Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Leukemia ; 15(8): 1286-92, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11480573

ABSTRACT

Thymic stromal lymphopoietin (TSLP) is a novel cytokine that was found to promote the development of murine B cells in vitro. Here we describe the cloning and characterization of the human homologue of murine TSLP. This protein, which is expressed in a number of tissues including heart, liver and prostate, prevented apoptosis and stimulated growth of the human acute myeloid leukemia (AML)-derived cell line MUTZ-3. Anti-interleukin (IL)-7 receptor antibodies (Abs) neutralized this effect indicating that TSLP binds to at least part of the IL-7 receptor complex. TSLP induced phosphorylation of signal transducer and activator of transcription (STAT)-5. In contrast to IL-7, TSLP-triggered STAT-5 phosphorylation was not preceded by activation of janus kinase (JAK) 3. These findings would be in accordance with the notion, raised previously for the mouse system, that TSLP leads to STAT-5 phosphorylation by activating other kinases than the JAKs. Some other signaling pathways stimulated by many cytokines are not involved in TSLP activity; thus, TSLP did not stimulate activation of ERK1,2 and p70S6K. Furthermore, neutralizing Abs raised against cytokines known to stimulate the growth of MUTZ-3 cells did not inhibit the proliferative effects of TSLP, suggesting that TSLP-induced growth was a direct effect. In summary, we describe the cloning of human TSLP and its proliferative effects on a myeloid cell line. TSLP-induced proliferation is preceded by phosphorylation of STAT-5, but not of JAK 3.


Subject(s)
Cytokines/genetics , Milk Proteins , Amino Acid Sequence , Base Sequence , Cloning, Molecular , Cytokines/analysis , Cytokines/pharmacology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Molecular Sequence Data , Phosphorylation , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , STAT5 Transcription Factor , Signal Transduction/drug effects , Signal Transduction/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Thymic Stromal Lymphopoietin
2.
Br J Haematol ; 109(1): 211-20, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10848802

ABSTRACT

Aplastic anaemia (AA) is an immune-mediated bone marrow failure associated with high serum levels of flt3 ligand (FL). We examined expression of the membrane-bound isoform of FL in peripheral blood and bone marrow cells from AA patients at diagnosis (n = 16) and after immunosuppressive (IS) treatment (n = 36). Flow cytometry demonstrated strongly increased FL levels on the cell surface of T lymphocytes in AA relative to normal controls (P < 0.0001). T-cell-specific expression of membrane-bound FL was confirmed by confocal microscopy. FL mRNA and total cellular FL protein levels were increased about threefold. Overexpression of FL in AA was observed for up to 20 years after IS treatment. FL levels correlated inversely with CD34+ cell numbers and the colony-forming ability of AA bone marrow (R = -0.68 and -0.85 respectively). Histological examination of spleen specimens and bone marrow biopsies gave no evidence of degeneration or fibrosis due to prolonged exposure to high FL. Levels of membrane-bound FL were not increased in autoimmune diseases (n = 23), including rheumatoid arthritis and lupus erythematosus, nor in graft-versus-host disease (n = 8). Chronic overexpression of FL on the surface of T lymphocytes in AA, but not in other T-cell-mediated disorders, suggests that membrane-bound FL plays a role in cell-cell interactions in bone marrow failure and may be important for long-term haemopoietic recovery.


Subject(s)
Anemia, Aplastic/metabolism , Membrane Proteins/metabolism , T-Lymphocytes/metabolism , Adolescent , Adult , Aged , Anemia, Aplastic/pathology , Anemia, Aplastic/therapy , Blotting, Western , Bone Marrow/pathology , Case-Control Studies , Child , Colony-Forming Units Assay , Female , Fibrosis , Flow Cytometry/methods , Humans , Immunosuppressive Agents/therapeutic use , Male , Membrane Proteins/analysis , Membrane Proteins/therapeutic use , Microscopy, Confocal , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction/methods , Spleen/pathology , Statistics, Nonparametric
3.
Blood ; 95(11): 3489-97, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10828034

ABSTRACT

The ligand for the receptor tyrosine kinase fms-like tyrosine kinase 3 (flt3), also referred to as fetal liver kinase-2 (flk-2), has an important role in hematopoiesis. The flt3 ligand (flt3L) is a growth factor for hematopoietic progenitors and induces hematopoietic progenitor and stem cell mobilization in vivo. In addition, when mice are treated with flt3L immature B cells, natural killer (NK) cells and dendritic cells (DC) are expanded in vivo. To further elucidate the role of flt3L in hematopoiesis, mice lacking flt3L (flt3L-/-) were generated by targeted gene disruption. Leukocyte cellularity was reduced in the bone marrow, peripheral blood, lymph nodes (LN), and spleen. Thymic cellularity, blood hematocrit, and platelet numbers were not affected. Significantly reduced numbers of myeloid and B-lymphoid progenitors were noted in the BM of flt3L-/- mice. In addition a marked deficiency of NK cells in the spleen was noted. DC numbers were also reduced in the spleen, LN, and thymus. Both myeloid-related (CD11c(++) CD8alpha(-)) and lymphoid-related (CD11c(++) CD8alpha(+)) DC numbers were affected. We conclude that flt3L has an important role in the expansion of early hematopoietic progenitors and in the generation of mature peripheral leukocytes.


