular Sequence Data , Mutation , Oligonucleotides, Antisense/antagonists & inhibitors , Oligonucleotides, Antisense/metabolism , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-kit , Rats , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Colony-Stimulating Factor/genetics , Transfection , Tumor Cells, Cultured , Tyrosine/metabolism
3.
Blood ; 85(3): 790-8, 1995 Feb 01.
Article in English | MEDLINE | ID: mdl-7530509

ABSTRACT

The c-kit receptor tyrosine kinase (KIT) is activated upon ligand binding, thereby leading to a variety of signaling events that play a fundamental role in hematopoiesis. In addition to ligand-dependent activation, we have previously shown that KIT is constitutively activated in a ligand-independent manner by two point mutations, Val-559-->Gly (G559) mutation in the juxtamembrane domain and Asp-814-->Val (V814) mutation in the phosphotransferase domain. To investigate the biochemical consequence and biologic significance of these mutations, retroviral vectors encoding KITG559 or KITV814 were introduced into murine pro-B-type Ba/F3 cells and myeloid FDC-P1 cells, both of which require interleukin-3 (IL-3) for their growth and survival. In the cells, KITG559 or KITV814 were found to be constitutively phophorylated on tyrosine in the absence of stem cell factor (SCF) that is a ligand for KIT. Chemical cross-linking analysis showed that a substantial fraction of the phosphorylated KITG559 underwent dimerization even in the absence of SCF, whereas the phosphorylated KITV814 did not, suggesting the distinct mechanisms underlying constitutive activation of KIT by G559 and V814 mutations. Furthermore, the cells expressing either KITG559 or KITV814 were found to show a factor-independent growth, whereas the cells expressing wild-type KIT (KITWT) proliferated in response to SCF as well as IL-3. Moreover, subcutaneous injection of Ba/F3 cells expressing KITG559 or KITV814 into nude mice resulted in production of large tumors at all sites of the injection within 2 weeks, and all nude mice quickly succumbed to leukemia and died. These results suggest that, although the mechanisms underlying constitutive activation of KITG559 or KITV814 may be different, both of the activating mutations have a function to induce a factor-independent and tumorigenic phenotype. Also, the data of this study raise the possibility that the constitutively activating mutations of c-kit may play a causal role in development of hematologic malignancies.


Subject(s)
Point Mutation , Proto-Oncogene Proteins/metabolism , Proto-Oncogenes , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Colony-Stimulating Factor/metabolism , Amino Acid Sequence , Animals , Aspartic Acid , Cell Division , Cell Line , Cell Transformation, Neoplastic , Cell Transplantation , Female , Humans , Kinetics , Mice , Mice, Inbred BALB C , Mice, Nude , Mutagenesis, Site-Directed , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-kit , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Colony-Stimulating Factor/biosynthesis , Receptors, Colony-Stimulating Factor/genetics , Transfection , Valine
4.
J Histochem Cytochem ; 42(11): 1443-51, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7930526

ABSTRACT

We examined the ultrastructural localization of (a) a secondary granule matrix protein--eosinophil peroxidase (EPO)--by cytochemistry, (b) a secondary granule core protein (major basic protein, MBP) by immunogold labeling, and (c) a primary granule protein (the Charcot-Leyden crystal protein, CLC protein) by immunogold labeling in eosinophilic myelocytes (EMs) and mature, activated eosinophils that differentiated from umbilical cord blood progenitors cultured in the presence of recombinant human interleukin-5 (rhIL-5). These studies provide the first substructural localization of MBP to condensing cores of immature secondary granules of EMs, as well as identification of unicompartmental, MBP-rich secondary granules that are devoid of matrix compartments and EPO content and are not primary granules by virtue of their lack of CLC protein. These granules occur in quantity in IL-5-activated mature human eosinophils, which have previously been shown to actively transport EPO from the matrix compartments of their secondary granules to the extracellular milieu in smooth membrane-bound cytoplasmic vesicles, a secretory process termed piecemeal degranulation, whereby eosinophils progressively empty cytoplasmic granules of their contents in the absence of classical granule extrusion.


