Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Leukemia ; 31(4): 872-881, 2017 04.
Article in English | MEDLINE | ID: mdl-27740633

ABSTRACT

Traditional response criteria in myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) are based on bone marrow morphology and may not accurately reflect clonal tumor burden in patients treated with non-cytotoxic chemotherapy. We used next-generation sequencing of serial bone marrow samples to monitor MDS and AML tumor burden during treatment with epigenetic therapy (decitabine and panobinostat). Serial bone marrow samples (and skin as a source of normal DNA) from 25 MDS and AML patients were sequenced (exome or 285 gene panel). We observed that responders, including those in complete remission (CR), can have persistent measurable tumor burden (that is, mutations) for at least 1 year without disease progression. Using an ultrasensitive sequencing approach, we detected extremely rare mutations (equivalent to 1 heterozygous mutant cell in 2000 non-mutant cells) months to years before their expansion at disease relapse. While patients can live with persistent clonal hematopoiesis in a CR or stable disease, ultimately we find evidence that expansion of a rare subclone occurs at relapse or progression. Here we demonstrate that sequencing of serial samples provides an alternative measure of tumor burden in MDS or AML patients and augments traditional response criteria that rely on bone marrow blast percentage.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Clonal Evolution/genetics , Epigenesis, Genetic/drug effects , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Aged , Aged, 80 and over , Bone Marrow/pathology , Exome , Female , Genes, p53 , High-Throughput Nucleotide Sequencing , Histone Deacetylase Inhibitors/administration & dosage , Humans , Leukemia, Myeloid, Acute/diagnosis , Male , Middle Aged , Mutation , Myelodysplastic Syndromes/diagnosis , Polymorphism, Single Nucleotide , Remission Induction , Treatment Outcome , Tumor Burden
4.
Leukemia ; 28(9): 1851-60, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24518205

ABSTRACT

Recent studies demonstrate that inflammatory signals regulate hematopoietic stem cells (HSCs). Granulocyte colony-stimulating factor (G-CSF) is often induced with infection and has a key role in the stress granulopoiesis response. However, its effects on HSCs are less clear. Herein, we show that treatment with G-CSF induces expansion and increased quiescence of phenotypic HSCs, but causes a marked, cell-autonomous HSC repopulating defect associated with induction of Toll-like receptor (TLR) expression and signaling. The G-CSF-mediated expansion of HSCs is reduced in mice lacking TLR2, TLR4 or the TLR signaling adaptor MyD88. Induction of HSC quiescence is abrogated in mice lacking MyD88 or in mice treated with antibiotics to suppress intestinal flora. Finally, loss of TLR4 or germ-free conditions mitigates the G-CSF-mediated HSC repopulating defect. These data suggest that low-level TLR agonist production by commensal flora contributes to the regulation of HSC function and that G-CSF negatively regulates HSCs, in part, by enhancing TLR signaling.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Signal Transduction/physiology , Toll-Like Receptors/physiology , Animals , Hematopoietic Stem Cells/physiology , Intestines/microbiology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/physiology , Receptors, Granulocyte Colony-Stimulating Factor/physiology , Signal Transduction/drug effects , fms-Like Tyrosine Kinase 3/physiology
5.
Leukemia ; 27(6): 1275-82, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23443460

ABSTRACT

Recent studies suggest that most cases of myelodysplastic syndrome (MDS) are clonally heterogeneous, with a founding clone and multiple subclones. It is not known whether specific gene mutations typically occur in founding clones or subclones. We screened a panel of 94 candidate genes in a cohort of 157 patients with MDS or secondary acute myeloid leukemia (sAML). This included 150 cases with samples obtained at MDS diagnosis and 15 cases with samples obtained at sAML transformation (8 were also analyzed at the MDS stage). We performed whole-genome sequencing (WGS) to define the clonal architecture in eight sAML genomes and identified the range of variant allele frequencies (VAFs) for founding clone mutations. At least one mutation or cytogenetic abnormality was detected in 83% of the 150 MDS patients and 17 genes were significantly mutated (false discovery rate ≤0.05). Individual genes and patient samples displayed a wide range of VAFs for recurrently mutated genes, indicating that no single gene is exclusively mutated in the founding clone. The VAFs of recurrently mutated genes did not fully recapitulate the clonal architecture defined by WGS, suggesting that comprehensive sequencing may be required to accurately assess the clonal status of recurrently mutated genes in MDS.


