Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Zhonghua Yi Xue Za Zhi ; 101(29): 2316-2321, 2021 Aug 03.
Article in Chinese | MEDLINE | ID: mdl-34333948

ABSTRACT

Objective: To explore the application value of single-port laparoscopic and thoracoscopic McKeown esophagectomy for esophageal cancer. Methods: A retrospective study was conducted to collect clinical data of 34 patients with esophageal cancer who were admitted to the Department of Thoracic Surgery, the First Affiliated Hospital of Fujian Medical University, from August to November 2020. All of them, 24 males and 10 females aged from 43 to 75 with an average of (62±4) years, underwent single-port laparoscopic and thoracoscopic McKeown esophagectomy.In the thoracic part, esophageal separation and mediastinal lymph node dissection were performed with conventional 4-ports thoracoscopy in the left lateral-prone position. In the abdominal part, gastric separation and lymph node dissection were accomplished with single-port laparoscopic retrograde three-step gastric separation: firstly, the lesser omentum was dissociated, the left liver lobe was suspended with purse-string needle and thread, after that the left gastric blood vessel and lymph node was dissected; secondly, the esophageal hiatus was separated, and the gastric cardia was cut off; finally, the spleen and stomach ligaments were dissociated to complete these steps. The operation time,volume of intraoperative blood loss, time for out-of-bed activities, time of postoperative drainage tube removal,volume of thoracic drainage fluid, short-term postoperative commplications,the postoperative pathological diagnoses, the time of their discharge from hospital and results of follow-up were observed. Results: All patients underwent successfully single-port laparoscopic and thoracoscopic esophagectomy without any conversion to open surgery; the operative time of the patients was 194-285 (240±21)min, including 53-105(60±13)min for the thoracic part and 40-73(49±7)min for the abdominal part. The volume of intraoperative blood loss was 15-110(60±20) ml. The numbers of mediastinal lymph node dissected and abdominal lymph nodes harvested were 10-25(13±3), 6-16(9±3)respectively. The 34 patients resumed out-of-bed activities on the 2nd to 3rd day after the operation. The thoracic closed drainage tube and left cervical drainage were removed 2 days after the operation. The thoracic Abel drainage tube was removed 5 days after the operation. The total volume of postoperative thoracic drainage fluid was 100-500(300±100)ml. No obvious sign of anastomotic leakage, anastomotic stenosis, chylothorax or gastric emptying disorder was found after the operation. Eight cases were complicated with temporal hoarseness, and 4 patients with pneumonia which was cured by antibiotics. The mean postoperative hospital stay was 8 days(6, 8), and patients were discharged after they could take a routine semi-liquid diet. The postoperative pathological diagnoses of all patients were squamous cell carcinoma, and the postoperative pathological stage was T1-3N0-1M0. Thirty-four patients were followed up for 60 (40, 75) days after the operation. During the follow-up period, no complication or death occurred. In addition, neither recurrence nor metastasis was observed. Conclusion: Single-port laparoscopic and thoracoscopic McKeown esophagectomy for esophageal carcinoma is safe and feasible, with good short-term efficacy. Therefore we consider it an alternative minimally invasive surgery for esophageal cancer.


Subject(s)
Carcinoma, Squamous Cell , Esophageal Neoplasms , Laparoscopy , Carcinoma, Squamous Cell/surgery , Esophageal Neoplasms/surgery , Esophagectomy , Female , Humans , Lymph Node Excision , Male , Postoperative Complications , Retrospective Studies , Thoracoscopy
2.
Cytokine ; 15(4): 188-95, 2001 Aug 21.
Article in English | MEDLINE | ID: mdl-11563879

ABSTRACT

Stem cell factor (SCF) initiates its biological effects by binding to its receptor Kit. Cell surface Kit is proteolytically cleaved to generate soluble Kit. Structure-function analysis of the extracellular region of Kit has implicated the first three immunoglobulin-like domains in SCF binding, and the fourth immunoglobulin-like domain in receptor dimerization. However, the role of the fifth immunoglobulin-like domain is unknown. To test the hypothesis that the fifth immunoglobulin-like domain is important for proteolytic cleavage of Kit from the cell surface, we constructed a mutant form of Kit in which the first four immunoglobulin-like domains are linked to the transmembrane and cytoplasmic domains (designated Kit-Del5). Kit-wild type (Kit-WT) and Kit-Del5 were expressed in the murine mast cell line IC2. Flow cytometry demonstrated that both Kit-WT and Kit-Del5 are displayed on the IC2 cell surface, and immunoblotting confirmed the presence of Kit proteins of the expected molecular weights, 154 kDa and 134 kDa, respectively. Although IC2-Kit-WT cells proteolytically cleave cell surface Kit, generating a 98 kDa soluble form of Kit, IC2-Kit-Del5 cells do not. These findings demonstrate that the fifth immunoglobulin-like domain of Kit is required for proteolytic cleavage of Kit from the cell surface.


