201
|
Omura T, Sakai H, Murakami H. Acceleration of granulocyte colony-stimulating factor-induced neutrophilic nuclear lobulation by overexpression of Lyn tyrosine kinase. EUROPEAN JOURNAL OF BIOCHEMISTRY 2002; 269:381-9. [PMID: 11784333 DOI: 10.1046/j.0014-2956.2001.02661.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Stimulation with granulocyte colony-stimulating factor (G-CSF) induces myeloid precursor cells to differentiate into neutrophils, and tyrosine phosphorylation of certain cellular proteins is crucial to this process. However, the signaling pathways for neutrophil differentiation are still obscure. As the Src-like tyrosine kinase, Lyn, has been reported to play a role in G-CSF-induced proliferation in avian lymphoid cells, we examined its involvement in G-CSF-induced signal transduction in mammalian cells. Expression plasmids for wild-type Lyn (Lyn) and kinase-negative Lyn (LynKN) were introduced into a murine granulocyte precursor cell line, GM-I62M, that can respond to G-CSF with neutrophil differentiation, and cell lines that overexpressed these molecules (GM-Lyn, GM-LynKN) were established. Upon G-CSF stimulation, both the GM-Lyn and GM-LynKN cells began to differentiate into neutrophils, showing early morphological changes within a few days, much more rapidly than did the parental cells, which started to exhibit nuclear lobulation about 10 days after the cells were transferred to G-CSF-containing medium. However, the time course of expression of the myeloperoxidase gene, another neutrophil differentiation marker, was not affected by the overexpression of Lyn or LynKN. Therefore, in normal cells, protein interactions with Lyn, but not its kinase activity, are important for the induction of G-CSF-induced neutrophilic nuclear lobulation in mammalian granulopoiesis.
Collapse
Affiliation(s)
- Tomomi Omura
- Department of Biotechnology, Faculty of Engineering, Okayama University, Japan
| | | | | |
Collapse
|
202
|
Rao S, Furukawa K, Mizumi T, Ushida T, Tateishi T. SEM analysis of primary murine macrophage cells challenged by PE beads, in vitro and in vivo UHMWPE wear debris and evaluation of subsequent release of IL-6. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2001. [DOI: 10.1016/s0928-4931(01)00318-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
203
|
Paulus JM, Levin J, Debili N, Albert A, Vainchenker W. Genesis of clone size heterogeneity in megakaryocytic and other hemopoietic colonies: the stochastic model revisited. Exp Hematol 2001; 29:1256-69. [PMID: 11698121 DOI: 10.1016/s0301-472x(01)00728-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE We previously showed that the distributions of the numbers of doublings (NbD) undergone by individual megakaryocyte progenitors before commitment to polyploidization are markedly skewed and can consistently be fitted to straight lines when plotted on semilogarithmic coordinates. The slope of such lines, which yields the probability of polyploidization per doubling, is made less steep by stimulators of megakaryocyte colony formation and is less steep in mixed erythroid-megakaryocyte than in pure megakaryocyte colonies. Therefore, megakaryocytopoiesis provides a unique model for the study of clonal heterogeneity in a hemopoietic lineage, which is the subject of this review. DATA SOURCES Articles relevant to the interpretation of these data were selected from the authors' and public databases. DATA SYNTHESIS Exponential NbD distributions were first explained by postulating that following the assembly of thrombopoiesis-specific regulators, megakaryocyte progenitors require only a single random event to arrest proliferation and commit to polyploidization. However, this stochastic model was refuted by data indicating that intrinsic properties of individual progenitors affect the NbD they achieve. We suggest that the unequal repartition of critical compounds (including receptors, signaling molecules, and gene regulators) inherent in the stem cell-progenitor transition causes a heritable heterogeneity in megakaryocyte progenitor responsiveness to polyploidization inducers. This model would be compatible with 1) the evidence for intraclonal synchronization in megakaryocyte and other hemopoietic clones generated by committed progenitors; 2) the low probability of polyploidization of the relatively insensitive bipotent megakaryocyte progenitors; and 3) the thesis that stimulators act in part by recruiting megakaryocyte progenitor cells endowed with lesser responsiveness to polyploidization inducers and higher proliferative potential. CONCLUSION The responsiveness of individual megakaryocyte progenitors to polyploidization inducers may be a major determinant of the exponential shape of NbD distributions.