Subject(s)
Mutation , Myelodysplastic Syndromes/genetics , Female , Gene Frequency , Humans , Leukemia, Myeloid, Acute/genetics , Male , Middle Aged , Recurrence
6.
Leukemia ; 25(2): 211-7, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21079612

ABSTRACT

Under normal conditions, the great majority of hematopoietic stem/progenitors cells (HSPCs) reside in the bone marrow. The number of HSPCs in the circulation can be markedly increased in response to a number of stimuli, including hematopoietic growth factors, myeloablative agents and environmental stresses such as infection. The ability to 'mobilize' HSPCs from the bone marrow to the blood has been exploited clinically to obtain HSPCs for stem cell transplantation and, more recently, to stimulate therapeutic angiogenesis at sites of tissue ischemia. Moreover, there is recent interest in the use of mobilizing agents to sensitize leukemia and other hematopoietic malignancies to cytotoxic agents. Key to optimizing clinical mobilizing regimens is an understanding of the fundamental mechanisms of HSPC mobilization. In this review, we discuss recent advances in our understanding of the mechanisms by which granulocyte colony-stimulating factor (G-CSF), the prototypical mobilizing agent, induces HSPC mobilization.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization/methods , Hematologic Neoplasms/therapy , Humans
7.
Immunity ; 14(2): 193-204, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11239451

ABSTRACT

To investigate the role of signal transducer and activator of transcription (STAT) proteins in granulocyte colony-stimulating factor (G-CSF)-regulated biological responses, we generated transgenic mice with a targeted mutation of their G-CSF receptor (termed d715F) that abolishes G-CSF-dependent STAT-3 activation and attenuates STAT-5 activation. Homozygous mutant mice are severely neutropenic with an accumulation of immature myeloid precursors in their bone marrow. G-CSF-induced proliferation and granulocytic differentiation of hematopoietic progenitors is severely impaired. Expression of a constitutively active form of STAT-3 in d715F progenitors nearly completely rescued these defects. Conversely, expression of a dominant-negative form of STAT-3 in wild-type progenitors results in impaired G-CSF-induced proliferation and differentiation. These data suggest that STAT-3 activation by the G-CSFR is critical for the transduction of normal proliferative signals and contributes to differentiative signals.


Subject(s)
DNA-Binding Proteins/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocytes/drug effects , Granulocytes/metabolism , Milk Proteins , Trans-Activators/metabolism , Animals , Base Sequence , Cell Differentiation/drug effects , Cell Division/drug effects , DNA Primers/genetics , Granulocyte Colony-Stimulating Factor/metabolism , Granulocytes/cytology , Hematopoiesis/genetics , In Vitro Techniques , Mice , Mice, Knockout , Mice, Transgenic , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , STAT3 Transcription Factor , STAT5 Transcription Factor , Signal Transduction
8.
Blood ; 95(10): 3025-31, 2000 May 15.
Article in English | MEDLINE | ID: mdl-10807765

ABSTRACT

The mechanisms that regulate hematopoietic progenitor cell (HPC) mobilization from the bone marrow to blood have not yet been defined. HPC mobilization by granulocyte colony-stimulating factor (G-CSF), cyclophosphamide (CY), or interleukin-8 but not flt-3 ligand is markedly impaired in G-CSF receptor-deficient (G-CSFR-deficient) mice. G-CSFR is expressed on mature hematopoietic cells, HPCs, and stromal cells, which suggests that G-CSFR signals in one or more of these cell types was required for mobilization by these agents. To define the cell type(s) responsible for G-CSF-dependent mobilization, a series of chimeric mice were generated using bone marrow transplantation. Mobilization studies in these chimeras demonstrated that expression of the G-CSFR on transplantable hematopoietic cells but not stromal cells is required for CY- or G-CSF-induced mobilization. Moreover, in irradiated mice reconstituted with both wild type and G-CSFR-deficient bone marrow cells, treatment with CY or G-CSF resulted in the equal mobilization of both types of HPCs. This result held true for a broad spectrum of HPCs including colony-forming cells, CD34(+) lineage(-) and Sca(+) lineage(-) cells, and long-term culture initiating cells. Collectively, these data provide the first definitive evidence that expression of the G-CSFR on HPCs is not required for their mobilization by G-CSF and suggest a model in which G-CSFR-dependent signals act in trans to mobilize HPCs from the bone marrow.