Subject(s)
Immunoglobulins/chemistry , Proto-Oncogene Proteins c-kit/chemistry , Animals , Cell Division , Cell Line , Cell Membrane/metabolism , Cytoplasm/metabolism , Flow Cytometry , Gene Deletion , Humans , Mice , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/metabolism , Tetradecanoylphorbol Acetate/pharmacology
3.
Blood ; 98(7): 2077-83, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11567993

ABSTRACT

Cellular trafficking of growth factor receptors, including cross-talk among receptors at the cell surface, may be important for signal transduction in normal hematopoietic cells. To test this idea, the signaling domain of Mpl (the thrombopoietin receptor) was targeted to the plasma membrane, or to the cytoplasm of murine marrow cells, and the ability of the cells to proliferate and differentiate in response to Mpl dimerized at the plasma membrane or free in the cytoplasm was assessed. Constructs encoding the signaling domain of Mpl linked to an FK506 binding protein domain (to permit dimerization by the membrane-permeable ligand AP20187) with or without a myristylation sequence (to target the receptor to the plasma membrane) and a hemagglutinin epitope tag were generated and introduced into murine marrow cells using a murine stem cell virus (MSCV)-based retroviral vector. Both populations of transduced marrow cells proliferated in Iscoves modified Dulbecco medium-10% FCS-100 nM AP20187 without exogenous growth factors for more than 100 days and achieved greater than a 10(7)-fold expansion of cells by day 50 (n = 4 transductions). Growth was dimerizer dependent, and myeloid, erythroid, and megakaryocytic progenitors were generated. Activation of Mpl either at the plasma membrane or in the cytoplasm allowed for the terminal maturation of transduced progenitor cells. Introduction of membrane-targeted or cytoplasmic Mpl into fetal liver cells from homozygous JAK2 knock-out mice or wild-type littermates demonstrated that both forms of Mpl require JAK2 for signaling. These data show that the activation of Mpl independent of its normal plasma membrane location can support production of the full range of normal hematopoietic progenitor cells in vitro.


Subject(s)
Cell Membrane/metabolism , Hematopoietic Stem Cells/drug effects , Milk Proteins , Neoplasm Proteins , Proto-Oncogene Proteins/physiology , Receptors, Cytokine , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line , Cell Membrane/chemistry , Cytoplasm/chemistry , DNA-Binding Proteins/pharmacology , DNA-Binding Proteins/physiology , Dimerization , Hematopoietic Stem Cells/cytology , Janus Kinase 2 , Mice , Microscopy, Fluorescence , Protein Transport , Protein-Tyrosine Kinases/pharmacology , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins/pharmacology , Receptors, Thrombopoietin , STAT5 Transcription Factor , Signal Transduction , Trans-Activators/pharmacology , Trans-Activators/physiology
4.
Leuk Res ; 25(5): 413-21, 2001 May.
Article in English | MEDLINE | ID: mdl-11301110

ABSTRACT

Stem cell factor (SCF) is a growth factor that promotes the survival, proliferation, and differentiation of hematopoietic cells. SCF and its receptor, Kit, are normally present in both cell surface and soluble forms. Both forms of Kit can bind SCF. However, the function of soluble Kit is unknown. In order to determine if soluble Kit can modulate SCF activity, we produced a fusion protein, Kit-Fc, comprised of the extracellular domain of murine Kit and the Fc portion of human IgG(1) and investigated its ability to bind 125I-SCF and to inhibit SCF-stimulated hematopoietic colony growth in vitro. Stable cell lines expressing Kit-Fc were generated and Kit-Fc was purified to greater than 95% purity. Scatchard analysis demonstrated that Kit-Fc binds iodinated SCF with high affinity (Kd 570 pM). Kit-Fc also bound to transmembrane SCF displayed on the surface of fibroblasts. The murine mast cell line IC2 was engineered to express murine Kit on the cell surface and was demonstrated to proliferate in the presence of SCF. Kit-Fc completely blocked SCF-stimulated proliferation of IC2-Kit cells, but not IL-3-stimulated growth of IC2-Kit cells, demonstrating the specificity of Kit-Fc. We investigated the ability of Kit-Fc to block SCF-stimulated murine hematopoietic colony growth. Kit-Fc blocked SCF-stimulated erythroid colony growth as effectively as a neutralizing anti-Kit monoclonal antibody, ACK2, but did not block erythropoietin-stimulated erythroid colony growth. Likewise, Kit-Fc blocked SCF-stimulated myeloid colony growth as effectively as ACK2 antibody, but did not block IL-3- or GM-CSF-stimulated myeloid colony growth. These results indicate that a form of soluble Kit binds SCF with high affinity, and can specifically block the ability of SCF to stimulate hematopoietic colony growth, suggesting that one function of soluble Kit may be to modulate SCF bioactivity.