Subject(s)
Blood Proteins/analysis , Eosinophils/chemistry , Ribonucleases , Cell Differentiation/drug effects , Cells, Cultured , Eosinophil Granule Proteins , Eosinophils/cytology , Eosinophils/ultrastructure , Fetal Blood/chemistry , Fetal Blood/cytology , Glycoproteins/analysis , Hematopoiesis/physiology , Hematopoietic Stem Cells/chemistry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Immunohistochemistry , Interleukin-5/pharmacology , Lysophospholipase , Microscopy, Electron
5.
J Exp Med ; 179(6): 1757-66, 1994 Jun 01.
Article in English | MEDLINE | ID: mdl-7515098

ABSTRACT

Extracellular matrix (ECM) molecules such as fibronectin (FN), collagens, and laminin have important roles in hematopoiesis. However, little is known about the precise mechanisms by which ECM molecules regulate proliferation of human hematopoietic progenitor cells. In this study, we have investigated the effects of ECM molecules, particularly of FN, on the proliferation of a myeloid leukemia cell line, M07E, which proliferates in response to either human granulocyte/macrophage colony-stimulating factor (GM-CSF) or stem cell factor (SCF). The [3H]thymidine incorporation and cell enumeration assays showed that FN strikingly inhibited GM-CSF- or SCF-induced proliferation of M07E cells in a dose-dependent manner, whereas little or no inhibition was induced by collagen types I and IV. The growth suppression of M07E cells was not due to the inhibitory effect of FN on ligand binding or very early events in the signal transduction pathways from the GM-CSF or SCF receptors. DNA content analysis using flow cytometry after staining with propidium iodide revealed that the treatment of M07E cells with FN did not block the entry of the cells into the cell cycle after stimulation with GM-CSF or SCF, whereas the treatment resulted in the appearance of subdiploid peak. Furthermore, FN was found to induce oligonucleosomal DNA fragmentation and chromatin condensation in the cells even in the presence of GM-CSF or SCF, suggesting the involvement of programmed cell death (apoptosis) in the FN-induced growth suppression. The growth suppression or apoptosis induced by FN was rescued by the addition of either anti-FN antibody, anti-very late antigen 5 monoclonal antibody (anti-VLA5 mAb), or GRGDSP peptide, but not by that of anti-VLA4 mAb or GRGESP peptide, suggesting that the FN effects on M07E cells were mediated through VLA5. In addition, the FN-induced apoptosis was detectable in VLA5-positive human hematopoietic cell lines other than M07E cells, but not in any of the VLA5-negative cell lines. These results suggest that FN is capable of inducing apoptosis via its interaction with VLA5, and also raise the possibility that the FN-VLA5 interaction may contribute, at least in part, to negative regulation of hematopoiesis.


Subject(s)
Apoptosis/drug effects , Cell Division/drug effects , Extracellular Matrix Proteins/pharmacology , Fibronectins/pharmacology , Hematopoietic Stem Cells/cytology , Neutrophils/cytology , Neutrophils/physiology , Receptors, Fibronectin/physiology , Cell Line , Cells, Cultured , Collagen/pharmacology , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/physiology , Humans , Interleukin-3/pharmacology , Kinetics , Laminin/pharmacology , Neutrophils/drug effects , Receptors, Fibronectin/drug effects , Receptors, Very Late Antigen/drug effects , Receptors, Very Late Antigen/physiology , Stem Cell Factor , Thymidine/metabolism , Tumor Cells, Cultured
6.
Blood ; 83(9): 2619-26, 1994 May 01.
Article in English | MEDLINE | ID: mdl-7513208

ABSTRACT

The c-kit proto-oncogene encodes a receptor tyrosine kinase that is known to play a crucial role in hematopoiesis, especially in mast cell growth and differentiation. Although a number of dominant loss-of-function mutations of c-kit gene have been well characterized in mice, rats, and humans, little is known about the c-kit mutations contributing to ligand-independent activation of the c-kit receptor tyrosine kinase (KIT). In a murine mastocytoma cell line, P-815, KIT has been found to be constitutively phosphorylated on tyrosine and activated in a ligand-independent manner. Sequencing of the whole coding region of c-kit cDNA showed that c-kit cDNA of P-815 cells carries a point mutation in codon 814, resulting in amino acid substitution of Tyr for Asp. Murine wild-type c-kit cDNA and mutant-type c-kit cDNA encoding Tyr in codon 814 were expressed in cells of a human embryonic kidney cell line, 293T. In the transfected cells, mutant-form KITTyr814 was strikingly phosphorylated on tyrosine and activated in immune complex kinase reaction regardless of stimulation with a ligand for KIT (stem cell factor), whereas tyrosine phosphorylation and activation was barely detectable in wild-form KIT. The data presented here provide evidence for a novel activating mutation of c-kit gene that might be involved in neoplastic growth or oncogenesis of some cell types, including mast cells.