Subject(s)
Bone Marrow Transplantation , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/physiology , Receptors, Granulocyte Colony-Stimulating Factor/physiology , Animals , Granulocyte Colony-Stimulating Factor/physiology , Hematopoietic Stem Cells/pathology , Mice , Transplantation Chimera
9.
Semin Hematol ; 37(1 Suppl 2): 25-32, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10718156

ABSTRACT

Hematopoietic progenitor cells (HPC) can be mobilized from the bone marrow into the peripheral circulation in response to diverse stimuli, including hematopoietic growth factors, cytotoxic agents, and certain chemokines. Despite significant differences in their biologic activities, these stimuli result in the mobilization of HPC with a similar phenotype, suggesting that a common mechanism for mobilization may exist. To explore the mechanisms of granulocyte colony-stimulating factor (G-CSF)-induced mobilization, we examined HPC mobilization in mice that are genetically deficient for the G-CSF receptor (G-CSFR). Their response was determined to each of three major types of mobilizing stimuli: cytotoxic agents (cyclophosphamide), chemokines (Interleukin-8[IL-8]), and hematopoietic growth factors (G-CSF, fit-3 ligand, and IL-12). These studies demonstrate that the G-CSFR is required for mobilization in response to cyclophosphamide and IL-8, but not fit-3 ligand or IL-12, and suggest that the G-CSFR may play an important and previously unexpected role in HPC migration.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Animals , Bone Marrow Cells , Cytokines/pharmacology , Humans , Membrane Proteins/pharmacology , Receptors, Granulocyte Colony-Stimulating Factor/physiology
10.
Immunity ; 11(2): 153-61, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10485650

ABSTRACT

To investigate the specificity of cytokine signals in hematopoietic differentiation, we generated mice with a targeted mutation of their G-CSF receptor (G-CSFR) such that the cytoplasmic (signaling) domain of the G-CSFR is replaced with the cytoplasmic domain of the erythropoietin receptor. In homozygous mutant mice, expression of this chimeric receptor had no apparent affect on lineage commitment and was able to support the production of morphologically mature neutrophils. However, mutant neutrophils displayed reduced chemotaxis, and G-CSF-stimulated mobilization of neutrophils and hematopoietic progenitors from the bone marrow to blood was markedly impaired. Thus, the G-CSFR is generating unique signals that are required for certain specialized hematopoietic cell functions but are not required for granulocytic differentiation or lineage commitment.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells/physiology , Receptors, Granulocyte Colony-Stimulating Factor/physiology , Animals , Chemotaxis, Leukocyte/drug effects , Female , Flow Cytometry , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoiesis/drug effects , Hematopoietic Stem Cell Mobilization , Interleukin-8/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/drug effects , Neutrophils/immunology , Receptors, Erythropoietin/physiology
11.
J Clin Invest ; 103(6): 825-32, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10079103

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) is a hematopoietic growth factor that is widely used to treat neutropenia. In addition to stimulating polymorphonuclear neutrophil (PMN) production, G-CSF may have significant effects on PMN function. Because G-CSF receptor (G-CSFR)-deficient mice do not have the expected neutrophilia after administration of human interleukin-8 (IL-8), we examined the effect of the loss of G-CSFR on IL-8-stimulated PMN function. Compared with wild-type PMNs, PMNs isolated from G-CSFR-deficient mice demonstrated markedly decreased chemotaxis to IL-8. PMN emigration into the skin of G-CSFR-deficient mice in response to IL-8 was also impaired. Significant chemotaxis defects were also seen in response to N-formyl-methionyl-leucyl-phenylalanine, zymosan-activated serum, or macrophage inflammatory protein-2. The defective chemotactic response to IL-8 does not appear to be due to impaired chemoattractant receptor function, as the number of IL-8 receptors and chemoattractant-induced calcium influx, actin polymerization, and release of gelatinase B were comparable to those of wild-type PMNs. Chemoattractant-induced adhesion of G-CSFR-deficient PMNs was significantly impaired, suggesting a defect in beta2-integrin activation. Collectively, these data demonstrate that selective defects in PMN activation are present in G-CSFR-deficient mice and indicate that G-CSF plays an important role in regulating PMN chemokine responsiveness.