Subject(s)
Proto-Oncogene Proteins c-kit/pharmacology , Stem Cell Factor/antagonists & inhibitors , 3T3 Cells/metabolism , Animals , Cell Line , Cricetinae , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Immunoglobulin G/pharmacology , Mice , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Solubility , Stem Cell Factor/metabolism , Stem Cell Factor/pharmacology
5.
Blood ; 97(7): 1990-8, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11264163

ABSTRACT

Platelet-derived growth factor (PDGF)-B and PDGF beta-receptor (PDGFR beta) deficiency in mice is embryonic lethal and results in cardiovascular, renal, placental, and hematologic disorders. The hematologic disorders are described, and a correlation with hepatic hypocellularity is demonstrated. To explore possible causes, the colony-forming activity of fetal liver cells in vitro was assessed, and hematopoietic chimeras were demonstrated by the transplantation of mutant fetal liver cells into lethally irradiated recipients. It was found that mutant colony formation is equivalent to that of wild-type controls. Hematopoietic chimeras reconstituted with PDGF-B(-/-), PDGFR beta(-/-), or wild-type fetal liver cells show complete engraftment (greater than 98%) with donor granulocytes, monocytes, B cells, and T cells and display none of the cardiovascular or hematologic abnormalities seen in mutants. In mouse embryos, PDGF-B is expressed by vascular endothelial cells and megakaryocytes. After birth, expression is seen in macrophages and neurons. This study demonstrates that hematopoietic PDGF-B or PDGFR beta expression is not required for hematopoiesis or integrity of the cardiovascular system. It is argued that metabolic stress arising from mutant defects in the placenta, heart, or blood vessels may lead to impaired liver growth and decreased production of blood cells. The chimera models in this study will serve as valuable tools to test the role of PDGF in inflammatory and immune responses. (Blood. 2001;97:1990-1998)


Subject(s)
Blood Vessels/abnormalities , Fetal Diseases/genetics , Hematopoiesis/physiology , Proto-Oncogene Proteins c-sis/physiology , Receptor, Platelet-Derived Growth Factor beta/physiology , Anemia/embryology , Anemia/genetics , Anemia/metabolism , Animals , Blood Vessels/embryology , Bone Marrow Transplantation , Embryonic and Fetal Development/genetics , Erythroblastosis, Fetal/genetics , Erythroblastosis, Fetal/metabolism , Female , Fetal Diseases/blood , Fetal Diseases/pathology , Fetal Heart/abnormalities , Fetal Tissue Transplantation , Genes, Lethal , Genetic Complementation Test , Genotype , Hematopoietic Stem Cell Transplantation , Inflammation , Kidney/abnormalities , Kidney/embryology , Liver/cytology , Liver/embryology , Male , Megakaryocytes/cytology , Mice , Mice, Knockout , Neovascularization, Physiologic/genetics , Placenta/physiopathology , Pregnancy , Proto-Oncogene Proteins c-sis/deficiency , Proto-Oncogene Proteins c-sis/genetics , Radiation Chimera , Receptor, Platelet-Derived Growth Factor beta/deficiency , Receptor, Platelet-Derived Growth Factor beta/genetics , Specific Pathogen-Free Organisms , Stress, Physiological/embryology , Stress, Physiological/genetics , Stress, Physiological/metabolism
6.
Blood ; 94(6): 1979-86, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10477727