Subject(s)
Mast-Cell Sarcoma/genetics , Point Mutation , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Colony-Stimulating Factor/metabolism , Amino Acid Sequence , Animals , Base Sequence , Blotting, Southern , DNA, Complementary/chemistry , Enzyme Activation , Mice , Molecular Sequence Data , Phosphorylation , Phosphotyrosine , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-kit , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Colony-Stimulating Factor/genetics , Transfection , Tumor Cells, Cultured , Tyrosine/analogs & derivatives , Tyrosine/metabolism
7.
Leukemia ; 8 Suppl 1: S18-22, 1994 Apr.
Article in English | MEDLINE | ID: mdl-7512180

ABSTRACT

The c-kit proto-oncogene encodes a receptor tyrosine kinase that is known to play a crucial role in mast cell growth and differentiation. In a human mast cell leukemia cell line (HMC-1), KitR was found to be constitutively phosphorylated on tyrosine, activated and associated with phosphatidylinositol 3-kinase (P13K) in the absence of autocrine production of SCF. Sequencing of c-kit cDNA revealed that c-kit genes of HMC-1 cells were composed of a normal, wild-type allele and a mutant allele with two point mutations in codon 560 and codon 816, resulting in intracellular amino acid substitutions of Gly-560 for Val and Val-816 for Asp, respectively. Murine c-kit mutants encoding Gly-559 and/or Val-814, corresponding to human Gly-560 and/or Val-816, were constructed by site-directed mutagenesis and expressed in cells of a human embryonic kidney cell line (293T). In the transfected cells, KitR (Gly-559 + Val-814) and KitR (Val-814) were strikingly phosphorylated on tyrosine and activated in the absence of SCF, whereas tyrosine phosphorylation and activation of KitR (Gly-559) or wild-type KitR was modest or little, respectively. These results suggest that constitutive activation of KitR in HMC-1 results from the activating mutations of c-kit gene, and raise the possibility that the activating mutations, particularly at codon 814 of murine c-kit or at codon 816 of human c-kit, may participate in oncogenesis of mast cells.


Subject(s)
Leukemia, Mast-Cell/genetics , Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Colony-Stimulating Factor/genetics , Humans , Proto-Oncogene Mas , Proto-Oncogene Proteins c-kit , Tumor Cells, Cultured
8.
Blood ; 83(4): 916-25, 1994 Feb 15.
Article in English | MEDLINE | ID: mdl-7509212

ABSTRACT

The Ws mutant allele of rats represents a 12-base deletion at the tyrosine kinase domain of the c-kit gene. Although homozygous Ws/Ws rats were deficient in both connective tissue-type mast cells (CTMC) and mucosal-type mast cells (MMC), mast cells did develop when bone marrow cells of Ws/Ws rats were cultured in the presence of concanavalin A-stimulated spleen cell conditioned medium (ConA-SCM). Although the proliferative response of rat cultured mast cells (RCMC) derived from Ws/Ws rats to ConA-SCM was comparable to that of RCMC derived from control normal (+/+) rats, the proliferative response of Ws/Ws RCMC to rat recombinant stem cell factor (rrSCF; a ligand for the c-kit receptor tyrosine kinase) was much lower than that of +/+ RCMC. However, a slight c-kit kinase activity was detectable in Ws/Ws RCMC, and the proliferation of Ws/Ws RCMC was accelerated when rrSCF was added to ConA-SCM. Because CTMC contain rat mast cell protease-I (RMCP-I) and MMC contain RMCP-II, the phenotype of +/+ and Ws/Ws RCMC in various culture conditions was evaluated by immunohistochemistry of RMCPs. Both +/+ and Ws/Ws RCMC showed the MMC-like phenotype (RMCP-I-/II+) when they were cultured with ConA-SCM alone. Most +/+ RCMC and about half of Ws/Ws RCMC acquired a novel protease (RMCP-I+/II+) phenotype when they were cultured with rrSCF alone. However, because the number of Ws/Ws RCMC dropped to one-tenth in the medium containing rrSCF alone, the absolute number of Ws/Ws RCMC with the RMCP-I+/II+ phenotype did not increase significantly. The effect of rrSCF in inducing the novel phenotype was suppressed when ConA-SCM was added to rrSCF. In contrast, +/+ and Ws/Ws RCMC cocultured with +/+ fibroblasts showed the RMCP-I+/II+ phenotype even in the presence of ConA-SCM. Moreover, a fibroblast cell line derived from SI/SI mouse embryos that did not produce SCF did not support the survival of both +/+ and Ws/Ws RCMC but did induce the RMCP-I+/II+ phenotype in about half of +/+ and Ws/Ws RCMC when their survival was supported by the addition of ConA-SCM. The normal signal transduction through the c-kit receptor did not appear to be prerequisite for the acquisition of the RMCP-I+/II+ phenotype.