Subject(s)
Chemotactic Factors/pharmacology , Neutrophil Activation , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Actins/metabolism , Animals , Antigens, CD/analysis , Calcium/metabolism , Cell Adhesion/genetics , Cell Degranulation , Chemokine CXCL2 , Chemokines/pharmacology , Chemotaxis, Leukocyte , Collagenases/metabolism , Interleukin-8/pharmacology , Matrix Metalloproteinase 9 , Mice , Mice, Mutant Strains , Monokines/pharmacology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Interleukin/analysis , Receptors, Interleukin-8A , Skin/immunology , Superoxides/metabolism , Tetradecanoylphorbol Acetate/pharmacology
12.
J Exp Med ; 188(6): 1173-84, 1998 Sep 21.
Article in English | MEDLINE | ID: mdl-9743535

ABSTRACT

Cytokines stimulate granulopoiesis through signaling via receptors whose expression is controlled by lineage-specific transcription factors. Previously, we demonstrated that granulocyte colony-stimulating factor (G-CSF) receptor mRNA was undetectable and granulocyte maturation blocked in CCAAT enhancer binding protein alpha (C/EBPalpha)-deficient mice. This phenotype is distinct from that of G-CSF receptor-/- mice, suggesting that other genes are likely to be adversely affected by loss of C/EBPalpha. Here we demonstrate loss of interleukin 6 (IL-6) receptor and IL-6-responsive colony-forming units (CFU-IL6) in C/EBPalpha-/- mice. The observed failure of granulopoiesis could be rescued by the addition of soluble IL-6 receptor and IL-6 or by retroviral transduction of G-CSF receptors, demonstrating that loss of both of these receptors contributes to the absolute block in granulocyte maturation observed in C/EBPalpha-deficient hematopoietic cells. The results of these and other studies suggest that additional C/EBPalpha target genes, possibly other cytokine receptors, are also important for the block in granulocyte differentiation observed in vivo in C/EBPalpha-deficient mice.


Subject(s)
DNA-Binding Proteins/physiology , Granulocytes/physiology , Hematopoiesis , Nuclear Proteins/physiology , Receptors, Granulocyte Colony-Stimulating Factor/biosynthesis , Receptors, Interleukin-6/biosynthesis , Transcription Factors/physiology , Up-Regulation/physiology , Animals , CCAAT-Enhancer-Binding Proteins , Cell Differentiation/genetics , Colony-Forming Units Assay , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drug Synergism , Enhancer Elements, Genetic , Fetus , Hematopoiesis/drug effects , Hematopoiesis/genetics , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Interleukin-6/pharmacology , Liver/cytology , Liver/drug effects , Liver/physiology , Mice , Mice, Knockout , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , Receptors, Granulocyte Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte Colony-Stimulating Factor/deficiency , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Interleukin-6/antagonists & inhibitors , Receptors, Interleukin-6/deficiency , Receptors, Interleukin-6/genetics , Solubility , Transcription Factors/genetics , Up-Regulation/genetics
13.
J Clin Invest ; 102(3): 483-92, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9691084

ABSTRACT

The role of mutations of the granulocyte colony-stimulating factor receptor (G-CSFR) in the pathogenesis of severe congenital neutropenia (SCN) and the subsequent development of acute myeloid leukemia (AML) is controversial. Mice carrying a targeted mutation of their G-CSFR that reproduces the mutation found in a patient with SCN and AML have been generated. The mutant G-CSFR allele is expressed in a myeloid-specific fashion at levels comparable to the wild-type allele. Mice heterozygous or homozygous for this mutation have normal levels of circulating neutrophils and no evidence for a block in myeloid maturation, indicating that resting granulopoiesis is normal. However, in response to G-CSF treatment, these mice demonstrate a significantly greater fold increase in the level of circulating neutrophils. This effect appears to be due to increased neutrophil production as the absolute number of G-CSF-responsive progenitors in the bone marrow and their proliferation in response to G-CSF is increased. Furthermore, the in vitro survival and G-CSF-dependent suppression of apoptosis of mutant neutrophils are normal. Despite this evidence for a hyperproliferative response to G-CSF, no cases of AML have been detected to date. These data demonstrate that the G-CSFR mutation found in patients with SCN is not sufficient to induce an SCN phenotype or AML in mice.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoiesis/drug effects , Neutropenia/genetics , Receptors, Granulocyte Colony-Stimulating Factor/deficiency , Acute Disease , Animals , Apoptosis/drug effects , Bone Marrow/pathology , Cell Division/drug effects , Disease Susceptibility , Female , Genotype , Humans , Leukemia, Myeloid/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelodysplastic Syndromes/etiology , Neutropenia/congenital , Neutropenia/pathology , Neutrophils/pathology , Receptors, Granulocyte Colony-Stimulating Factor/drug effects , Receptors, Granulocyte Colony-Stimulating Factor/genetics
14.
Blood ; 92(2): 353-61, 1998 Jul 15.
Article in English | MEDLINE | ID: mdl-9657731

