Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Virology ; 446(1-2): 173-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24074579

ABSTRACT

Erythrovirus B19 (B19V) is a member of the family Parvoviridae. Infection with B19V has been linked to a variety of diseases including erythroid, thyroid, neurological and autoimmune diseases. Here we show that infection of primary CD36+ cells with B19V coincides with downregulation of thyroid, retinoid, and estrogen hormone receptors. In addition we show changes in expression of a variety of related downstream signaling genes participating in cancer and cardiac-related diseases in B19V-infected erythroid primary cells.


Subject(s)
Host-Pathogen Interactions , Myeloid Progenitor Cells/virology , Parvovirus B19, Human/physiology , Receptors, Estrogen/biosynthesis , Receptors, Retinoic Acid/biosynthesis , Receptors, Thyroid Hormone/biosynthesis , Virus Replication , CD36 Antigens/analysis , Cells, Cultured , Down-Regulation , Humans , Myeloid Progenitor Cells/chemistry , Signal Transduction
2.
Virol J ; 8: 397, 2011 Aug 10.
Article in English | MEDLINE | ID: mdl-21831263

ABSTRACT

BACKGROUND: Human immunodeficiency virus type 1 (HIV-1) viral protein R (Vpr) is a virion-associated regulatory protein that functions at several points within the viral life cycle and has been shown to accumulate primarily in the nucleus and at the nuclear envelope. However, most studies have investigated Vpr localization employing cell types irrelevant to HIV-1 pathogenesis. To gain a better understanding of how cellular phenotype might impact HIV-1 Vpr intracellular localization, Vpr localization was examined in several cell lines representing major cellular targets for HIV-1 infection within the peripheral blood, bone marrow, and central nervous system (CNS). RESULTS: Utilizing a green fluorescent protein-tagged Vpr, we detected Vpr mainly in foci inside the nucleus, at the nuclear envelope, and around the nucleoli, with dispersed accumulation in the cytoplasm of human endothelial kidney 293T cells. No differences were observed in Vpr localization pattern with respect to either the location of the tag (N- or C-terminus) or the presence of other viral proteins. Subsequently, the Vpr localization pattern was explored in two primary HIV-1 target cells within the peripheral blood: the CD4+ T lymphocyte (represented by the Jurkat CD4+ T-cell line) and the monocyte-macrophage (represented by the U-937 cell line). Vpr was found primarily in speckles within the cytoplasm of the Jurkat T cells, whereas it accumulated predominantly intranuclearly in U-937 monocytic cells. These patterns differ from that observed in a bone marrow progenitor cell line (TF-1), wherein Vpr localized mainly at the nuclear envelope with some intranuclear punctuate staining. Within the CNS, we examined two astroglioma cell lines and found that Vpr displayed a perinuclear and cytoplasmic distribution. CONCLUSIONS: The results suggest that the pattern of Vpr localization depends on cellular phenotype, probably owing to interactions between Vpr and cell type-specific host factors. These interactions, in turn, are likely coupled to specific roles that Vpr plays in each cell type within the context of the viral life cycle. Phenotype-specific Vpr localization patterns might also provide an explanation with respect to Vpr secretion or release from HIV-1-infected cells within the peripheral blood and CNS.


Subject(s)
Gene Products, vpr/analysis , HIV-1/pathogenicity , Host-Pathogen Interactions , Astrocytes/chemistry , Astrocytes/virology , CD4-Positive T-Lymphocytes/chemistry , CD4-Positive T-Lymphocytes/virology , Cell Line , Cell Nucleolus/chemistry , Cell Nucleus/chemistry , Cytoplasm/chemistry , Endothelial Cells/chemistry , Endothelial Cells/virology , Humans , Macrophages/chemistry , Macrophages/virology , Myeloid Progenitor Cells/chemistry , Myeloid Progenitor Cells/virology , Nuclear Envelope/chemistry
3.
Curr HIV Res ; 8(4): 336-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20353391