Subject(s)
Mast Cells/cytology , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Colony-Stimulating Factor/genetics , Sequence Deletion , Animals , Antibodies, Monoclonal/pharmacology , Bone Marrow Cells , Cell Adhesion Molecules/pharmacology , Cell Division/drug effects , Cells, Cultured , Concanavalin A , Culture Media, Conditioned , Endopeptidases/biosynthesis , Endopeptidases/metabolism , Fibroblasts/physiology , Fluorescent Antibody Technique , Hematopoietic Cell Growth Factors/pharmacology , Kinetics , Lymphocyte Activation , Lymphocytes/immunology , Lymphocytes/physiology , Mast Cells/drug effects , Mast Cells/enzymology , Nerve Growth Factors/immunology , Nerve Growth Factors/physiology , Phenotype , Protein-Tyrosine Kinases/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-kit , Rats , Rats, Mutant Strains , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Colony-Stimulating Factor/biosynthesis , Spleen/immunology , Spleen/physiology , Stem Cell Factor , Time Factors
9.
Oncogene ; 9(1): 157-62, 1994 Jan.
Article in English | MEDLINE | ID: mdl-7508100

ABSTRACT

The W locus of mice encodes the c-kit receptor tyrosine kinase. Recently, we characterized a novel mutant allele, Wn, and demonstrated that the c-kit protein synthesized in Wn/Wn cultured mast cells (CMC) was reduced in size and not expressed on their surface (Tsujimura et al., 1993). In this study, we further examined biochemical nature of the mutant form of c-kit protein, by using Wn/Wn CMC and 293T cells transfected with Wn-type c-kit cDNA (c-kitWn). The c-kit product synthesized in Wn/Wn CMC was truncated almost all cytoplasmic domain and was less glycosylated. In c-kitWn-transected cells, both glycosylation and extracellular expression of c-kit protein was also impaired, however, no truncation was detected. These results indicate that Wn-mutant form of c-kit product is insufficient in maturation, which is associated with impairments in the transport to the plasma membrane, and retention of the mutant protein in endoplasmic reticulum is suggested. This is the first demonstration of the c-kit mutation affecting posttranslational processing its product.


Subject(s)
Alleles , Point Mutation , Protein Processing, Post-Translational , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Colony-Stimulating Factor/metabolism , Animals , Cells, Cultured , Mast Cells/chemistry , Mice , Protein Conformation , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-kit , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Colony-Stimulating Factor/chemistry , Receptors, Colony-Stimulating Factor/genetics , Transfection
10.
Exp Hematol ; 21(13): 1686-94, 1993 Dec.
Article in English | MEDLINE | ID: mdl-7694869

ABSTRACT

The interaction of the proto-oncogene c-kit product with its ligand (stem cell factor or SCF) is considered to play crucial roles in hematopoiesis. In a series of human acute myeloblastic leukemia (AML) cells, the effects of recombinant human (rh) SCF on AML cells were examined in short-term and long-term cultures. c-kit expression was detected in 26 of 31 AML cases, and short-term treatment of AML cells with rhSCF led to proliferation in 13 of 18 AML cases that expressed the c-kit product. In seven of the 13 cases showing proliferative response to rhSCF, AML cells were exclusively composed of immature blast cells. We therefore used the seven AML cases for examining the effect of rhSCF on the differentiation and proliferation of AML cells in a long-term culture. Proliferation of AML cells was found to be maintained with rhSCF more than 2 weeks in five of seven cases and 4 weeks in two cases, whereas most of the AML cells died before 2 weeks in the absence of rhSCF. Further, in four of five AML cases, all of which expressed the CD34 antigen and showed a proliferative response to rhSCF in a long-term culture, rhSCF appeared to promote differentiation of blast cells toward lineages of various cell types, such as granulocytic and/or monocytic and mast-cell lineages. These results suggest that, at least in a fraction of AML cases, rhSCF can induce not only proliferation but also differentiation of AML cells, and also that phenotypic manifestation of AML cells may not mean definite cell commitment but can be changed by stimulation with rhSCF.