ABSTRACT

Granulocyte colony-stimulating factor (G-CSF) is the principal growth factor regulating the production of neutrophils, yet its role in lineage commitment and terminal differentiation of hematopoietic progenitor cells is controversial. In this study, we describe a system to study the role of G-CSF receptor (G-CSFR) signals in granulocytic differentiation using retroviral transduction of G-CSFR-deficient, primary hematopoietic progenitor cells. We show that ectopic expression of wild-type G-CSFR in hematopoietic progenitor cells supports G-CSF-dependent differentiation of these cells into mature granulocytes, macrophages, megakaryocytes, and erythroid cells. Furthermore, we show that two mutant G-CSFR proteins, a truncation mutant that deletes the carboxy-terminal 96 amino acids and a chimeric receptor containing the extracellular and transmembrane domains of the G-CSFR fused to the cytoplasmic domain of the erythropoietin receptor, are able to support the production of morphologically mature, chloroacetate esterase-positive, Gr-1/Mac-1-positive neutrophils in response to G-CSF. These results demonstrate that ectopic expression of the G-CSFR in hematopoietic progenitor cells allows for multilineage differentiation and suggest that unique signals generated by the cytoplasmic domain of the G-CSFR are not required for G-CSF-dependent granulocytic differentiation.


Subject(s)
Cell Lineage/physiology , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocytes/cytology , Granulocytes/physiology , Receptors, Granulocyte Colony-Stimulating Factor/physiology , Signal Transduction , Animals , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cells, Cultured , Flow Cytometry , Granulocyte Colony-Stimulating Factor/physiology , Granulocytes/drug effects , Humans , Mice
15.
Blood ; 90(7): 2522-8, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9326216

ABSTRACT

Hematopoietic progenitor cells (HPC) can be mobilized from the bone marrow into the peripheral circulation in response to a number of stimuli including hematopoietic growth factors, cytotoxic agents, and certain chemokines. Despite significant differences in their biological activities, these stimuli result in the mobilization of HPC with a similar phenotype, suggesting that a common mechanism for mobilization may exist. In this study, the role of granulocyte colony-stimulating factor (G-CSF) in progenitor mobilization was examined using G-CSF receptor (G-CSFR)-deficient mice. In contrast to wild-type mice, no increase in circulating colony-forming cells (CFU-C), CD34+ lineage- progenitors, or day 12 colony-forming unit-spleen progenitors (CFU-S) was detected in G-CSFR-deficient mice after cyclophosphamide administration. This defect was not due to a failure to regenerate HPC following cyclophosphamide administration as the number of CFU-C in the bone marrow of G-CSFR-deficient mice was increased relative to wild-type mice. Likewise, no increase in circulating CFU-C was detected in G-CSFR-deficient mice following interleukin-8 (IL-8) administration. In contrast, mobilization of HPC in response to flt-3 ligand was nearly normal. These results show that the G-CSFR is required for mobilization in response to cyclophosphamide or IL-8 but not flt-3 ligand and suggest that the G-CSFR may play an important and previously unexpected role in HPC migration.


Subject(s)
Cyclophosphamide/pharmacology , Hematopoietic Stem Cell Mobilization , Interleukin-8/pharmacology , Membrane Proteins/pharmacology , Receptors, Granulocyte Colony-Stimulating Factor/physiology , Animals , Blood Cell Count , Bone Marrow Cells/drug effects , Colony-Forming Units Assay , Granulocyte Colony-Stimulating Factor/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Granulocyte Colony-Stimulating Factor/deficiency , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Recombinant Proteins/pharmacology , Spleen/cytology
16.
Blood ; 90(7): 2583-90, 1997 Oct 01.
Article in English | MEDLINE | ID: mdl-9326224