ABSTRACT

Stem cell factor (SCF) exerts its biological effects by binding to a specific receptor, the tyrosine kinase c-Kit, which is expressed on the cell surface. Although normal cellular trafficking of growth factor receptors may play a critical role in the modulation of receptor function, the mechanisms that regulate the distribution of c-Kit on the cell surface and the internalization of c-Kit have not been fully defined. We investigated whether signal transduction via Src family kinases is required for normal c-Kit trafficking. Treatment of the SCF-responsive human hematopoietic cell line MO7e with the inhibitor of Src family kinases PP1 blocked SCF-induced capping of c-Kit and internalization of c-Kit. c-Kit was able to associate with clathrin in the presence of PP1, suggesting that entry of c-Kit into clathrin-coated pits occurs independently of Src family kinases. SCF-induced internalization of c-Kit was also diminished in the D33-3 lymphoid cell line in which expression of Lyn kinase was disrupted by homologous recombination. These results indicate that Src family kinases play a role in ligand-induced trafficking of c-Kit.


Subject(s)
Coated Pits, Cell-Membrane/physiology , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction/physiology , Stem Cell Factor/physiology , src-Family Kinases/metabolism , Cell Membrane/physiology , Chemotaxis/drug effects , Chemotaxis/physiology , Clathrin/metabolism , Gene Expression Regulation, Enzymologic , Humans , Kinetics , Leukemia , Proto-Oncogene Proteins c-kit/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Recombination, Genetic , Stem Cell Factor/pharmacology , Tumor Cells, Cultured , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
7.
Blood ; 91(3): 898-906, 1998 Feb 01.
Article in English | MEDLINE | ID: mdl-9446650

ABSTRACT

Stem cell factor (SCF) binding to the c-kit receptor triggers homodimerization and intermolecular tyrosine phosphorylation of the c-kit receptor, thus initiating signal transduction. Receptor dimerization is a critical early step in this process. Prior biochemical studies of c-kit receptor dimerization have mainly used affinity cross-linking techniques, which are beset with problems including low efficiency of cross-linking and the usual requirement for radiolabeled SCF to detect the cross-linked complex. We used the fluorescence resonance energy transfer (FRET) technique to examine the effects of SCF and other hematopoietic cytokines on c-kit receptor dimerization. The nonneutralizing anti-c-kit receptor monoclonal antibody 104D2 was directly conjugated to fluorescein isothiocyanate (FITC) or to the carbocyanine dye Cy3 and used to label cytokine-responsive human hematopoietic cell lines. The ability of SCF to induce c-kit receptor dimerization was assessed by flow cytometric analysis of FRET between the donor fluorochrome FITC and the acceptor fluorochrome Cy3. SCF induced a dose-dependent increase in c-kit receptor dimerization that correlated well with the concentrations of SCF required to stimulate cell proliferation. Receptor dimerization was detectable within 3 minutes after the addition of SCF and was maximal 30 minutes after the addition of SCF. Confocal microscopy showed redistribution of the c-kit receptor (from a diffuse distribution on the cell surface to "caps" at one end of the cell) within 3 minutes after SCF addition, followed by receptor internalization. Reappearance of the c-kit receptor on the cell surface required new protein synthesis, suggesting that the c-kit receptor is not recycled to the cell surface after internalization. Finally, erythropoietin (Epo), but not the structurally and functionally related cytokine thrombopoietin (Tpo), stimulated c-kit receptor dimerization detectable by FRET, and tyrosine phosphorylation of the c-kit receptor. These results suggest that exposure to Epo can activate the c-kit receptor and provide further evidence for cross-talk between the Epo and c-kit receptors in human hematopoietic cell lines. Studies with progeny of burst-forming unit-erythroid (BFU-E) suggest that the FRET technique is sufficiently sensitive to detect c-kit receptor dimerization on normal human hematopoietic cells.


Subject(s)
Proto-Oncogene Proteins c-kit/chemistry , Carbocyanines , Cell Division , Cell Line , Cytokines/pharmacology , Dimerization , Flow Cytometry , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Hematopoietic Stem Cells/chemistry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Kinetics , Microscopy, Confocal , Spectrometry, Fluorescence , Stem Cell Factor/metabolism , Stem Cell Factor/pharmacology
8.
Blood ; 89(6): 1896-904, 1997 Mar 15.
Article in English | MEDLINE | ID: mdl-9058709