Subject(s)
Hematopoietic Cell Growth Factors/pharmacology , Leukemia, Myeloid, Acute/pathology , Antigens, CD/analysis , Antigens, CD34 , Antigens, Differentiation, Myelomonocytic/analysis , CD13 Antigens , Cell Differentiation , Cell Division , Cytoplasmic Granules/pathology , Gene Expression , HLA-DR Antigens/analysis , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/immunology , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-kit , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Colony-Stimulating Factor/genetics , Recombinant Proteins/pharmacology , Stem Cell Factor , Tumor Cells, Cultured
11.
J Clin Invest ; 92(4): 1736-44, 1993 Oct.
Article in English | MEDLINE | ID: mdl-7691885

ABSTRACT

The c-kit proto-oncogene encodes a receptor tyrosine kinase. Binding of c-kit ligand, stem cell factor (SCF) to c-kit receptor (c-kitR) is known to activate c-kitR tyrosine kinase, thereby leading to autophosphorylation of c-kitR on tyrosine and to association of c-kitR with substrates such as phosphatidylinositol 3-kinase (PI3K). In a human mast cell leukemia cell line HMC-1, c-kitR was found to be constitutively phosphorylated on tyrosine, activated, and associated with PI3K without the addition of SCF. The expression of SCF mRNA transcript in HMC-1 cells was not detectable by means of PCR after reverse transcription (RT-PCR) analysis, suggesting that the constitutive activation of c-kitR was ligand independent. Sequencing of whole coding region of c-kit cDNA revealed that c-kit genes of HMC-1 cells were composed of a normal, wild-type allele and a mutant allele with two point mutations resulting in intracellular amino acid substitutions of Gly-560 for Val and Val-816 for Asp. Amino acid sequences in the regions of the two mutations are completely conserved in all of mouse, rat, and human c-kit. In order to determine the causal role of these mutations in the constitutive activation, murine c-kit mutants encoding Gly-559 and/or Val-814, corresponding to human Gly-560 and/or Val-816, were constructed by site-directed mutagenesis and expressed in a human embryonic kidney cell line, 293T cells. In the transfected cells, both c-kitR (Gly-559, Val-814) and c-kitR (Val-814) were abundantly phosphorylated on tyrosine and activated in immune complex kinase reaction in the absence of SCF, whereas tyrosine phosphorylation and activation of c-kitR (Gly-559) or wild-type c-kitR was modest or little, respectively. These results suggest that conversion of Asp-816 to Val in human c-kitR may be an activating mutation and responsible for the constitutive activation of c-kitR in HMC-1 cells.


Subject(s)
Leukemia, Mast-Cell/genetics , Point Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Colony-Stimulating Factor/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , DNA Primers , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Interleukin-3/pharmacology , Mice , Molecular Sequence Data , Phosphotyrosine , Polymerase Chain Reaction , Proto-Oncogene Mas , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-kit , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Colony-Stimulating Factor/biosynthesis , Receptors, Colony-Stimulating Factor/metabolism , Recombinant Proteins/pharmacology , Transfection , Tumor Cells, Cultured , Tyrosine/analogs & derivatives , Tyrosine/analysis
12.
Leuk Lymphoma ; 10(1-2): 35-41, 1993 May.
Article in English | MEDLINE | ID: mdl-7690631

ABSTRACT

The c-kit proto-oncogene encodes a receptor tyrosine kinase that is considered to play important roles in hematopoiesis. The proto-oncogene c-kit product is expressed on various types of human cell lines derived from leukemic cells of erythroid, megakaryocytic and mast-cell lineages. Also, the c-kit product is detectable in blast cells in most cases of acute myeloblastic leukemia (AML) and in some cases of chronic myelogenous leukemia (CML) in blastic crisis (BC). By contrast, little or no expression of c-kit is observed in human leukemia cell lines of lymphoid lineage and in blast cells in acute lymphoblastic leukemia (ALL). Tyrosine phosphorylation and activation of the c-kit product with the ligand for c-kit (stem cell factor: SCF) results in proliferation of some human leukemia cell lines, such as M07E, and blast cells in a substantial fraction of AML cases. In addition, SCF appears to have an activity in inducing differentiation of certain types of leukemic cells. In some cases, further, the c-kit product is found to be activated in leukemic cells even before the stimulation with SCF. These results suggest that c-kit may be involved in excessive proliferation and aberrant differentiation of human leukemia cells.