ABSTRACT

Multiple hematopoietic cytokines can stimulate granulopoiesis; however, their relative importance in vivo and mechanisms of action remain unclear. We recently reported that granulocyte colony-stimulating factor receptor (G-CSFR)-deficient mice have a severe quantitative defect in granulopoiesis despite which phenotypically normal neutrophils were still detected. These results confirmed a role for the G-CSFR as a major regulator of granulopoiesis in vivo, but also indicated that G-CSFR independent mechanisms of granulopoiesis must exist. To explore the role of interleukin-6 (IL-6) in granulopoiesis, we generated IL-6 x G-CSFR doubly deficient mice. The additional loss of IL-6 significantly worsened the neutropenia present in young adult G-CSFR-deficient mice; moreover, exogenous IL-6 stimulated granulopoiesis in vivo in the absence of G-CSFR signals. Near normal numbers of myeloid progenitors were detected in the bone marrow of IL-6 x G-CSFR-deficient mice and their ability to terminally differentiate into mature neutrophils was observed. These results indicate that IL-6 is an independent regulator of granulopoiesis in vivo and show that neither G-CSFR or IL-6 signals are required for the commitment of multipotential progenitors to the myeloid lineage or for their terminal differentiation.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Granulocyte Colony-Stimulating Factor/physiology , Hematopoiesis/physiology , Interleukin-6/physiology , Neutropenia/physiopathology , Receptors, Granulocyte Colony-Stimulating Factor/physiology , Receptors, Interleukin-6/physiology , Animals , Female , Hematopoietic Stem Cells/cytology , Interleukin-6/blood , Interleukin-6/deficiency , Interleukin-6/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutropenia/genetics , Receptors, Granulocyte Colony-Stimulating Factor/deficiency , Receptors, Granulocyte Colony-Stimulating Factor/genetics
17.
Immunity ; 5(5): 491-501, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8934575

ABSTRACT

We have generated mice carrying a homozygous null mutation in the granulocyte colony-stimulating factor receptor (G-CSFR) gene. G-CSFR-deficient mice have decreased numbers of phenotypically normal circulating neutrophils. Hematopoietic progenitors are decreased in the bone marrow, and the expansion and terminal differentiation of these progenitors into granulocytes is impaired. Neutrophils isolated from G-CSFR-deficient mice have an increased susceptibility to apoptosis, suggesting that the G-CSFR may also regulate neutrophil survival. These data confirm a role for the G-CSFR as a major regulator of granulopoiesis in vivo and provide evidence that the G-CSFR may regulate granulopoiesis by several mechanisms. However, the data also suggest that G-CSFR-independent mechanisms of granulopoiesis must exist.


Subject(s)
Apoptosis/immunology , Neutrophils/metabolism , Receptors, Granulocyte Colony-Stimulating Factor/deficiency , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Animals , Cell Division/immunology , Hematopoiesis/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Mice , Mice, Mutant Strains , Mutagenesis, Site-Directed/immunology , Neutrophils/immunology
18.
Blood ; 87(3): 858-64, 1996 Feb 01.
Article in English | MEDLINE | ID: mdl-8562953

ABSTRACT

The role of cytokines in the control of hematopoietic cell differentiation remains controversial. Two general models for the cytokine control of hematopoietic differentiation have been proposed. In the stochastic model, cytokines provide proliferative and survival signals to the differentiating hematopoietic cell, but they do not provide specific lineage commitment signals. In the instructive model, cytokines transmit specific signals to multipotent hematopoietic cells, thereby directing lineage commitment. To distinguish between these two models with respect to granulocyte colony-stimulating factor (G-CSF) and granulocytic differentiation, we used the 32Dcl3 cell line, which is capable of differentiating into granulocytes in response to G-CSF, 32D cells transfected with either bcl-2 or bcl-XL showed prolonged survival in medium containing no cytokine supplement. Cells surviving in these cultures developed the segmented nuclei characteristic of mature neutrophils. However, no induction of myeloperoxidase activity or increase in cathepsin G transcripts were detected. These data support a hybrid model for the role of G-CSF in granulocytic differentiation; although some features of granulocytic differentiation, namely nuclear segmentation, do not require G-CSF and appear therefore to be preprogrammed in 32D cells, the complete maturation of these cells to granulocytes appears to be dependent on G-CSF.