Subject(s)
B-Lymphocytes/cytology , Dendritic Cells/cytology , Hematopoiesis/physiology , Hematopoietic Stem Cells/immunology , Killer Cells, Natural/cytology , Membrane Proteins/physiology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Bone Marrow/immunology , Colony-Forming Units Assay , Dendritic Cells/drug effects , Dendritic Cells/immunology , Genomic Library , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Interleukin-7/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Kinetics , Leukocytes/cytology , Ligands , Lymph Nodes/immunology , Lymphocyte Culture Test, Mixed , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly I-C/pharmacology , Recombinant Proteins/pharmacology , Spleen/immunology , Thymus Gland/immunology
4.
J Biol Chem ; 275(5): 3431-7, 2000 Feb 04.
Article in English | MEDLINE | ID: mdl-10652336

ABSTRACT

CD7 is a 40-kDa protein found primarily on T, NK, and pre-B cells; the function of the CD7 protein in the immune system is largely unknown. The K12 (SECTM1) protein was originally identified by its location just upstream of the CD7 locus. The K12 gene encodes a transmembrane protein of unknown function. In order to clone a K12-binding protein, we generated a soluble version of the human K12 protein by fusing its extracellular domain to the Fc portion of human IgG(1). Flow cytometry experiments showed that the K12-Fc fusion protein bound at high levels to both human T and NK cells. Precipitation experiments using K12-Fc on (35)S-radiolabeled NK cells lysates indicated that the K12 cognate was an approximately 40-kDa protein. A human peripheral blood T cell cDNA expression library was screened with the K12-Fc protein, and two independent, positive cDNA clones were identified and sequenced. Both cDNAs encoded the same protein, which was CD7. Thus, K12 and CD7 are cognate proteins that are located next to each other on human chromosome 17q25. Additionally, we have cloned the gene encoding the mouse homologue of K12, shown that it maps near the mouse CD7 gene on chromosome 11, and established that the mouse K12 protein binds to mouse, but not human, CD7. Mouse K12-Fc inhibited in a dose-dependent manner concanavalin A-induced proliferation, but not anti-TcRalpha/beta induced proliferation, of mouse lymph node T cells. Human K12-Fc stimulated the up-regulation of CD25, CD54, and CD69 on human NK cells in vitro.


Subject(s)
Antigens, CD7/genetics , Chromosomes, Human, Pair 17 , Membrane Proteins/genetics , T-Lymphocytes/metabolism , Amino Acid Sequence , Animals , Antigens, CD7/metabolism , Chromosome Mapping , Cloning, Molecular , DNA, Complementary/analysis , DNA, Complementary/genetics , Humans , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Sequence Alignment
5.
Cytokine ; 11(9): 679-88, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10479404

ABSTRACT

The Flt-3 receptor is expressed in primitive haematopoietic cells and its ligand exerts proliferative effects on these cells in vitro in synergy with other cytokines. To increase our knowledge of the functional properties of the human Flt-3 ligand (FL) as relating to in vitro expansion of haematopoietic stem cells, the effects on murine haematopoiesis of FL alone or in combination with other growth factors were studied. Analysis of Flk-2/Flt-3 mRNA expression indicated that Flk-2/Flt-3 was preferentially expressed in primitive haematopoietic cell populations. To examine the expression of the Flk-2/Flt-3 receptor on megakaryocyte progenitors (CFU-Meg), Flk-2/Flt-3 positive and negative CD34(+)populations were separated from human bone marrow and cultured in a plasma clot culture system. CFU-Meg colonies were found in the Flk-2/Flt-3 negative fraction. Myeloid (CFU-GM) derived colonies appeared in the presence of FL alone. Neither FL+IL-3 nor FL+IL-3+IL-6 had any effect on the generation of megakaryocyte colonies (CFU-MK), due to the lack of FL receptor expression on megakaryocyte progenitors. Bone marrow cells remaining after 5-fluorouracil (5-FU) treatment of mice represent a very primitive population of progenitors enriched for reconstituting stem cells. This cell population expressed FL receptors, as revealed by RT-PCR analysis. Addition of FL alone did not enhance the replication of such cells in liquid cultures as compared to controls. However, a significantly greater generation of myeloid progenitors (CFU-GM) in clonogenic assays was observed in the presence of FL+IL-3, FL+GM-CSF or FL+CSF-1. In addition, the effects of FL on in vitro expansion of murine haematopoietic stem cells were studied using lineage-negative (lin(-)) Sca-1 positive (Sca-1(+)) c-kit positive (c-kit(+)) marrow cells from 5-FU treated mice. FL enhanced the survival of primitive murine lin(-)Sca-1(+)c-kit(+)cells. FL and IL-6 were able to significantly expand murine progenitor stem cells in vitro and promote their survival. These studies strongly suggest that FL significantly and selectively enhanced the generation of myeloid progenitors in vitro and increased myeloid progenitor responsiveness to later acting growth factors. In addition, FL synergized with IL-6 to support in vitro expansion of haematopoietic progenitors and promoted the survival of lin(-)Sca-1(+)c-kit(+)cells.