Collapse
Affiliation(s)
- J M Paulus
- Laboratory of Hematology and Service of Medical Statistics, Hôpital du Sart Tilman, University of Liège, 4000 Liège, Belgium.
| | | | | | | | | |
Collapse
|
204
|
Abstract
There has been great interest in the ex vivo expansion of human long-term repopulating hematopoietic stem cells (LTR-HSCs) for a variety of clinical applications such as umbilical cord blood transplantation. The glucoprotein130 signal, activated by a complex of interleukin 6 (IL-6) and soluble IL-6 receptor (IL-6/sIL-6R), acts dramatically in synergy with the c-Kit or Flk2/Flt3 signal to expand immature human HSCs. We demonstrate a significant ex vivo expansion of human LTR-HSCs capable of repopulating in newly discovered nonobese diabetes/Shi-severe combined immunodeficient (NOD/Shi-SCID) mice. The proportion of human CD45+ cells in recipient marrow was 10 times higher in animals receiving the cultured cells with stem cell factor, Flk2/Flt3 ligand, thrombopoietin, and IL-6/sIL-6R than in those receiving comparable numbers of fresh cord blood CD34+ cells. The expansion rate provided by this combination was estimated to be 4.2-fold by a limiting dilution method. Addition of IL-3 to the culture with the cytokine combination abrogated the repopulating ability of the expanded cells. The culture method with the IL-6/sIL-6R complex and other cytokines may pave the way for ex vivo expansion of human transplantable HSCs suitable for clinical applications.
Collapse
Affiliation(s)
- T Nakahata
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Japan.
| |
Collapse
|
205
|
Miyamoto K, Tsuji K, Maekawa T, Asano S, Nakahata T. Inhibitory effect of interleukin 3 on early development of human B-lymphopoiesis. Br J Haematol 2001; 114:690-7. [PMID: 11552999 DOI: 10.1046/j.1365-2141.2001.02956.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Interleukin 3 (IL-3) is known as a stimulator of proliferation and differentiation of non-lymphoid cells, but information about the activity of IL-3 in lymphopoiesis is limited. In the present study, we examined the effect of IL-3 on human B-lymphopoiesis using the co-culture system on a murine stromal cell line, MS-5, with stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF). Although a large number of CD45+CD19+ B cells were generated in the co-culture of cord blood CD34+ cells, the addition of IL-3 to the co-culture suppressed the B-cell generation in a dose-dependent manner. In the co-culture, CD34+IL-3 receptor alpha-chain (IL-3Ralpha)+ cells, but not IL-3Ralpha- cells, produced B cells, and IL-3 suppressed the B-cell generation from CD34+IL-3Ralpha+ cells, suggesting that IL-3 exerts an inhibitory effect through the binding to IL-3Ralpha on CD34+ cells. Because the delayed addition of IL-3 and the short-term exposure to IL-3 showed that IL-3 acted as an inhibitor at an early stage of B-cell development from CD34+ cells, before the generation of CD19+ B cells, the effect of IL-3 on the early development of B cells from immature CD34+CD38- cells was examined. Individual colonies, produced from clone-sorted CD34+CD38- cells by SCF, thrombopoietin and a complex of IL-6/soluble IL-6R with or without IL-3, were divided into two fractions and analysed for B-cell potential using co-culture on MS-5 with SCF and G-CSF, and haematopoietic potential using methylcellulose clonal culture. Although some colonies cultured without IL-3 showed both B-cell and haematopoietic potential, all the colonies cultured with IL-3 showed only haematopoietic potential. These results indicate that IL-3 has an inhibitory effect on the B-cell generation from human lymphohaematopoietic progenitor cells.
Collapse
Affiliation(s)
- K Miyamoto
- Department of Clinical Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | | | | | | |
Collapse
|
206
|
Soler C, García-Manteiga J, Valdés R, Xaus J, Comalada M, Casado FJ, Pastor-Anglada M, Celada A, Felipe A. Macrophages require different nucleoside transport systems for proliferation and activation. FASEB J 2001; 15:1979-88. [PMID: 11532978 DOI: 10.1096/fj.01-0022com] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
To evaluate the mechanisms involved in macrophage proliferation and activation, we studied the regulation of the nucleoside transport systems. In murine bone marrow-derived macrophages, the nucleosides required for DNA and RNA synthesis are recruited from the extracellular medium. M-CSF induced macrophage proliferation and DNA and RNA synthesis, whereas interferon gamma (IFN-gamma) led to activation, blocked proliferation, and induced only RNA synthesis. Macrophages express at least the concentrative systems N1 and N2 (CNT2 and CNT1 genes, respectively) and the equilibrative systems es and ei (ENT1 and ENT2 genes, respectively). Incubation with M-CSF only up-regulated the equilibrative system es. Inhibition of this transport system blocked M-CSF-dependent proliferation. Treatment with IFN-gamma only induced the concentrative N1 and N2 systems. IFN-gamma also down-regulated the increased expression of the es equilibrative system induced by M-CSF. Thus, macrophage proliferation and activation require selective regulation of nucleoside transporters and may respond to specific requirements for DNA and RNA synthesis. This report also shows that the nucleoside transporters are critical for macrophage proliferation and activation.