ABSTRACT

HIV-1 infection is associated with hematologic abnormalities including defective myelopoiesis. Most studies of myelopoiesis during HIV-1 infection were performed using unfractionated bone marrow-derived mononuclear cells, thus resulting in significant inter-individual variability in the numbers of cultured precursors. Here we evaluated the myelopoietic potential of circulating CD34+ progenitors by conducting a longitudinal analysis of antiretroviral therapy (ART)-induced changes of colony forming units-granulocyte and monocyte (CFU-GM) growth. Twelve HIV-infected individuals were studied longitudinally before and after initiation of ART (i.e. at a time when plasma HIV-RNA levels had become undetectable); thirty-one HIV-uninfected healthy individuals were enrolled as controls. Peripheral blood-derived CD34+ progenitors were purified by immunomagnetic sorting, and cultured in methylcellulose-based medium containing stem cell factor, granulocyte-monocyte colony-stimulating factor and interleukin-3. ART-induced changes in the proportion of CD8+ T cells expressing surface HLA-DR were also evaluated. We found that CFU-GM levels were increased in untreated HIV-infected individuals when compared to uninfected controls but declined significantly following ART, in parallel with the decline of HIV-RNA levels in plasma and with the down-regulation of HLA-DR expression on CD8+ T cells. These findings suggest that, in untreated HIV-infected individuals, chronic inflammation and/or immune activation is associated with defective myelopoiesis and accumulation of myeloid precursors. ART-induced suppression of HIV-1 replication is associated with normalization of CFU-GM levels.


Subject(s)
Antiretroviral Therapy, Highly Active , Granulocytes/physiology , HIV Infections/drug therapy , HIV Infections/pathology , Monocytes/physiology , Myeloid Progenitor Cells/physiology , Adult , Antigens, CD34/analysis , Cells, Cultured , Culture Media/chemistry , Female , HIV Infections/virology , HIV-1/isolation & purification , Humans , Longitudinal Studies , Male , Middle Aged , Myeloid Progenitor Cells/chemistry , Stem Cells
4.
J Virol ; 82(7): 3736-50, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18216121

ABSTRACT

Human cytomegalovirus (HCMV) establishes a latent infection in hematopoietic cells, from which it can reactivate to cause significant disease in immunocompromised individuals. HCMV expresses a functional homolog of the immunosuppressive cytokine interleukin-10 (termed cmvIL-10), and alternate splicing of the cmvIL-10 transcript results in expression of a latency-associated cmvIL-10 transcript (LAcmvIL-10). To determine whether LAcmvIL-10 encodes immunosuppressive functions, recombinant LAcmvIL-10 protein was generated, and its impact on major histocompatibility complex class II (MHC-II) expression was examined on granulocyte macrophage progenitor cells (GM-Ps) and monocytes. LAcmvIL-10 (and cmvIL-10) downregulated MHC-II on the surfaces of both cell types. This downregulation was associated with a decrease in total MHC-II protein and transcription of components of the MHC-II biosynthesis pathway. Unlike cmvIL-10, LAcmvIL-10 did not trigger phosphorylation of Stat3, and its ability to downregulate MHC-II was not blocked by neutralizing antibodies to the human IL-10 receptor, suggesting that LAcmvIL-10 either does not engage the cellular IL-10 receptor or utilizes it in a different manner from cmvIL-10. The impact of LAcmvIL-10 on dendritic cell (DC) maturation was also assessed. In contrast to cmvIL-10, LAcmvIL-10 did not inhibit the expression of costimulatory molecules CD40, CD80, and CD86 and the maturation marker CD83 on DCs, nor did it inhibit proinflammatory cytokines (IL-1alpha, IL-1beta, IL-6 and tumor necrosis factor alpha). Thus, LAcmvIL-10 retains some, but not all, of the immunosuppressive functions of cmvIL-10. As GM-Ps and monocytes support latent infection, expression of LAcmvIL-10 may enable HCMV to avoid immune recognition and clearance during latency.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Cytomegalovirus/immunology , Immune Tolerance , Viral Proteins/immunology , Virus Latency , Antigens, CD/analysis , Antigens, Surface/analysis , Cytokines/biosynthesis , Cytomegalovirus/physiology , Dendritic Cells/chemistry , Dendritic Cells/immunology , Down-Regulation , Flow Cytometry , Histocompatibility Antigens Class II/biosynthesis , Humans , Monocytes/chemistry , Monocytes/immunology , Myeloid Progenitor Cells/chemistry , Myeloid Progenitor Cells/immunology , Phosphorylation , Receptors, Interleukin-10/antagonists & inhibitors , STAT3 Transcription Factor/metabolism
5.
Immunity ; 24(6): 801-812, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16782035