Subject(s)
Hematopoietic Stem Cells/drug effects , Membrane Proteins/pharmacology , Proto-Oncogene Proteins/drug effects , Receptor Protein-Tyrosine Kinases/drug effects , Animals , Bone Marrow Diseases/chemically induced , Bone Marrow Diseases/pathology , Cell Lineage , Cells, Cultured , Colony-Forming Units Assay , Enzyme Induction/drug effects , Fluorouracil/toxicity , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cells/cytology , Humans , Interleukin-3/pharmacology , Interleukin-6/pharmacology , Megakaryocytes/cytology , Mice , Mice, Inbred C57BL , RNA, Messenger/biosynthesis , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , fms-Like Tyrosine Kinase 3
6.
Leukemia ; 13(4): 553-7, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10214861

ABSTRACT

Myelodysplastic syndromes (MDS) caused by a clonal hematopoietic stem cell disorder progress to either overt leukemia or cytopenia, which leads to lethal infection or bleeding. Although several clinical trials have attempted to reverse cytopenia by using hematopoietic growth factors (HGF), success has been limited due in part to a limited understanding of the role of HGF in MDS progression. The FLT3 ligand, which binds to and activates the FLT3 receptor, does not have a stimulatory effect on hematopoietic cells, but can synergize with other HGF to support the expansion of both immature and committed progenitors. Using ELISA technology we measured endogenous serum levels in 93 patients with MDS: 29 RA, 1 RARS, 31 RAEB, 23 RAEBt, 9 CMML. 48.3% of RA patients' sera had significantly elevated FLT3 ligand levels ranging from 404 to 5735 pg/ml, whereas none of the RAEB, RAEBt, or CMML patients sera had levels different from controls. No significant correlation was found between FLT3 ligand levels and peripheral blood counts, bone marrow cellularity, age, cytogenetic abnormalities, or survival. Our data suggest that FLT3 ligand levels can be upregulated early in the course of MDS, which may represent an appropriate response to a decreased number of normal progenitors, or alternatively a dysregulated HGF system.


Subject(s)
Membrane Proteins/blood , Myelodysplastic Syndromes/blood , Adolescent , Adult , Aged , Anemia, Refractory/blood , Anemia, Refractory/pathology , Anemia, Refractory, with Excess of Blasts/blood , Anemia, Refractory, with Excess of Blasts/pathology , Biomarkers , Biomarkers, Tumor/blood , Disease Progression , Female , Gene Expression Regulation , Hematopoiesis , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Myelodysplastic Syndromes/complications , Myelodysplastic Syndromes/pathology , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/drug effects , Receptor Protein-Tyrosine Kinases/physiology , Stem Cell Factor/physiology , fms-Like Tyrosine Kinase 3
7.
Blood ; 93(8): 2595-604, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10194439

ABSTRACT

The flt3 ligand (FL) is a growth factor for primitive hematopoietic cells. Serum levels of FL are inversely related to the number and proliferative capacity of early hematopoietic progenitors. We sought to elucidate the molecular mechanism underlying this regulation. Expression of FL was examined in peripheral blood (PB) and bone marrow (BM) cells under normal steady-state hematopoiesis and during transient BM failure induced by chemoradiotherapy in 16 patients with hematological malignancies. Using anti-FL antibodies in Western analysis, flow cytometry, and confocal microscopy, we detected high levels of preformed FL inside but not on the surface of T lymphocytes in steady-state hematopoiesis. Intracellular FL colocalized with giantin and ERGIC-53, indicating that it is stored within and close to the Golgi apparatus. After chemotherapy-induced hematopoietic failure, FL rapidly translocated to the surface of T lymphocytes and the levels of FL released to serum increased approximately 100-fold. Expression of FL mRNA was enhanced only about sevenfold; a similar, twofold to sixfold increase in mRNA was observed in the thymus and BM of mice with irradiation-induced aplasia. Upregulation of FL mRNA was delayed when compared with the appearance of cell surface-associated and soluble protein isoforms. The described changes in FL expression in response to chemotherapy-induced aplasia were observed in all patients, irrespective of the diagnosis and treatment regimen. Our data demonstrate that mobilization of preformed FL from intracellular stores rather than de novo synthesis is responsible for increased FL levels in BM failure.


Subject(s)
Antineoplastic Agents/adverse effects , Bone Marrow/pathology , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/physiopathology , Hematopoiesis/physiology , Membrane Proteins/genetics , T-Lymphocytes/physiology , Adult , Aged , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Bone Marrow Cells/physiology , Female , Gene Expression Regulation, Neoplastic/drug effects , Hematologic Neoplasms/pathology , Hematopoiesis/drug effects , Humans , Male , Membrane Proteins/biosynthesis , Membrane Proteins/blood , Mice , Microscopy, Confocal , Middle Aged , RNA, Messenger/genetics , Time Factors , Transcription, Genetic
8.
Exp Hematol ; 27(1): 155-61, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9923454