Subject(s)
Leukemia/pathology , Protein-Tyrosine Kinases/analysis , Proto-Oncogene Proteins/physiology , Cell Adhesion , Cell Differentiation , Cell Division , Gene Expression Regulation, Leukemic , Humans , Leukemia/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-kit , Proto-Oncogenes
13.
Cancer Res ; 53(3): 675-80, 1993 Feb 01.
Article in English | MEDLINE | ID: mdl-8425202

ABSTRACT

Human interleukin 2 (IL-2) is a member of the class of crucial regulators of lymphocyte proliferation. The action of IL-2 is known to be mediated through binding to a specific IL-2 receptor (IL-2R) which comprises at least two distinct proteins: IL-2R alpha (p55) and IL-2R beta (p70-75). However, the expression and function of IL-2R are largely unknown in acute myeloblastic leukemia cells. In a human granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-3, or stem cell factor-dependent myeloid leukemia cell line (M07E), IL-2 was found to stimulate proliferation in a dose-dependent manner and to augment GM-CSF- and stem cell factor-induced proliferation of M07E cells. The expression of IL-2R beta on M07E cells was detectable with 125I-IL-2 binding and affinity cross-linking analyses and with a monoclonal antibody against IL-2R beta, Mik-beta 1. Although the expression of IL-2R beta was not down-regulated but somewhat up-regulated by treatment with GM-CSF in both mRNA and protein levels, GM-CSF was found to compete (75%) with radiolabeled IL-2 for binding to IL-2R on M07E cells, whereas no competition of GM-CSF binding was observed with IL-2 even at a 400-fold molar excess. These results suggest that IL-2R may be functionally expressed in some cases of acute myeloblastic leukemia cells and raise the possibility that IL-2 may have some effects on human myelopoiesis.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interleukin-2/metabolism , Leukemia, Megakaryoblastic, Acute/pathology , Receptors, Interleukin-2/physiology , Binding, Competitive , Cell Division/drug effects , Cross-Linking Reagents/pharmacology , Humans , Interleukin-2/antagonists & inhibitors , Interleukin-2/pharmacology , Iodine Radioisotopes , Kinetics , Leukemia, Megakaryoblastic, Acute/genetics , Leukemia, Megakaryoblastic, Acute/metabolism , RNA, Messenger/genetics , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/metabolism , Sensitivity and Specificity , Transcription, Genetic/genetics , Tumor Cells, Cultured
14.
Int Arch Allergy Immunol ; 100(3): 219-29, 1993.
Article in English | MEDLINE | ID: mdl-8453309

ABSTRACT

The ontogeny of human mast cells (HMCs) is not known, but agranular precursors committed to HMC development are present in cord blood mononuclear cells. Individual cord bloods were cocultured with murine fibroblasts and prepared for light microscopy, immunofluorescent determination of tryptase and electron microscopy after sequential times in culture. Mast cells and their progenitors were identified and quantitated during their evolution from agranular precursors in human cord blood. Each of these methods revealed similar numbers of HMC progenitors. The earliest, visible granule contents were particulate; later, scrolls superimposed on particulate materials were noted. HMC progenitors did not contain crystal granules, but fully mature HMCs present in long-term cocultures did. The developmental sequence for the evolution of the HMC lineage in vitro, made available by this new culture system, is identical to that described for the development of HMCs in vivo in sequentially examined samples of human fetal tissues.


Subject(s)
Mast Cells/cytology , Stem Cells/ultrastructure , Animals , Basophils/cytology , Cell Nucleus/ultrastructure , Cells, Cultured , Cytoplasmic Granules/ultrastructure , Fetal Blood/cytology , Fibroblasts/cytology , Humans , Mice , Microscopy, Electron , Time Factors
15.
Proc Natl Acad Sci U S A ; 90(2): 735-9, 1993 Jan 15.
Article in English | MEDLINE | ID: mdl-7678463

ABSTRACT

Both human and mouse c-kit ligand induced differentiation of human mast cells in a long-term culture of the mononuclear cells of umbilical cord blood. Growth factor activity for human mast cells present in conditioned medium of BALB/3T3 fibroblasts was due to mouse c-kit ligand. Recombinant c-kit ligand induced differentiation and proliferation of mast cell progenitors in early stages of culture. However, apparent selective growth of mast cells by c-kit ligand in cord blood cell cultures is mainly due to the effect of the cytokine to selectively maintain survival of immature mast cells. Electron microscopic analysis indicated that human mast cells developed by c-kit ligand were similar to human mast cells in the lung and gut mucosa, while those developed in coculture of cord blood cells with Swiss albino/3T3 fibroblasts were similar to skin mast cells. This conclusion was supported by the fact that the majority of mast cells developed by c-kit ligand contained only tryptase in their granules, whereas those developed in the cocultures contained both tryptase and chymase. It was also found that mast cells developed by c-kit ligand were immature even after culture for 14 weeks. Nevertheless, these cells express Fc epsilon RI, and could be sensitized with human IgE for anti-IgE-induced release of histamine, prostaglandin D2, and leukotriene C4.