ABSTRACT

Toll-like receptors (TLRs) are best known for their ability to recognize microbial or viral components and initiate innate immune responses. We showed here that TLRs and their coreceptors were expressed by multipotential hematopoietic stem cells, whose cell cycle entry was triggered by TLR ligation. TLR expression also extended to some of the early hematopoietic progenitors, although not the progenitor cells dedicated to megakaryocyte and erythroid differentiation. TLR signaling via the Myd88 adaptor protein drove differentiation of myeloid progenitors, bypassing some normal growth and differentiation requirements, and also drove lymphoid progenitors to become dendritic cells. CD14 contributed to the efficiency of lipopolysaccharide (LPS) recognition by stem and progenitor cells, and LPS interacted directly with the TLR4/MD-2 complex on these cells in bone marrow. Thus, the preferential pathogen-mediated stimulation of myeloid differentiation pathways may provide a means for rapid replenishment of the innate immune system during infection.


Subject(s)
Hematopoietic Stem Cells/immunology , Immunity, Innate , Multipotent Stem Cells/immunology , Toll-Like Receptors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Differentiation , Dendritic Cells/cytology , Dendritic Cells/immunology , Granulocytes/cytology , Granulocytes/immunology , Hematopoietic Stem Cells/chemistry , Hematopoietic Stem Cells/cytology , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/immunology , Mice , Multipotent Stem Cells/chemistry , Multipotent Stem Cells/cytology , Myeloid Differentiation Factor 88 , Myeloid Progenitor Cells/chemistry , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/immunology , Signal Transduction , Toll-Like Receptor 4/analysis , Toll-Like Receptor 4/metabolism , Toll-Like Receptors/analysis
6.
Article in English | MEDLINE | ID: mdl-16124415

ABSTRACT

The mechanism of anemia in severe falciparum malaria is still not completely understood. The purpose of this study was to determine whether apoptosis in the erythroid lineage causes anemia in falciparum malaria. Bone marrow aspirated from 8 severe falciparum malaria patients, 3 normal volunteers and 5 retrospective normal bone marrow smears were investigated. By light microscopic study, 5 of 8 hyperparasitemic patients had hypocellular bone marrows and erythroid hypoplasia, whereas the other 3 patients had normal cellularity. The mean myeloid : erythroid ratio of these 5 patients was significantly (p < or = 0.05) higher than normal. Apoptosis of bone marrow nucleated cells (BMNC) could be determined from the exposure of phosphatidylserine (PS) on the cell membrane but not DNA fragmentation (180-250 bp) or ultrastructural morphology. The percentages of apoptotic BMNC and apoptotic erythroid cells in bone marrow from each patient and controls varied from low to high, and were not associated with parasitemia. This study suggests that destruction of erythroid lineage, particularly through apoptosis regulation, cannot solely account for anemia in falciparum malaria.


Subject(s)
Anemia/etiology , Apoptosis , Bone Marrow Cells/pathology , Malaria, Falciparum/complications , Plasmodium falciparum/pathogenicity , Anemia/parasitology , Animals , Bone Marrow Cells/parasitology , Case-Control Studies , DNA Fragmentation , Electrophoresis, Agar Gel , Erythroid Cells/chemistry , Erythroid Cells/parasitology , Hematopoiesis , Humans , Malaria, Falciparum/parasitology , Myeloid Progenitor Cells/chemistry , Myeloid Progenitor Cells/parasitology , Phosphatidylserines/blood , Plasmodium falciparum/isolation & purification
7.
Biol Blood Marrow Transplant ; 9(9): 571-82, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14506659