ABSTRACT

The potential of recombinant human (rh)Flt3 ligand (FL), alone or in combination with other recombinant growth factors, to mobilize peripheral blood precursor cells (PBPCs) was examined in an animal model. Adult outbred New Zealand White rabbits received subcutaneous injections daily for 14 days in a standardized protocol; whole blood cell counts and colony-forming unit-granulocyte/macrophage (CFU-GM) colonies were measured 3 times weekly during the injection period and for an additional observation period of 14 days. Two animals in each group were treated as follows: 200 or 500 microg/kg FL, 10 microg/kg granulocyte colony-stimulating factor (G-CSF), 10 or 75 microg/kg stem cell factor (SCF), 10 microg/kg G-CSF + 500 microg/kg FL, 10 microg/kg G-CSF + 75 microg/kg SCF + 500 microg/kg FL. Both G-CSF and FL induced a sustained and dose-dependent increase in the leukocyte count to a maximum of 5-fold. They were additive in combination, leading to a tenfold increase in white blood cell counts. No consistent pattern was observed for platelet counts or red blood cells. No toxic side effects were seen. Both G-CSF and FL mobilized CFU-GM in a dose-dependent fashion to a 59-fold increase for G-CSF and 116-fold for FL. Maximum mobilization occurred on day 4 with G-CSF and on day 11 with FL. G-CSF + FL in combination acted synergistically, inducing a 503-fold increase of CFU-GM over baseline. The addition of SCF to this combination did not alter leukocyte counts or CFU-GM mobilization. Our results indicate that FL is a potent and safe agent for the mobilization of PB-PCs and is synergistic with G-CSF.


Subject(s)
Granulocyte Colony-Stimulating Factor/therapeutic use , Hematopoietic Stem Cell Mobilization , Membrane Proteins/therapeutic use , Animals , Behavior, Animal/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Female , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocytes/cytology , Hematopoiesis , Hematopoietic Stem Cells/drug effects , Humans , Leukocyte Count/drug effects , Leukocytes/cytology , Leukocytes/drug effects , Ligands , Macrophages/cytology , Male , Membrane Proteins/pharmacology , Rabbits , Recombinant Proteins , Stem Cell Factor/pharmacology , Stem Cells , Time Factors
9.
Blood ; 92(6): 2003-11, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9731058

ABSTRACT

flt3/flk-2 ligand (FL) is a cytokine that exhibits synergistic activities in combination with other early acting factors on subpopulations of hematopoietic stem/progenitor cells. In addition to normal hematopoietic precursors, expression of the FL receptor, flt3R, has been frequently demonstrated on the blast cells from patients with acute B-lineage lymphoblastic, myeloid, and biphenotypic (also known as hybrid or mixed) leukemias. Because many of these leukemic cell types express FL, the possibility has been raised that altered regulation of FL-mediated signaling might contribute to malignant transformation or expansion of the leukemic clone. In humans, FL is predominantly synthesized as a transmembrane protein that must undergo proteolytic cleavage to generate a soluble form. To investigate the consequences of constitutively expressing the analogous murine FL isoform in murine hematopoietic stem/progenitor cells, lethally irradiated syngeneic mice (18 total) were engrafted with post-5-fluorouracil-treated bone marrow cells transduced ex vivo with a recombinant retroviral vector (MSCV-FL) encoding murine transmembrane FL. Compared with control mice (8 total), MSCV-FL mice presented with a mild macrocytic anemia but were otherwise healthy for more than 5 months posttransplant (until 22 weeks). Subsequently, all primary MSCV-FL recipients observed for up to 1 year plus 83% (20 of 24) of secondary MSCV-FL animals that had received bone marrow from asymptomatic primary hosts reconstituted for 4 to 5 months developed transplantable hematologic malignancies (with mean latency periods of 30 and 23 weeks, respectively). Phenotypic and molecular analyses indicated that the tumor cells expressed flt3R and displayed B-cell and/or myeloid markers. These data, establishing that dysregulated expression of FL in primitive hematopoietic cells predisposes flt3R+ precursors to leukemic transformation, underscore a potential role of this cytokine/receptor combination in certain human leukemias.


Subject(s)
Hematopoiesis/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Leukemia, Experimental/genetics , Membrane Proteins/biosynthesis , Animals , Bone Marrow/metabolism , Clone Cells , Disease Susceptibility , Gene Expression Regulation, Neoplastic/genetics , Genetic Vectors/genetics , Leukemia, Experimental/etiology , Leukemia, Experimental/pathology , Ligands , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Recombinant Proteins/biosynthesis , Tumor Cells, Cultured
10.
Blood ; 92(3): 765-9, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9680342

ABSTRACT

Several hematopoietic cytokines have been investigated for their potential to provide protection from the lethal consequences of bone marrow aplasia after total body irradiation (TBI). Some can increase the dose of irradiation tolerated by the animals; none allow endogenous recovery after doses such as administered in clinical blood or marrow transplantation. We tested the radioprotective potential of FLT-3 ligand, an early acting hematopoietic cytokine, alone and in combination with a late acting cytokine, granulocyte-colony stimulating factor (G-CSF). Adult outbred New Zealand White rabbits were submitted to TBI of 1,200 or 1,400 cGy by a Co60 source. Recombinant human (rh) FLT-3 ligand at a dose of 500 microg/kg and/or rhG-CSF at a dose of 10 microg/kg were administered for 14 days subcutaneously daily, beginning either 2 days before or the day after TBI. All control animals given no growth factors died of aplasia at day 10 (range, 5 to 16). All 8 animals given G-CSF had severe aplasia and 7 died at day 8 (range, 5 to 10); 1 animal survived, with G-CSF being administered before TBI. In contrast, 11 of 12 animals given FLT-3 ligand, with or without G-CSF, survived. Radioprotection was best in the group given FLT-3 ligand together with G-CSF before TBI. In these animals median platelet counts were never <10 x 10(9)/L and median white blood cell counts never <0.5 x 10(9)/L. These data show that hematopoietic recovery can occur after 1,400 cGy TBI in rabbits, if protected by FLT-3 ligand, and suggest a radioprotective clinical potential of this cytokine.