ABSTRACT

Thrombopoietin (Tpo) is a major regulator of megakaryopoiesis both in vivo and in vitro. Tpo initiates its biologic effects by binding to the Mpl receptor, which is a member of the hematopoietin receptor family. To define the Tpo binding characteristics of the Mpl receptor, we iodinated purified 70-kD recombinant human Tpo using the Bolton-Hunter reagent. Autoradiographic analysis of (125)I-Tpo binding to normal human marrow mononuclear cells showed many grains specifically associated with megakaryocytes; there were no grains specifically associated with myeloblasts or erythroblasts. Equilibrium binding experiments with (125)I-Tpo and normal human platelets showed a single class of high-affinity receptors (kd, 190 pmol/L) with approximately 30 Mpl receptors per platelet. Affinity cross-linking with (125)I-Tpo showed that the Mpl receptor on platelets is of molecular weight approximately 98 kD. Despite their sequence similarity, erythropoietin and Tpo did not cross-compete for binding to BaF3 cells engineered to coexpress Mpl receptor and erythropoietin receptor. Progeny of normal human burst-forming units-erythroid (BFU-E) contained Mpl receptor mRNA, and flow cytometric analysis showed the presence of Mpl receptor protein on the surface of these cells. These data indicate that display of the Mpl receptor is not limited to the megakaryocytic lineage, but also includes progeny of BFU-E. Like receptors for other hematopoietic cytokines, the binding affinity of the Mpl receptor for Tpo is high, with relatively few receptors displayed per cell. These results suggest that the effects of Tpo to speed red blood cell recovery after myelosuppressive therapy in vivo and to enhance colony-forming unit-erythroid generation in vitro may be mediated by direct interaction of Tpo and erythroid progenitor cells.


Subject(s)
Blood Platelets/metabolism , Neoplasm Proteins , Proto-Oncogene Proteins/biosynthesis , Receptors, Cytokine , Thrombopoietin/blood , Adult , Autoradiography , Binding, Competitive , Bone Marrow Cells , Cell Differentiation , Cell Line , Colony-Forming Units Assay , Erythroid Precursor Cells/metabolism , Humans , Protein Binding , Proto-Oncogene Proteins/metabolism , Receptors, Thrombopoietin
9.
Blood ; 88(9): 3383-90, 1996 Nov 01.
Article in English | MEDLINE | ID: mdl-8896403

ABSTRACT

FLT3 ligand is a hematopoietic growth factor that plays a key role in growth of primitive hematopoietic cells. FLT3 receptor mRNA is found in early hematopoietic progenitors and in human myeloid leukemia blasts. Much less is known about the surface expression of FLT3 receptor on human hematopoietic cells. Using human 125I-FLT3 ligand, we have identified and characterized surface FLT3 receptors on normal and malignant human hematopoietic cells and cell lines. Our results showed that surface display of FLT3 receptor was greatest in fresh myeloid leukemia blast cells and myeloid leukemia cell lines. Erythroleukemic and megakaryocytic leukemia cell lines (n = 5) bound little to no 125I-FLT3 ligand. Scatchard analysis of 125I-FLT3 ligand binding data shows that three myeloid leukemia cell lines, ML-1, AML-193, and HL-60, as well as normal human marrow mononuclear cells, exhibit high affinity FLT3 receptors. Crosslinking of 125I-FLT3 ligand to FLT3 receptors on the surface of ML-1 myeloid leukemia cells indicates that the FLT3 ligand. The rates of FLT3 ligand internalization and degradation were determined by binding 125I-FLT3 ligand to ML-1 cells and acid stripping to distinguish surface bound from internalized ligand. Internalized 125I-FLT3 ligand was detected within 5 minutes after binding to ML-1 cells. In addition, we evaluated the effect of FLT3 ligand on megakaryocytic colony growth and nuclear endoreduplication, alone or in the presence of thrombopoietin. FLT3 ligand did not promote colony forming unit megakaryocyte (CFU-Meg) colony growth or megakaryocyte nuclear maturation, nor did FLT3 ligand augment the effects of thrombopoietin on these measures of megakaryopoiesis. These data indicate that the FLT3 receptor shares several characteristics with the c-kit receptor including dimerization and rapid internalization. However, the more restricted cellular distribution of the FLT3 receptor may target the effects of FLT3 ligand to primitive hematopoietic cells and to myeloid and lymphoid progenitor cells, in contrast to the pleiotropic effects of the c-kit receptor ligand, stem cell factor.