Subject(s)
Cell Differentiation/drug effects , Fetal Blood/cytology , Hematopoietic Cell Growth Factors/pharmacology , Leukocytes, Mononuclear/physiology , Mast Cells/physiology , Animals , Cell Division , Humans , Immunoglobulin E/metabolism , Leukocytes, Mononuclear/drug effects , Mast Cells/cytology , Mice , Mice, Inbred BALB C , Recombinant Proteins/pharmacology , Stem Cell Factor
16.
Int J Oncol ; 2(2): 213-9, 1993 Feb.
Article in English | MEDLINE | ID: mdl-21573539

ABSTRACT

We have investigated the effects of c-kit ligand (stem cell factor [SCF]) and interleukin-3 (IL-3) on proliferation and differentiation of a human chronic myelogenous leukemia cell line, KU812F, which can differentiate toward erythroid and basophilic lineages. When purified c-kit-positive cells (approximately 20% of KU812F cells) were used as a target, SCF induced not only proliferation but also augumented erythroid differentiation of the cells, while IL-3 did promote basophilic differentiation. Further, analyses of in situ hybridization and cell sorting with anti-c-kit antibody showed that the expression of c-kit decreased along with differentiation from immature to mature basophils and erythroid cells.

17.
Blood ; 80(6): 1454-62, 1992 Sep 15.
Article in English | MEDLINE | ID: mdl-1381628

ABSTRACT

Mutant mice of mi/mi genotype are osteopetrotic and deficient in tissue mast cells due to a defect in osteoclasts and mast cells. In an effort to further understand the mechanisms behind why mi/mi mouse-derived cultured mast cells (mi/mi-CMC) responded to interleukin-3 (IL-3), but not to the proliferative stimuli presented by fibroblasts, mi/mi-CMC and congenic normal (+/+) mouse-derived CMC (+/+-CMC), both of which expressed the phenotypic characteristics of immature mast cells, were cocultured with Swiss albino/3T3 fibroblasts in a medium containing IL-3. In the in vitro CMC/fibroblast coculture, mi/mi-CMC did not acquire the phenotypes of connective tissue-type mast cells (CTMC), while +/+-CMC did. In addition, attachment of mi/mi-CMC to the fibroblasts was found to be significantly lower than that of +/+-CMC. Because the interaction of c-kit product with its ligand (stem cell factor [SCF]) is known to play an important role not only in proliferation and differentiation of mast cells but also in attachment of CMC to fibroblasts, the expression and function of c-kit were investigated in mi/mi-CMC and +/+-CMC. Recombinant rat SCF (rrSCF164) induced a dose-dependent proliferation of +/+-CMC. Also, rrSCF164 induced +/+-CMC to acquire the phenotypes of CTMC in the medium containing IL-3. By contrast, rrSCF164 did not stimulate the proliferation of mi/mi-CMC nor induce a phenotypic change of the cells from immature mast cells to mature, CTMC-like mast cells. Immunoblotting with antiphosphotyrosine antibody showed that rrSCF164 induced considerable tyrosine phosphorylation of 145- to 165-Kd protein, the product of c-kit, in +/+-CMC, whereas tyrosine phosphorylation of the protein was barely detectable in mi/mi-CMC. Northern blot and flow cytometry analyses showed that mi/mi-CMC expressed much less c-kit at both protein and message levels than +/+-CMC. Further, mi/mi-CMC were found to differ from +/+-CMC in the expression of mouse mast cell protease-6 (MMCP-6) and MMCP-2 messenger RNA transcripts. These results suggest that the gene product of the mi locus may be important in regulating the expression of gene products such as c-kit, and that mast cell deficiency of mi/mi mice appears to be due, at least in part, to impaired signaling through the c-kit receptor because of the low c-kit expression.