ABSTRACT

Our laboratory has previously reported a nonmyelosuppressive preparative regimen for hematopoietic cell transplantation that leads to mixed chimerism and allograft tolerance in miniature swine across minor and major histocompatibility disparities. Stable chimerism persisted in most of these animals but was restricted to T cells and confined to peripheral blood. Because of the importance of myeloid and erythroid progenitors for the treatment of hematologic disorders, the objective of this study was to assess whether such cells existed in the bone marrow of these lymphoid chimeras as an indication of functional engraftment. Colony-formation assays were performed on donor inocula before infusion and on bone marrow cells harvested from the transplant recipients. Donor-origin myeloid/erythroid progenitor colonies were detected in bone marrow from 6 of 7 lymphoid chimeric recipients. A delayed donor leukocyte infusion successfully converted a stable lymphoid chimera to full multilineage chimerism within 2 weeks. Donor-origin myeloid/erythroid progenitors could be detected in the bone marrow of a host-matched recipient after myeloablation and adoptive transfer of mobilized cells from one of the engrafted lymphoid chimeras. These data suggest that even when only lymphoid chimerism is readily detected by flow cytometry, dormant myeloid/erythroid progenitors can exist and subsequent conversion to full donor chimerism can be achieved. The ability to establish multilineage engraftment and chimerism without significant toxicity may have important clinical implications for the management of nonmalignant hematopoietic disorders and hematologic malignancies.


Subject(s)
Peripheral Blood Stem Cell Transplantation/methods , Transplantation Chimera/growth & development , Transplantation Conditioning/methods , Transplantation Tolerance/immunology , Animals , Antigens, CD/analysis , Blotting, Southern , Bone Marrow Cells/chemistry , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , CD3 Complex/analysis , Colony-Forming Units Assay , Erythroid Precursor Cells/chemistry , Flow Cytometry , Granulocytes/chemistry , Histocompatibility Antigens Class I/analysis , Histocompatibility Antigens Class II , Interleukin-3/pharmacology , Leukapheresis , Lymphocytes/chemistry , Monocytes/chemistry , Multipotent Stem Cells/chemistry , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Myeloid Progenitor Cells/chemistry , Pluripotent Stem Cells/chemistry , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Polymerase Chain Reaction , Stem Cell Factor/pharmacology , Swine , Swine, Miniature , T-Lymphocytes/immunology , Tissue Donors , Transplantation Chimera/immunology
8.
Blood ; 102(7): 2436-43, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-12805065

ABSTRACT

Adult murine bone marrow hematopoietic stem cells (HSCs) can be purified by sorting Hoechst 33342-extruding side population (SP) cells. Herein we investigated whether SP cells reside within embryonic tissues and exhibit hematopoietic progenitor activity. We isolated yolk sac (YS) and embryonic tissues 7.5 to 11.5 days after coitus (dpc), resolved an SP in each, and demonstrated that these SP cells exhibit distinct phenotypic and functional characteristics throughout development. YS and embryonic SP isolated 8.0 dpc expressed vascular endothelial-cadherin (VE-cadherin) and vascular endothelial receptor 2 (Flk-1), markers not expressed by bone marrow SP but expressed by endothelial cells and progenitors. SP at this stage did not express CD45 or produce hematopoietic colonies in vitro. In contrast, SP isolated 9.5 to 11.5 dpc contained a significantly higher proportion of cells expressing cKit and CD45, markers highly expressed by bone marrow SP. Furthermore, YS SP isolated 9.5 to 11.5 dpc demonstrated 40- to 90-fold enrichment for hematopoietic progenitor activity over unfractionated tissue. Our data indicate that YS and embryonic SP cells detected prior to the onset of circulation express the highest levels of endothelial markers and do not generate blood cells in vitro; however, as development progresses, they acquire hematopoietic potential and phenotypic characteristics similar to those of bone marrow SP.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Bone Marrow/embryology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Animals , Antigens, Surface/analysis , Benzimidazoles , Biomarkers , Bone Marrow Cells/chemistry , Calcium Channel Blockers/pharmacology , Erythroid Precursor Cells/chemistry , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/physiology , Female , Fetus , Fluorescent Dyes , Hematopoietic Stem Cells/chemistry , Immunophenotyping , In Vitro Techniques , Leukocyte Common Antigens/analysis , Male , Mice , Mice, Inbred C57BL , Myeloid Progenitor Cells/chemistry , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/physiology , Pregnancy , Proto-Oncogene Proteins c-kit/analysis , Staining and Labeling , Vascular Endothelial Growth Factor Receptor-2/analysis , Verapamil/pharmacology , Yolk Sac/cytology , Yolk Sac/drug effects , Yolk Sac/physiology
9.
Cell Tissue Res ; 313(1): 55-62, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12827492