Subject(s)
Anemia, Aplastic/prevention & control , Bone Marrow/drug effects , Hematopoietic Stem Cells/drug effects , Membrane Proteins/therapeutic use , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents/therapeutic use , Whole-Body Irradiation/adverse effects , Anemia, Aplastic/etiology , Animals , Apoptosis/drug effects , Bone Marrow/radiation effects , Drug Synergism , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte Colony-Stimulating Factor/therapeutic use , Hematopoietic Stem Cells/radiation effects , Membrane Proteins/pharmacology , Rabbits , Radiation Injuries, Experimental/etiology , Radiation-Protective Agents/pharmacology , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use
11.
Eur J Haematol ; 60(5): 297-306, 1998 May.
Article in English | MEDLINE | ID: mdl-9654159

ABSTRACT

We compared the effect of human flt3 ligand (FL) and stem cell factor (SCF) on cord blood (CB)-derived CD34+ cells expressing different levels of flt3 or c-kit tyrosine kinase (TK) receptor in clonal cell culture. The c-kit receptor was expressed by 58.5+/-16.7% of CB CD34+ cells (n=19), in which c-kit(high), c-kit(low) and c-kit cell populations could be identified. In contrast, the flt3 receptor (FR) was weakly expressed on 58.6+/-8.3% (n=9) of CB CD34+ cells. FL+erythropoietin (Epo) failed to support erythroid burst (BFU-E) formation by any subpopulation of CD34+ cells. However, SCF + Epo supported BFU-E and erythrocyte-containing mixed (CFU-mix) colony formation from all subpopulations. Interestingly, FL markedly augmented CFU-mix colony formation supported by interleukin (IL)-3 + Epo when CD34+c-kit(low) or CD34+FR+ cells were used as the target. On the other hand, SCF significantly enhanced CFU-mix colony formation supported by IL-3 + Epo when CD34+c-kit(high) or low and CD34+FR+ cells were used. The replating potential of CFU-mix supported by IL-3 + Epo+ FL was greater when CD34+c-kit(low) or CD34+FR+ cells were used. When the CD34+c-kit(low) cells were used, the number of lineages expressed in secondary cultures of CFU-mix colonies derived from primary cultures containing IL-3 + Epo + FL or SCF was significantly larger than when the primary cultures contained IL-3 + Epo. Furthermore, the number of long-term culture-initiating cells found in CD34+FR+ cells was larger than that in FR cells. CB-derived CD34+c-kit(low) cells represent a less mature population than c-kit(high) cells, as reported previously. Therefore, these results indicate that both FL and SCF can act on primitive multipotential progenitors. However, it is still uncertain whether CB-derived CD34+FR+ cells are less mature than CD34+FR- cells.


Subject(s)
Antigens, CD34/analysis , Fetal Blood/cytology , Hematopoiesis/drug effects , Membrane Proteins/pharmacology , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/pharmacology , Cells, Cultured , Colony-Forming Units Assay , Culture Media, Serum-Free , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Erythropoietin/pharmacology , Flow Cytometry , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Humans , Interleukin-3/pharmacology , Membrane Proteins/metabolism
12.
Curr Opin Hematol ; 5(3): 192-6, 1998 May.
Article in English | MEDLINE | ID: mdl-9664159

ABSTRACT

Flt3 ligand (FL) is a member of a small family of growth factors that stimulate the proliferation of hematopoietic cells by binding to and activating distinct tyrosine kinase receptors. Other members of this family include M-CSF and the c-kit ligand. Expression of the Flt3 receptor is primarily restricted to the most primitive hematopoietic progenitor cells, and FL stimulates the proliferation in vitro and the expansion and mobilization in vivo of stem and progenitor cells. FL as a single factor has little proliferative activity on these stem and progenitor cells, but it synergizes with a wide range of other colony-stimulating factors and interleukins to stimulate proliferation of these cells. FL is also an effective agent for mobilizing stem and progenitor cells to peripheral blood. Recent data demonstrating anti-tumor activity of FL suggest that this protein plays a major role in activating the immune system via its ability to stimulate the production of both dendritic and natural killer cells. These biologic activities of FL may potentially prove quite useful in a number of clinical settings.


Subject(s)
Hematopoiesis/drug effects , Membrane Proteins/therapeutic use , Clinical Trials as Topic , Dendritic Cells/drug effects , Genetic Therapy , Hematopoietic Stem Cell Mobilization , Humans , Immunotherapy
13.
J Biol Chem ; 273(28): 17626-33, 1998 Jul 10.
Article in English | MEDLINE | ID: mdl-9651358

ABSTRACT

FLT3 ligand (FLT3L) stimulates primitive hematopoietic cells by binding to and activating the FLT3 receptor (FLT3R). We carried out a structure-activity study of human FLT3L in order to define the residues involved in receptor binding. We developed a rapid method to screen randomly mutagenized FLT3L using a FLT3R-Fc fusion protein to probe the relative binding activities of mutated ligand. Approximately 60,000 potential mutants were screened, and the DNA from 59 clones was sequenced. Thirty-one single amino acid substitutions at 24 positions of FLT3L either enhanced or reduced activity in receptor binding and cell proliferation assays. Eleven representative proteins were purified and analyzed for receptor affinity, specific activity, and physical properties. Receptor affinity and bioactivity were highly correlated. FLT3L affinity for receptor improved when four individual mutations that enhance FLT3L receptor affinity were combined in a single molecule. A model of FLT3L three-dimensional structure was generated based on sequence alignment and x-ray structure of macrophage colony-stimulating factor. Most residues implicated in receptor binding are widely dispersed in the primary structure of FLT3L, yet they localize to a surface patch in the tertiary model. A mutation that maps to and is predicted to disrupt the proposed dimerization interface between FLT3L monomers exhibits a Stokes radius that is concentration-dependent, suggesting that this mutation disrupts the FLT3L dimer.