Subject(s)
Mast Cells/cytology , Mice, Mutant Strains/genetics , Osteopetrosis/genetics , Proto-Oncogene Proteins/genetics , 3T3 Cells , Animals , Cell Differentiation , Cell Division , Cells, Cultured , Chymases , Female , Fibroblasts/cytology , Genotype , Male , Mastocytosis/genetics , Mice , Phenotype , Proto-Oncogene Proteins c-kit , RNA, Messenger/analysis , Receptors, Cell Surface/genetics , Serine Endopeptidases/genetics , Transcription, Genetic
18.
J Leukoc Biol ; 51(6): 557-69, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1613392

ABSTRACT

Human mast cells developed in vitro when cord blood mononuclear cells were cocultured for 3 months with 3T3 embryonic mouse skin fibroblasts. The metachromatic cells that arose in these cultures contained histamine, a functional Fc epsilon receptor and granule proteases (tryptase, chymase), and they were definitively identified by the ultrastructural demonstration of crystal granules. We present a detailed ultrastructural analysis of this newly available system for the reliable development of human mast cells in vitro and provide criteria for definitive identification of the mast cell and basophil lineages in humans.


Subject(s)
Fetal Blood/cytology , Leukocytes, Mononuclear/ultrastructure , Mast Cells/ultrastructure , 3T3 Cells , Animals , Cells, Cultured , Endothelium/ultrastructure , Humans , Macrophages/ultrastructure , Mice , Skin/cytology
19.
Am J Pathol ; 140(4): 795-807, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1562046

ABSTRACT

The mechanism of piecemeal degranulation by human eosinophils was investigated. Mature eosinophils that developed in rhIL-5-containing conditioned media from cultured human cord blood mononuclear cells were prepared for ultrastructural studies using a combined technique to image eosinophil peroxidase by cytochemistry in the same sections on which postembedding immunogold was used to demonstrate Charcot-Leyden crystal protein. Vesicular transport of eosinophil peroxidase from the specific granule matrix compartment to the cell surface was associated with piecemeal degranulation. This process involved budding of eosinophil peroxidase-loaded vesicles and tubules from specific granules. Some eosinophil peroxidase that was released from eosinophils remained bound to the cell surface; some was free among the cultured cells. Macrophages and basophils bound the released eosinophil peroxidase to their plasma membranes, internalized it in endocytotic vesicles, and stored it in their respective phagolysosomes and secretory granules. Charcot-Leyden crystal protein was diffusely present in the nucleus and cytoplasm of IL-5-stimulated mature eosinophils. Extensive amounts were generally present in granule-poor and subplasma membrane areas of the cytoplasm in contrast to eosinophil peroxidase, which was secreted and bound to the external surface of eosinophil plasma membranes. These studies establish vesicular transport as a mechanism for emptying the specific eosinophil granule matrix compartment during IL-5-associated piecemeal degranulation.


Subject(s)
Blood Cells/cytology , Eosinophils/cytology , Fetal Blood , Interleukin-5/pharmacology , Monocytes/cytology , Cells, Cultured , Cellular Senescence , Cytoplasmic Granules/metabolism , Eosinophils/metabolism , Glycoproteins/metabolism , Humans , Lysophospholipase , Peroxidases/metabolism , Recombinant Proteins , Tissue Distribution
20.
Am J Pathol ; 139(6): 1309-18, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1750506

ABSTRACT

Culture systems designed to maintain or develop human mast cells have proved difficult, yet these systems would provide valuable resources for future investigations of human mast cell biology. Cocultures of either isolated mature human lung mast cells (Levi-Schaffer et al., J Immunol 1987, 139:494-500) or human cord blood mononuclear cells (Furitsu, Proc Natl Acad Sci USA 1989, 86:10039-10043) with 3T3 embryonic mouse skin fibroblasts have implicated fibroblasts as an important factor in the successful maintenance and development of human mast cells in vitro. The authors cultured isolated, mature human lung mast cells either with or without 3T3 cells for 1 month and examined their ultrastructural phenotype. Mast cell viability in each circumstance was equivalent, but mast cell yield was improved in the presence of 3T3 cells. The ultrastructural phenotype was identical in both culture systems. Mast cells were shown to maintain the phenotype of their in vivo lung counterparts (ie, scroll granules predominanted, and numerous lipid bodies were present). This ultrastructural phenotype differs from that of mast cells that develop in cocultures of human cord blood cells and 3T3 cells, where developing mast cells with crystalline granules and few lipid bodies prevail, a phenotype much like that of human skin mast cells in vivo (Furitsu, Proc Natl Acad Sci USA 1989, 86:10039-10043).


Subject(s)
3T3 Cells/physiology , Cytological Techniques , Fetal Blood/cytology , Lung/cytology , Mast Cells/physiology , 3T3 Cells/ultrastructure , Animals , Cells, Cultured , Cellular Senescence , Humans , Mast Cells/ultrastructure , Mice , Phenotype , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...