Subject(s)
Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid/metabolism , Receptor Protein-Tyrosine Kinases/biosynthesis , Autoradiography , Cell Line , Humans , Ligands , Radioligand Assay , Receptor Protein-Tyrosine Kinases/analysis
10.
Blood ; 88(6): 2026-32, 1996 Sep 15.
Article in English | MEDLINE | ID: mdl-8822921

ABSTRACT

Thrombopoietin (Tpo), the ligand for the c-Mpl receptor, is a major regulator of megakaryopoiesis. Treatment of mice with Tpo raises the platelet count fourfold within a few days. Conversely, c-mpl knock-out mice have platelet counts that are 15% that of normal. The subunit structure of the c-Mpl receptor is not fully understood. Some cytokines that stimulate megakaryopoiesis (IL-6, IL-11, leukemia inhibitory factor, and oncostatin M) bind to receptors that use gp130 as a signal transduction subunit. For these reasons, we determined whether gp130 function was required for Tpo-induced signal transduction. Murine marrow cells were cultured in semi-solid media in the presence of Tpo or IL-3, with or without a neutralizing anti-gp130 monoclonal antibody (RX187) or a soluble form of c-Mpl receptor (soluble Mpl) that blocks Tpo bioactivity, and the numbers of colony-forming unit-megakaryocyte (CFU-Meg) colonies were counted on day 5. Murine marrow cells were also cultured in suspension under serum-free conditions for 5 days, and megakaryocyte DNA content was measured by flow cytometry, as an index of nuclear maturation. The addition of RX187 did not block Tpo-induced CFU-Meg colony growth nor CFU-Meg nuclear maturation in suspension culture. However, IL-3-induced CFU-Meg colony growth and megakaryocyte nuclear maturation decreased in the presence of RX187. Soluble Mpl completely ablated Tpo-induced CFU-Meg growth, and partially blocked IL-3-stimulated CFU-Meg growth. Thus the effects of Tpo on megakaryopoiesis in vitro do not depend on cytokines that signal through gp130. Furthermore, it is unlikely that gp 130 serves as a beta chain for the c-Mpl receptor, as Tpo signalling is unimpaired in the presence of RX187. In contrast, the effects of IL-3 on CFU-Meg growth are mediated in part through Tpo and through gp130-signalling cytokines.


Subject(s)
Antigens, CD/physiology , Hematopoiesis/drug effects , Megakaryocytes/cytology , Membrane Glycoproteins/physiology , Neoplasm Proteins , Proto-Oncogene Proteins/physiology , Receptors, Cytokine , Thrombopoietin/pharmacology , Animals , Cell Differentiation/drug effects , Colony-Forming Units Assay , Cytokine Receptor gp130 , DNA/metabolism , Heterozygote , Immunologic Techniques , Interleukin-3/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Receptors, Thrombopoietin , Signal Transduction
11.
Blood ; 88(1): 75-81, 1996 Jul 01.
Article in English | MEDLINE | ID: mdl-8704204

ABSTRACT

The phenotypes of mice that harbor a defect in the genes encoding either stem cell factor (SCF) or its receptor, c-kit, indicate that this ligand/receptor pair is necessary for maintenance of normal hematopoiesis in the adult. Our objective was to determine whether SCF, like erythropoietin, is necessary for acute erythroid expansion during recovery from hemolytic anemia. Monoclonal antibody ACK2, which recognizes the murine c-kit receptor, was used to selectively block the hematopoietic growth-promoting effects of SCF. Mice were treated with phenylhydrazine on day 0 and day 1 to induce hemolytic anemia and also received no antibody, control IgG, or ACK2 on day 0. The mice were killed on day 3 and the hematocrit (Hct), reticulocyte count, and numbers of erythroid and myeloid hematopoietic progenitor cells (colony-forming unit-erythroid [CFU-E], burst-forming unit [BFU]-E, and CFU-granulocyte-macrophage [GM]) were quantitated in the femoral marrow and spleen using hematopoietic colony-forming assays. Induction of hemolytic anemia with phenylhydrazine resulted in a drop in the Hct from approximately 50% to 30%, and an approximate 8- to 10-fold increase in the reticulocyte count. The numbers of CFU-E increased modestly in the femur, and approximately 25- to 50-fold in the spleen, in comparison with normal mice. BFU-E and CFU-GM values did not increase in the femur but expanded 6- to 10-fold in the spleen, in comparison with normal mice. This confirms that much of the erythroid expansion in response to hemolytic anemia occurs in the murine spleen. Neutralizing quantities of the ACK2 antibody reduced femoral CFU-E, BFU-E, and CFU-GM content to less than half that found in phenylhydrazine-treated control mice and nearly totally ablated splenic hematopoiesis. These results suggest that c-kit receptor function may be required for optimal response to acute erythropoietic demand and that erythropoiesis in the splenic microenvironment is more dependent on SCF/c-kit receptor interaction than is erythropoiesis in the marrow microenvironment. Because expansion of late erythropoiesis in the spleen was preferentially blocked, we tested the hypothesis that homing of more primitive hematopoietic cells to the spleen was dependent on c-kit receptor function. Lethally irradiated mice were injected with marrow cells obtained from mice that had received phenylhydrazine plus control IgG or with marrow cells obtained from mice that had received phenylhydrazine plus ACK2. In parallel experiments, normal murine marrow cells were treated in vitro with control IgG or with ACK2 and were injected into lethally irradiated mice. The fraction of BFU-E and CFU-GM retrieved from the marrow and spleen of the recipient mice 4 hours later was reduced by approximately 75% when progenitor cells had been exposed to ACK2, in comparison with control IgG. These data suggest that interaction of SCF with the c-kit receptor affects the homing behavior of hematopoietic progenitor cells in the adult animal.