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is one of the major cytokines involved in control of haemopoiesis both in bone marrow and in extramedullar sites. Its biological activity depends upon the composition and physicochemical properties of the microenvironment provided by the supporting stroma. GM-CSF activity is modulated and controlled by the stromal heparan-sulphate proteoglycans, but their optimal interaction occurs only at low pH. We questioned whether the microenvironment organisation of the interface between stroma and haemopoietic cells provides such conditions. We studied myeloid progenitor proliferation in contact with bone marrow-derived and extramedullar stromas using electron microscopy and selective labelling of pericellular components. We present evidence that, upon interaction, the two cell types reorganise their interface both in shape and molecular composition. Haemopoietic cells extend projections that considerably increase the area of intercellular contact, and stromal cells form lamellipodia and carry out a redistribution of membrane-associated sialylated glycoconjugates and proteoglycans. Such rearrangements lead to extensive capping of negatively charged molecules at the interface between the supporting stroma and the haemopoietic cells, leading potentially to a local decrease in pH. Our results indicate that the distribution of negative charges at the cellular interface may be responsible for the selectivity of cell response to GM-CSF.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Myelopoiesis/physiology , Animals , Cell Division/physiology , Cell Line , Cell Surface Extensions/ultrastructure , Cells, Cultured , Coculture Techniques , Connective Tissue Cells/physiology , Ferritins/analysis , Fibroblasts/cytology , Fibroblasts/physiology , Glycocalyx/chemistry , Glycocalyx/ultrastructure , Glycosaminoglycans/analysis , Glycosaminoglycans/isolation & purification , Glycosaminoglycans/physiology , Hydrogen-Ion Concentration , Indoles/analysis , Mice , Mice, Inbred C3H , Microscopy, Electron , Microscopy, Electron, Scanning , Microscopy, Phase-Contrast , Myeloid Progenitor Cells/chemistry , Myeloid Progenitor Cells/physiology , Myeloid Progenitor Cells/ultrastructure , N-Acetylneuraminic Acid/metabolism , Neuraminidase/metabolism , Organometallic Compounds/analysis , Protein Binding , Proteoglycans/analysis , Proteoglycans/physiology , Pseudopodia/ultrastructure , Ruthenium Red/analysis , Ruthenium Red/pharmacology , Scattering, Radiation , Stromal Cells/chemistry , Stromal Cells/physiology , Stromal Cells/ultrastructure
10.
J Leukoc Biol ; 70(3): 455-60, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11527996

ABSTRACT

Simple methods that separate progenitor cells of different hemopoietic lineages would facilitate studies on lineage commitment and differentiation. We used an antibody specific for the chemokine receptor CCR1 to examine mononuclear cells isolated from cord blood samples. When CD34(+) cells were separated into CD34(+)CCR1(+) and CD34(+)CCR1(-) cells and plated in colony-forming assays, the granulocyte/macrophage progenitors were found almost exclusively in the CD34(+)CCR1(+) cells. In contrast, the CD34(+)CCR1(-) cells contained the majority of the erythroid progenitors. There was a highly significant difference (P<0.002) in the total percentage distribution of both granulocyte-macrophage colony-forming cells and erythroid burst-forming units between the two populations. This is the first report of separation of erythroid progenitors from granulocyte/macrophage progenitors using a chemokine receptor antibody in cord blood samples. These results suggest that at the clonogenic progenitor cell stage the expression of CCR1 might be lineage-specific. This method should prove useful for studies on erythroid progenitor and granulocyte/macrophage differentiation.