Collapse
Affiliation(s)
- C Soler
- Departament de Fisiologia (Biologia del Macròfag) and Fundació August Pi i Sunyer, Campus de Bellvitge, E-08028 Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
207
|
Hofmann JF, Sykora M, Redemann N, Beug H. G1-Cdk activity is required for both proliferation and viability of cytokine-dependent myeloid and erythroid cells. Oncogene 2001; 20:4198-208. [PMID: 11464286 DOI: 10.1038/sj.onc.1204550] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2001] [Revised: 04/05/2001] [Accepted: 04/12/2001] [Indexed: 11/09/2022]
Abstract
Hematopoietic cytokines are critically required for survival and cell proliferation of myeloid and erythroid progenitors. It is poorly understood how the apoptotic machinery of progenitor cells senses the absence of specific cytokines. Here we show that G1-Cdk activity is essential for cytokine-mediated viability of myeloid and erythroid progenitors. Cytokine deprivation is associated with rapid downregulation of G1-Cdk activity, cell cycle arrest, and apoptosis. Specific inhibition of G1-Cdk activity results in apoptotic cell death in the presence of saturating cytokine levels. In contrast, specific cell cycle arrest in G2/M does not affect viability. When cell proliferation is arrested by cytokine withdrawal, primary erythroid progenitors expressing v-ErbA maintain G1-Cdk activity and undergo delayed apoptosis. Cdk-inhibitors strongly enhance apoptosis in starved v-ErbA cells, indicating that sustained Cdk activity is required for protection from apoptosis by v-ErbA.
Collapse
Affiliation(s)
- J F Hofmann
- Research Institute of Molecular Pathology, Dr. Bohr Gasse 7, A-1030 Vienna, Austria
| | | | | | | |
Collapse
|
208
|
Bourette RP, De Sepulveda P, Arnaud S, Dubreuil P, Rottapel R, Mouchiroud G. Suppressor of cytokine signaling 1 interacts with the macrophage colony-stimulating factor receptor and negatively regulates its proliferation signal. J Biol Chem 2001; 276:22133-9. [PMID: 11297560 DOI: 10.1074/jbc.m101878200] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Macrophage colony-stimulating factor receptor (M-CSF-R) is a tyrosine kinase that regulates proliferation, differentiation, and cell survival during monocytic lineage development. Upon activation, M-CSF-R dimerizes and autophosphorylates on specific tyrosines, creating binding sites for several cytoplasmic SH2-containing signaling molecules that relay and modulate the M-CSF signal. Here we show that M-CSF-R interacts with suppressor of cytokine signaling 1 (Socs1), a negative regulator of various cytokine and growth factor signaling pathways. Using the yeast two-hybrid system, in vitro glutathione S-transferase-M-CSF-R pull-down, and in vivo coimmunoprecipitation experiments, we demonstrated a direct interaction between the SH2 domain of Socs1 and phosphorylated tyrosines 697 or 721 of the M-CSF-R kinase insert region. Moreover, Socs1 is tyrosine-phosphorylated in response to M-CSF. Ectopic expression of Socs1 in FDC-P1/MAC and EML hematopoietic cell lines decreased their growth rates in the presence of limiting concentrations of M-CSF. However, Socs1 expression did not totally suppress long term cell growth in the presence of saturating M-CSF concentrations, in contrast to other cytokines such as stem cell factor and interleukin 3. Taken together, these results suggest that Socs1 is an M-CSF-R-binding partner involved in negative regulation of proliferation signaling and that it differentially affects cytokine receptor signals.