Subject(s)
Hematopoietic Stem Cells/pathology , Proto-Oncogene Proteins c-kit/physiology , Spleen/pathology , Stem Cell Factor/physiology , Anemia, Hemolytic/chemically induced , Anemia, Hemolytic/pathology , Animals , Antibodies, Monoclonal/pharmacology , Bone Marrow/pathology , Cell Movement/physiology , Colony-Forming Units Assay , Erythropoiesis/physiology , Female , Hematopoietic Stem Cells/metabolism , Immunoglobulin G/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Phenotype , Phenylhydrazines/toxicity , Proto-Oncogene Proteins c-kit/drug effects , Radiation Chimera , Rats , Stem Cell Factor/antagonists & inhibitors
12.
Blood ; 85(8): 2052-8, 1995 Apr 15.
Article in English | MEDLINE | ID: mdl-7536489

ABSTRACT

Stem cell factor (SCF) triggers cell growth by binding to cell surface c-kit receptors. Soluble forms of several cytokine receptors have been described and may play a role in the modulation of cytokine activity in vivo. For these reasons, we investigated whether human hematopoietic cells produce soluble c-kit receptors. The human leukemia cell lines OCIM1 and MO7e display approximately 80,000 and approximately 35,000 high-affinity cell surface c-kit receptors, respectively. Soluble c-kit receptors were detected by enzyme immunoassay in OCIM1 and MO7e culture supernatants. We determined the molecular weight and binding affinity of soluble c-kit receptor produced by OCIM1 cells, soluble c-kit receptor purified from human serum, and recombinant soluble c-kit receptor expressed in CHO cells. The three soluble c-kit receptors each have a molecular weight of 98 kD. Quantitative binding experiments with 125I-SCF indicate that the soluble c-kit receptors obtained from human serum or OCIM1 cells have binding affinities for SCF of approximately 200 to 300 pmol/L, in contrast to the recombinant form, which has a binding affinity of approximately 1.5 nmol/L. All three forms of the soluble c-kit receptor were able to compete with c-kit receptors on OCIM1 cells for 125I-SCF binding. Thus human hematopoietic cells can produce a soluble form of the c-kit receptor that retains high-affinity SCF binding activity. We speculate that the soluble c-kit receptor may bind SCF and function as a receptor antagonist in vivo.


Subject(s)
Hematopoietic Stem Cells/metabolism , Proto-Oncogene Proteins/isolation & purification , Receptor Protein-Tyrosine Kinases/isolation & purification , Receptors, Colony-Stimulating Factor/isolation & purification , Amino Acid Sequence , Base Sequence , Binding, Competitive , Burkitt Lymphoma/pathology , Hematopoietic Cell Growth Factors/metabolism , Humans , Leukemia, Erythroblastic, Acute/pathology , Leukemia, Megakaryoblastic, Acute/pathology , Molecular Sequence Data , Molecular Weight , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/chemistry , Neoplasm Proteins/isolation & purification , Neoplastic Stem Cells/metabolism , Protein Binding , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-kit , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/chemistry , Receptors, Colony-Stimulating Factor/biosynthesis , Receptors, Colony-Stimulating Factor/chemistry , Recombinant Proteins/metabolism , Solubility , Stem Cell Factor , Tumor Cells, Cultured
13.
Blood ; 85(7): 1719-26, 1995 Apr 01.
Article in English | MEDLINE | ID: mdl-7535585