Subject(s)
Cell Culture Techniques/methods , Erythroid Precursor Cells/cytology , Myeloid Progenitor Cells/cytology , Receptors, Chemokine/biosynthesis , Antibodies/immunology , Antigens, CD34/analysis , Biomarkers/analysis , Cell Differentiation , Cell Lineage , Cells, Cultured , Chemokine CCL4 , Colony-Forming Units Assay , Culture Media, Serum-Free , Erythroid Precursor Cells/chemistry , Fetal Blood/cytology , Flow Cytometry , Granulocytes/cytology , Granulocytes/immunology , Humans , Macrophage Inflammatory Proteins/pharmacology , Macrophages/cytology , Macrophages/immunology , Myeloid Progenitor Cells/chemistry , Myeloid Progenitor Cells/immunology , RNA, Messenger/biosynthesis , Receptors, CCR1 , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology
11.
Diagn Mol Pathol ; 9(4): 184-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11129441

ABSTRACT

A translocation resulting in a fusion of ETV6 (TEL) gene at 12p13 and CBFA2 (AML1) gene at 21q22 is variably reported in 16-36% of cases of childhood acute lymphoblastic leukemia (ALL). This t(12;21)(p13;q22) is not detectable by conventional cytogenetic methods and was reported to be associated with B-cell precursor ALL with presumed favorable prognosis. We have examined 18 cases of well characterized childhood B-cell precursor ALL with cytogenetic, immunophenotypic, and clinical data for the presence of the t(12;21) using fluorescence in situ hybridization (FISH). Fourteen of the 18 cases (78%) were positive for fusion ETV6/CBFA2. One of seven adult ALL patients was positive (12% of cells positive in this 21 year old patient). By contrast, no evidence of t(12;21) by FISH was noted in two childhood T-ALL cases and 10 normal bone marrow samples. Twelve of the 14 positive childhood cases had CD13 and/or CD33 expression (myeloid markers) while only one of the four negative cases was CD13 and CD33 positive. Eight of 12 cases positive for t(12;21), and with conventional cytogenetic data, had structural and/or numerical chromosome abnormalities other than the detected t(12;21). One case had relapse with gradual increase in percentage of cells positive for t(12;21) and development of an isochromosome 21 carrying the fusion signals. The data reveal a strong association of t(12;21) with B-cell precursor ALL, especially with myeloid marker expression.


Subject(s)
Biomarkers, Tumor/genetics , Burkitt Lymphoma/genetics , Myeloid Progenitor Cells/pathology , Oncogene Proteins, Fusion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Translocation, Genetic , Adolescent , Adult , Burkitt Lymphoma/diagnosis , Burkitt Lymphoma/pathology , Child , Child, Preschool , Core Binding Factor Alpha 2 Subunit , Female , Humans , In Situ Hybridization, Fluorescence , Infant , Male , Myeloid Progenitor Cells/chemistry , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
12.
Blood ; 96(12): 3887-93, 2000 Dec 01.
Article in English | MEDLINE | ID: mdl-11090074

ABSTRACT

The t(11;19)(q23;p13.1) chromosomal translocation in acute myeloid leukemias fuses the gene encoding transcriptional elongation factor ELL to the MLL gene with consequent expression of an MLL-ELL chimeric protein. To identify potential mechanisms of leukemogenesis by MLL-ELL, its transcriptional and oncogenic properties were investigated. Fusion with MLL preserves the transcriptional elongation activity of ELL but relocalizes it from a diffuse nuclear distribution to the nuclear bodies characteristic of MLL. Using a serial replating assay, it was demonstrated that the MLL-ELL chimeric protein is capable of immortalizing clonogenic myeloid progenitors in vitro after its retroviral transduction into primary murine hematopoietic cells. However, a structure-function analysis indicates that the elongation domain is not essential for myeloid transformation because mutants lacking elongation activity retain a potent ability to immortalize myeloid progenitors. Rather, the highly conserved carboxyl terminal R4 domain is both a necessary and a sufficient contribution from ELL for the immortalizing activity associated with MLL-ELL. The R4 domain demonstrates potent transcriptional activation properties and is required for transactivation of a HoxA7 promoter by MLL-ELL in a transient transcriptional assay. These data indicate that neoplastic transformation by the MLL-ELL fusion protein is likely to result from aberrant transcriptional activation of MLL target genes. Thus, in spite of the extensive diversity of MLL fusion partners, these data, in conjunction with previous studies of MLL-ENL, suggest that conversion of MLL to a constitutive transcriptional activator may be a general model for its oncogenic conversion in myeloid leukemias. (Blood. 2000;96:3887-3893)