Subject(s)
Membrane Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Receptor Protein-Tyrosine Kinases/chemistry , Sequence Homology, Amino Acid , Structure-Activity Relationship
14.
Exp Hematol ; 26(5): 365-73, 1998 May.
Article in English | MEDLINE | ID: mdl-9590652

ABSTRACT

The hematopoietic growth factors stem cell factor (SCF) and flt3 ligand (flt3L) are produced within the hematopoietic microenvironment in a membrane-bound and soluble isoform. To elucidate the relevance of the two isoforms in the network of early-acting cytokines, we examined the interaction of membrane-bound SCF with the soluble forms of SCF and flt3L in long-term cultures of human bone marrow cells. Feeder layers of the murine SCF-deficient Steel stromal cell line transfected with human cDNA stably expressing SCF as a transmembrane molecule were used to support growth of mononuclear cells and CD34+ progenitors derived from normal human bone marrow or from hypoplastic marrow of patients with aplastic anemia (AA). The output of nonadherent progenitor cells representing colony-forming units (CFU) and high-proliferative potential colony-forming cells (HPP-CFC) was scored weekly in secondary methylcellulose cultures; the number of colonies derived from long-term culture-initiating cells (LTC-IC) was determined in nonadherent and adherent cells at 5 weeks. Membrane-bound SCF expressed in the stromal layer was more effective than soluble SCF and soluble flt3L in maintaining clonogenic progenitors. Furthermore, the transmembrane form of SCF effectively synergized with both exogenously supplied factors, although the effect of flt3L was superior to the effect of soluble SCF. In cultures of normal bone marrow cells, addition of flt3L enhanced the total number of CFU and HPP-CFC-type progenitors, primarily of the granulocyte/macrophage lineage, by six- to ninefold after 3 weeks and of LTC-IC-derived colonies by 13-fold after 5 weeks of culture. In cultures of AA cells, both the number and the survival rate of clonogenic precursors were severely impaired even in the presence of flt3L, which, however, yielded a two- to sixfold enhancement of CFU and HPP-CFC numbers at 1 to 2 weeks. In comparison with the hematopoietic function of human Dexter-type stroma cultures, murine feeders expressing high levels of membrane-associated human SCF were equivalent in supporting hematopoiesis during the initial 3 to 4 culture weeks when supplemented with flt3L. These results demonstrate that soluble flt3L interacts with membrane-bound SCF in supporting the long-term growth of bone marrow progenitor cells. The hypothesis that SCF and flt3L function synergistically during the very early stages of human hematopoiesis is thereby reinforced.


Subject(s)
Anemia, Aplastic/pathology , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/physiology , Membrane Proteins/pharmacology , Membrane Proteins/physiology , Stem Cell Factor/pharmacology , Stem Cell Factor/physiology , Adolescent , Adult , Age Factors , Animals , Cell Division/drug effects , Cell Line , Cell Survival/drug effects , Cells, Cultured , Child , Drug Synergism , Female , Hematopoiesis , Hematopoietic Stem Cells/cytology , Humans , Male , Mice , Middle Aged , Stromal Cells/drug effects , Time Factors
15.
J Virol ; 72(5): 4015-21, 1998 May.
Article in English | MEDLINE | ID: mdl-9557689

ABSTRACT

Epstein-Barr virus (EBV) is a ubiquitous herpesvirus associated with infectious mononucleosis and several tumors. The BARF1 gene is transcribed early after EBV infection from the BamHI A fragment of the EBV genome. Evidence shown here indicates that the BARF1 protein is secreted into the medium of transfected cells and from EBV-carrying B cells induced to allow lytic replication of the virus. Expression cloning identified colony-stimulating factor-1 (CSF-1) as a ligand for BARF1. Computer-assisted analyses indicated that subtle amino acid sequence homology exists between BARF1 and c-fins, the cellular proto-oncogene that is the receptor for CSF-1. Recombinant BARF1 protein was found to be biologically active, and it neutralized the proliferative effects of human CSF-1 in a dose-dependent fashion when assayed in vitro. Since CSF-1 is a pleiotropic cytokine best known for its differentiating effects on macrophages, these data suggest that BARF1 may function to modulate the host immune response to EBV infection.