ABSTRACT

Thrombopoietin (Tpo), the ligand for the c-mpl receptor, is a major regulator of platelet production in vivo. Treatment of mice with purified recombinant Tpo increases platelet count fourfold and expands colony-forming unit-megakaryocyte (CFU-Meg) numbers. Other cytokines including interleukin-3 (IL-3), IL-6, IL-11, erythropoietin (Epo), and stem cell factor (SCF) can stimulate megakaryopoiesis. Therefore, we examined the effects of recombinant murine Tpo in combination with these cytokines on megakaryopoiesis in vitro. Murine marrow cells were cultured in agar in Iscove's modified Dulbecco's medium (IMDM) supplemented with 10% horse serum and beta-mercaptoethanol in the presence of recombinant growth factors, and CFU-Meg colonies were counted on day 5. Megakaryocyte ploidy was analyzed using murine marrow cells cultured for 5 days in IMDM supplemented with 1% nutridoma-SP and recombinant growth factors. Megakaryocytes were identified by labeling with the 4A5 antibody and ploidy was analyzed by flow cytometry. Tpo supported the growth of CFU-Meg in a dose-dependent manner. Although the addition of SCF (50 ng/mL), Epo (2 U/mL), or IL-11 (50 ng/mL) alone exerted only a modest effect on CFU-Meg growth, the combination of SCF plus Tpo, Epo plus Tpo, or IL-11 plus Tpo resulted in a synergistic enhancement of the number of CFU-Meg colonies. IL-3 alone supported CFU-Meg colony growth, and the effects of IL-3 plus Tpo or IL-6 plus Tpo on colony growth appeared to be approximately additive. Fifty percent of megakaryocytes generated in cultures containing IL-3 or Epo displayed < or = 16 N ploidy. In contrast, cultures containing Tpo uniquely generated large numbers (30% to 35% of the total) of megakaryocytes with > or = 64N ploidy. These results show that Tpo stimulates both proliferation of committed megakaryocytic progenitor cells and maturation of megakaryocytes, and that two multipotent cytokines, SCF and IL-11, as well as a late-acting erythroid cytokine, Epo, can synergize with Tpo to stimulate proliferation of CFU-Meg.


Subject(s)
Erythropoietin/pharmacology , Hematopoietic Cell Growth Factors/pharmacology , Interleukin-11/pharmacology , Megakaryocytes/drug effects , Polyploidy , Thrombopoietin/pharmacology , Animals , Cell Division/drug effects , Cells, Cultured , Colony-Forming Units Assay , Culture Media, Serum-Free , Drug Synergism , Interleukin-3/pharmacology , Mice , Recombinant Proteins/pharmacology , Stem Cell Factor
14.
J Clin Invest ; 90(5): 1879-88, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1385477

ABSTRACT

IL-3 is a glycoprotein cytokine involved in the hematopoietic response to infectious, immunologic, and inflammatory stimuli. In addition, clinical administration of recombinant IL-3 augments recovery in states of natural and treatment-related marrow failure. IL-3 acts by binding to high affinity cell surface receptors present on hematopoietic cells. To determine the site(s) at which IL-3 binds to it receptor, we analyzed a series of interspecies chimera of the growth factor for species-specific receptor binding and biological activity. The results suggest that IL-3 binds to its receptor and triggers a proliferative stimulus through two noncontiguous helical domains located near the amino terminus and the carboxy terminus of the molecule. To corroborate these findings, we have also mapped the binding epitopes of 10 mAb of human or murine IL-3, and have defined four distinct epitopes. Two of these epitopes comprise the amino-terminal receptor binding domain. A third epitope corresponds to the carboxy-terminal receptor interactive domain, and the fourth epitope, apparently not involved in the interaction of IL-3 and its receptor, lies between these sites. And on the basis of sandwich immunoassays using pairs of these mAbs, the two receptor interactive regions appear to reside in close juxtaposition in the tertiary structure of the molecule. These results provide a correlation of the structure-function relationships of IL-3 that should prove useful in evaluating the details of IL-3-IL-3 receptor interaction and in the rational design of clinically useful derivatives of this growth factor.


Subject(s)
Interleukin-3/pharmacology , Recombinant Fusion Proteins/pharmacology , Adult , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Cross Reactions , Epitopes/analysis , Humans , Hylobates , Interleukin-3/chemistry , Interleukin-3/metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Rats , Rats, Inbred Lew , Receptors, Interleukin-3/metabolism , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...