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/pharmacology , Leukemia, Myeloid/etiology , Myeloid Progenitor Cells/drug effects , Neoplasm Proteins , Peptide Elongation Factors/pharmacology , Proto-Oncogenes , Transcription Factors/pharmacology , Acute Disease , Amino Acid Sequence , Animals , Bone Marrow Cells , Cell Nucleus/chemistry , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , DNA-Binding Proteins/chemistry , Embryo, Mammalian/cytology , Fibroblasts/cytology , Histone-Lysine N-Methyltransferase , Interleukin-3/pharmacology , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Myeloid Progenitor Cells/chemistry , Myeloid Progenitor Cells/cytology , Myeloid-Lymphoid Leukemia Protein , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/pharmacology , Peptide Elongation Factors/genetics , Protein Structure, Tertiary , RNA Polymerase II/metabolism , Structure-Activity Relationship , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription, Genetic/drug effects , Transcriptional Elongation Factors , Transfection , Translocation, Genetic
13.
Cytometry ; 42(6): 371-8, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11135291

ABSTRACT

We developed a flow cytometric method for the enumeration and classification of nonmalignant immature granulocytes (IG). In this study, IG are defined as most immature (IG stage 1: promyelocytes and myelocytes) and as more mature (IG stage 2: metamyelocytes). Blood specimens from 46 patients with documented infectious or inflammatory disease and known presence of IG (by routine manual microscopy) were analyzed. For a reference manual differential count, we used a 400 white blood cell (WBC) differential and separated granulocytes into promyelocytes and myelocytes combined, metamyelocytes, and included band cells in the mature, segmented neutrophil population. The flow cytometric method is based on three-color staining of whole, anticoagulated blood with CD45-PerCP, CD16-FITC, and CD11b-PE-labeled monoclonal antibodies and a three-step gating procedure. The flow cytometric results were confirmed by cell sorting and microscopic evaluation of the sorted cells. A total of 10,000 events, excluding debris, were recorded per specimen and IG stage 1 (CD16-/CD11b-), IG stage 2 (CD16-/CD11b+), and mature neutrophils (CD16+/CD11b+) were categorized. Regression and correlation between flow cytometric IG and the manual differential showed y = 1.34x + 0.95, r(2) = 0.86 for IG stages 1 and 2 combined versus promyelocytes, myelocytes, and metamyelocytes. For IG stage 1 versus microscopic counts of promyelocytes and myelocytes, the results were y = 1.53x + 1.24, r(2) = 0.76; for IG stage 2 versus manual metamyelocyte count, y = 0.77x + 0.21, r(2) = 0.58. Reproducibility of the flow cytometric method showed a coefficient of variation (CV) of 6.8% for all IG combined compared with a CV of 50.2% for manual differential IG count (based on a routine 100 WBC count). Samples were found stable at least 12 h at 25 degrees C and at least 48 h at 4 degrees C for flow cytometry. After staining and lysing, the sample was stable for at least 120 min at room temperature. We analyzed samples from patients with myelodysplastic and myeloproliferative disease separately. We found that CD16- mature neutrophils falsely elevated the flow cytometric IG count. Similar results were obtained in blood from patients treated with granulocyte-colony stimulating factor (G-CSF). Although this restricts the use of the method somewhat, we believe that this flow cytometric method is useful for enumerating reactive IG, as well as for evaluating automated methods for IG identification by hematology analyzers.


Subject(s)
Flow Cytometry/methods , Granulocytes/classification , Myeloid Progenitor Cells/classification , Granulocyte Colony-Stimulating Factor/therapeutic use , Granulocytes/chemistry , Granulocytes/ultrastructure , Hematologic Diseases/blood , Hematologic Diseases/therapy , Humans , Leukocyte Common Antigens/analysis , Leukocyte Count , Macrophage-1 Antigen/analysis , Myeloid Progenitor Cells/chemistry , Myeloid Progenitor Cells/ultrastructure , Receptors, IgG/analysis , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...