Subject(s)
Herpesvirus 4, Human/genetics , Macrophage Colony-Stimulating Factor/metabolism , Receptor, Macrophage Colony-Stimulating Factor/genetics , Viral Proteins/genetics , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Genes, Viral , Herpesvirus 4, Human/metabolism , Humans , Ligands , Macrophage Colony-Stimulating Factor/genetics , Proto-Oncogene Mas , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Sequence Homology, Amino Acid , Solubility , Viral Proteins/metabolism
17.
Blood ; 90(11): 4321-31, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9373243

ABSTRACT

B-cell commitment and early development from multipotent hematopoietic progenitor cells has until recently been considered to be dependent on direct interaction with stromal cells. We recently showed that the flt3 ligand (FL) has a unique ability to interact with interleukin-7 (IL-7) to directly and selectively promote B-cell development from murine bone marrow progenitor cells with a combined myeloid and lymphoid potential. Here we report that whereas IL-10 alone has no ability to stimulate growth of primitive (Lin-Sca-1(+)c-kit+) bone marrow progenitor cells, it potently enhances FL + IL-7-induced proliferation (sevenfold). This enhanced proliferation results from recruitment of progenitors unresponsive to FL + IL-7 alone, as well as from increased growth of individual clones, resulting in a 7,000-fold cellular expansion over 12 days. Single cell cultures and delayed addition studies suggested that the stimulatory effect of IL-10 was directly mediated on the progenitor cells. The cells generated in response to FL + IL-7 + IL-10 appeared to be almost exclusively proB cells, as shown by their expression of B220, CD24, CD43, and lack of expression of c mu, myeloid, erythroid, and T-cell surface antigens. Although IL-10 also enhanced kit ligand (KL) + IL-7-induced proliferation of Lin-Sca-1(+)c-kit+ progenitor cells, the resulting cells were predominantly myeloid progeny. Accordingly, FL + IL-7 + IL-10 was 100-fold more efficient in stimulating production of proB cells than KL + IL-7 + IL-10. In contrast to its ability to stimulate the earliest phase of proB cell formation and proliferation, IL-10 inhibited growth of proB cells generated in response to FL + IL-7. Analysis of CD19 expression on cells generated in FL + IL-7 + IL-10 showed that almost all cells generated under these conditions lacked expression of CD19, in contrast to cells generated in the absence of IL-10, which were predominantly CD19(+). Replating of sorted CD19(+) and CD19(-) proB cells in FL + IL-7 or FL + IL-7 + IL-10 showed that IL-10 efficiently blocked growth of CD19(+), but not CD19(-) cells. Both CD19(-) and CD19(+) cells expressed lambda5 and VpreB , shown to be specific for B-cell progenitors. In addition, sorted CD19(-) cells generated CD19(+) cells in response to FL + IL-7. Thus, IL-10 has a dual regulatory effect on early B-cell development from primitive murine bone marrow progenitor cells in that it enhances FL + IL-7-induced proB-cell formation and growth before acquisition of CD19 expression, whereas growth of CD19(+) proB cells is inhibited.


Subject(s)
Antigens, CD19/metabolism , B-Lymphocytes/drug effects , Hematopoietic Stem Cells/cytology , Interleukin-10/pharmacology , Interleukin-7/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/metabolism , Stem Cell Factor/metabolism , Animals , B-Lymphocytes/cytology , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured , Hematopoietic Stem Cells/immunology , Humans , Mice , Recombinant Proteins/metabolism , fms-Like Tyrosine Kinase 3
18.
Blood ; 90(11): 4363-8, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9373247

ABSTRACT

We recently showed that c-kit signal synergizes with glycoprotein (gp)130 signal mediated by a complex of interleukin (IL)-6 and soluble IL-6 receptor (IL-6/sIL-6R) to stimulate the expansion of human primitive hematopoietic progenitor cells and erythropoietin-independent erythropoiesis. In the present study, we examined the effect of a ligand for Flt3 (FL), whose receptor tyrosine kinase is closely related to c-kit, in combination with IL-6/sIL-6R on human hematopoiesis in vitro. In serum-containing methylcellulose clonal culture of cord blood CD34(+) cells, whereas FL alone stimulated only granulocyte-macrophage (GM) colony formation, erythroid bursts and mixed colonies in addition to GM colonies were induced by FL with IL-6/sIL-6R, but not IL-6/sIL-6R alone. In suspension culture, CD34(+) cells generated a small number of myeloid cells in the presence of FL or IL-6/sIL-6R alone. However, the addition of IL-6/sIL-6R to the culture with FL induced the generation of a significant number of erythroid cells and megakaryocytes in addition to myeloid cells. The combination of FL and IL-6/sIL-6R also induced a remarkable expansion of GM colony- and erythroid burst-forming cells and multipotential progenitors, although FL or IL-6/sIL-6R alone induced the generation of only a small number of progenitors for GM colonies. The synergistic effects of FL and IL-6/sIL-6R were confirmed in serum-free clonal and suspension cultures. In addition, the addition of anti-human gp130 monoclonal antibodies abrogated the synergistic action. These results indicate that Flt3 signal, as well as c-kit signal, synergizes with gp130 signal to stimulate human myelopoiesis, erythropoiesis and megakaryopoiesis, and the expansion of primitive multipotential hematopoietic progenitor cells.