Subject(s)
Apoptosis , Granulocyte Colony-Stimulating Factor/physiology , Hematopoietic Stem Cells/drug effects , Proto-Oncogene Proteins/physiology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers , Cell Differentiation/drug effects , Cells, Cultured , Cytoplasmic Granules/ultrastructure , Genes, Reporter , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocytes/cytology , Hematopoietic Stem Cells/cytology , Humans , Mice , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2 , Recombinant Fusion Proteins/biosynthesis , Transfection , bcl-X Protein
19.
J Biol Chem ; 270(22): 13553-60, 1995 Jun 02.
Article in English | MEDLINE | ID: mdl-7768958

ABSTRACT

Two tyrosine kinase-dependent pathways exist for activation of the respiratory burst by polymorphonuclear leukocyte (PMN) immunoglobulin G Fc receptors. Direct ligation of Fc gamma RII activates the respiratory burst, but ligation of the glycan phosphoinositol-linked Fc gamma RIIIB does not. Instead, this receptor and the integrin complement receptor CR3 synergize in activation of the respiratory burst (Zhou, M.-J., and Brown, E. J. (1994) J. Cell Biol. 125, 1407-1416). Here we show that direct ligation of Fc gamma RII leads to activation and Triton X-100 insolubility of the Src family kinase Fgr, without effect on the related myeloid Src family member Hck. In contrast, adhesion of PMN via Fc gamma RIIIB leads to activation and Triton X-100 insolubility of Hck but not Fgr. The exclusive association of Fc gamma RIIIB with Hck activation and Triton insolubility is not solely a result of its glycan phosphoinositol anchor, since decay accelerating factor (CD55), another prominent glycan phosphoinositol-anchored PMN protein, is associated with Fgr insolubility to a greater extent than Hck. Ligation of decay accelerating factor, with or without coligation of CR3, does not activate the PMN respiratory burst. Coligation of Fc gamma RIIIB with Fc gamma RII overcomes the pertussis toxin inhibition of H2O2 production in response to direct ligation of Fc gamma RII. These data support the hypothesis that activation of Hck upon Fc gamma RIIIB ligation has a role in generation of the synergistic respiratory burst.


Subject(s)
Antigen-Antibody Complex/metabolism , Neutrophils/metabolism , Octoxynol/chemistry , Protein-Tyrosine Kinases/metabolism , Receptors, IgG/metabolism , Enzyme Activation , Humans , Neutrophils/enzymology , Protein-Tyrosine Kinases/chemistry , Respiratory Burst , Signal Transduction , Solubility
20.
Blood ; 85(2): 472-9, 1995 Jan 15.
Article in English | MEDLINE | ID: mdl-7812001

ABSTRACT

The proto-oncogene c-fgr is a member of the c-src gene family of cytoplasmic tyrosine kinases. Previous studies have suggested that it is normally expressed in neutrophils, monocytes, macrophages, and natural killer cells. c-fgr is also expressed in the B cells of certain lymphoproliferative disorders, namely, Epstein-Barr virus-associated lymphoproliferative disease, and in chronic lymphocytic leukemia, but it has not previously been detected in normal or reactive human lymphoid tissue. In this study we have determined the pattern of p55c-fgr protein expression in normal human hematopoietic and lymphoid tissues at the single-cell level using immunohistochemical and immunofluorescent techniques. We show that p55c-fgr expression is developmentally regulated with high-level expression first evident at the myelocyte stage of myeloid differentiation. In addition, we show that p55c-fgr is expressed in circulating B lymphocytes isolated from chronic lymphocytic leukemia patients but is not expressed in normal circulating B lymphocytes. Surprisingly, p55c-fgr is also expressed in a subpopulation of normal B lymphocytes, the mantle zone B lymphocytes. This demonstration that p55c-fgr is expressed in a normal B-lymphocyte subpopulation suggests that its expression in certain B-cell lymphoproliferative disorders may be an indirect consequence of, rather than a primary cause of, the neoplastic transformation process.


Subject(s)
B-Lymphocyte Subsets/metabolism , Gene Expression Regulation, Developmental , Lymph Nodes/cytology , Lymphoid Tissue/cytology , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogenes , Bone Marrow Cells , Cell Differentiation , Cells, Cultured , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Erythroblastic, Acute/pathology , Leukemia, Lymphocytic, Chronic, B-Cell/blood , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplastic Stem Cells/metabolism , Protein-Tyrosine Kinases/biosynthesis , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-hck , Tumor Cells, Cultured , src-Family Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...