Subject(s)
Antigens, CD/pharmacology , Hematopoiesis/drug effects , Membrane Glycoproteins/pharmacology , Proto-Oncogene Proteins/pharmacology , Receptor Protein-Tyrosine Kinases/pharmacology , Receptors, Cell Surface/metabolism , Signal Transduction/drug effects , Stem Cell Factor/pharmacology , Antibodies, Monoclonal , Antigens, CD34/metabolism , Cells, Cultured , Cytokine Receptor gp130 , Drug Synergism , Fetal Blood/cytology , Humans , Interleukin-6/metabolism , Receptors, Interleukin-6/metabolism , fms-Like Tyrosine Kinase 3
19.
Blood ; 90(9): 3781-8, 1997 Nov 01.
Article in English | MEDLINE | ID: mdl-9345066

ABSTRACT

Peripheral blood progenitor cells (PBPC) are increasingly being used in the clinic as a replacement for bone marrow (BM) in the transplantation setting. We investigated the capacity of several different growth factors, including human flt3 ligand (FL), alone and in combination with granulocyte-macrophage colony-stimulating factor (GM-CSF ) or granulocyte colony-stimulating factor (G-CSF ), to mobilize colony forming cells (CFU) into the peripheral blood (PB) of mice. Mice were injected subcutaneously (SC) with growth factors daily for up to 10 days. Comparing the single agents, we found that FL alone was superior to GM-CSF or G-CSF in mobilizing CFU into the PB. FL synergized with both GM-CSF or G-CSF to mobilize more CFU, and in a shorter period of time, than did any single agent. Administration of FL plus G-CSF for 6 days resulted in a 1,423-fold and 2,717-fold increase of colony-forming unit-granulocyte-macrophage (CFU-GM) and colony-forming unit granulocyte, erythroid, monocyte, megakaryocyte (CFU-GEMM) in PB, respectively, when compared with control mice. We also followed the kinetics of CFU numerical changes in the BM of mice treated with growth factors. While GM-CSF and G-CSF alone had little effect on BM CFU over time, FL alone increased CFU-GM and CFU-GEMM threefold and fivefold, respectively. Addition of GM-CSF or G-CSF to FL did not increase CFU in BM over levels seen with FL alone. However, after the initial increase in BM CFU after FL plus G-CSF treatment for 3 days, BM CFU returned to control levels after 5 days treatment, and CFU-GM were significantly reduced (65%) after 7 days treatment, when compared with control mice. Finally, we found that transplantation of FL or FL plus G-CSF-mobilized PB cells protected lethally irradiated mice and resulted in long-term multilineage hematopoietic reconstitution.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Membrane Proteins/pharmacology , Animals , Blood Cell Count , Drug Synergism , Female , Hematopoietic Stem Cells/drug effects , Humans , Mice , Mice, Inbred C57BL
20.
Blood ; 90(10): 3903-13, 1997 Nov 15.
Article in English | MEDLINE | ID: mdl-9354657

ABSTRACT

Flt3 ligand (FL) has been proposed as a possible modulator of early hematopoietic cell growth. The purpose of this study was to analyze the impact of FL on ex vivo expansion of hematopoietic cells obtained from adult donors. We sought to precisely identify hematopoietic populations responsive to FL and to quantitate the ability of FL to enhance the survival and/or proliferation of early hematopoietic precursors in a stroma-free culture system. Towards that end, four CD34+ subsets were isolated and their response to FL was characterized. In methylcellulose, FL significantly increased colony formation by CD34+ CD38dim cells but not CD34+ CD38+ cells. In suspension culture, the enhancement of cell expansion by FL was 10 times greater with the CD34+ CD38dim fraction than the CD34+ CD38+ fraction. FL stimulated the generation of colony-forming unit-granulocyte-macrophage (CFU-GM) from the CD34+CD38dim fraction by 14.5- +/- 5.6-fold. To determine if CD34+ CD38dim cells responded uniformly to FL, the population was subdivided into a CD34+ CD38dim CD33dim HLA-DR+ (HLA-DR+) fraction and a CD34+ CD38dim CD33(dim) HLA-DRdim (HLA-DRdim) fraction. FL was far more effective at stimulating cell and progenitor growth from the HLA-DR+ fraction. To determine if FL enhanced or depleted the number of precommitted cells in expansion culture, CD34+ CD38dim and HLA-DR+ fractions were incubated in liquid culture and analyzed by flow cytometry. Inclusion of FL enhanced the absolute number of primitive CD34+ CD33dim cells and CD34+ HLA-DRdim cells after 5 to 12 days of cultivation. To confirm immunophenotypic data, the effect of FL on long-term culture-initiating cells (LTCIC) was determined. After 2 weeks of incubation of CD34+ CD38dim or HLA-DR+ cultures, LTCIC recoveries were significantly higher with FL in 5 of 6 trials (P < . 05). For HLA-DR+ cells, LTCIC recoveries averaged 214% +/- 87% of input with FL and 24% +/- 16% without FL. In contrast, HLA-DRdim LTCIC could not be maintained in stroma-free culture. We conclude that less than 10% of CD34+ cells respond vigorously to FL and that those cells are contained within the HLA-DR+ fraction. FL stimulates the expansion of total cells, CD34+ cells, and CFU-GM and enhances the pool of early CD34+ CD33(dim) cells, CD34+ HLA-DRdim cells, and LTCIC. These data indicate that it is possible to expand hematopoietic progenitors from adult donors without losing precursors from the precommitted cell pool.


Subject(s)
Antigens, CD , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Membrane Proteins/pharmacology , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Adult , Antigens, CD34 , Antigens, Differentiation , Cells, Cultured , HLA-DR Antigens , Hematopoietic Stem Cells/immunology , Humans , Immunophenotyping , Membrane Glycoproteins , NAD+ Nucleosidase
SELECTION OF CITATIONS
SEARCH DETAIL
...