Collapse
Affiliation(s)
- R P Bourette
- Centre de Génétique Moléculaire et Cellulaire, Unité Mixte de Recherche CNRS 5534, 43 Boulevard du 11 Novembre 1918, 69622 Villeurbanne, France.
| | | | | | | | | | | |
Collapse
|
209
|
Loeuillet C, Bernard G, Rémy-Martin J, Saas P, Hervé P, Douay L, Chalmers D. Distinct hematopoietic support by two human stromal cell lines. Exp Hematol 2001; 29:736-45. [PMID: 11378269 DOI: 10.1016/s0301-472x(01)00644-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
OBJECTIVE The hematopoietic microenvironment is complex, and the role of myofibroblast in its function is crucial. In order to obtain a stable model reflecting this particular cell type, we have previously established human bone marrow cell lines from primary myofibroblastic Stro1(+) population (pStro1(+)). We placed HPV16 E6 and E7 expression under the control of different promoters. Here, we have characterized and studied the hematopoietic support for two cell lines corresponding to the promoters alpha-SM (alphaSM-56 line) and SV40 (SV40-56 line). MATERIALS AND METHODS The expression profile was analyzed at the RNA level by gene array and at the protein level by Western blot, flow cytometry, and ELISA. Hematopoietic support determined using colony-forming unit (CFU) and stroma-adherent colony-forming cell (SA-CFC) assays. RESULTS The phenotype of cell lines was not significantly modified compared with primary myofibroblastic cells. They secreted a broad spectrum of hematopoietic cytokines and nonspecific mediators. The two lines allowed the growth of hematopoietic precursors and had different support capabilities. CONCLUSIONS We have extensively characterized two novel human bone marrow stromal cell lines. They retained a myofibroblastic phenotype and have substantial but different hematopoietic support capabilities. These lines provided a basis for determining stromal factors involved in stem-cell regulation.
Collapse
Affiliation(s)
- C Loeuillet
- EFS de Bourgogne-Franche Comté, Besançon, France.
| | | | | | | | | | | | | |
Collapse
|
210
|
Libby P, Aikawa M. Evolution and stabilization of vulnerable atherosclerotic plaques. JAPANESE CIRCULATION JOURNAL 2001; 65:473-9. [PMID: 11407724 DOI: 10.1253/jcj.65.473] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- P Libby
- Leducq Center for Cardiovascular Research, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
211
|
Elwood NJ, Smith CA. Current status of retroviral vector mediated gene transfer into human hematopoietic stem cells. Leuk Lymphoma 2001; 41:465-82. [PMID: 11378565 DOI: 10.3109/10428190109060338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Genetic modification of hematopoietic stem cells (HSCs) has been proposed as a treatment strategy for a variety of hematologic diseases, tracking marked cells or conferring resistance to chemotherapeutic agents. Despite early enthusiasm, the results of clinical studies involving gene transfer into HSCs has not resulted in therapeutic benefits for the vast majority of treated patients. This review describes the limitations and advances that have been made in the areas of gene transfer vectors, identification of the appropriate HSCs to target for genetic modifications and the methods used to perform gene transfer.
Collapse
Affiliation(s)
- N J Elwood
- Center for Genetic and Cellular Therapies, Department of Medicine, Box 2601, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
212
|
Affiliation(s)
- D J Anderson
- Division of Biology 216-76, Howard Hughes Medical Institute, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
213
|
Poon M, Gertz SD, Fallon JT, Wiegman P, Berman JW, Sarembock IJ, Taubman MB. Dexamethasone inhibits macrophage accumulation after balloon arterial injury in cholesterol fed rabbits. Atherosclerosis 2001; 155:371-80. [PMID: 11254907 DOI: 10.1016/s0021-9150(00)00605-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Macrophages play a critical role in the development and progression of atherosclerosis. This study was designed to examine the effect of the glucocorticoid, dexamethasone, (Dex), on macrophage accumulation after acute arterial injury. Twenty New Zealand white rabbits were fed a 2% cholesterol, 6% peanut oil, rabbit chow diet for one month prior to bilateral balloon dilatation of the femoral arteries. Ten rabbits received Dex (1 mg/kg, im.) the day before and then daily for 7 days after arterial injury; control rabbits received vehicle only. Seven days after injury, Dex treatment resulted in a 96% and 77% reduction (P < 0.002) in the mean number of macrophages accumulating in the intima and media, respectively. This effect was apparently not due to a reduction in the number of circulating monocytes or to the ability of monocytes from Dex treated animals to adhere to endothelium or migrate in response to a chemotactic signal, determined in vitro under static conditions. It was associated with a 61% reduction in monocyte chemoattractant protein-1 (MCP-1) antigen (P < 0.004) in the injured arterial wall (media+intima). Glucocorticoids may be useful in attenuating the inflammatory response and subsequent foam-cell accumulation after arterial injury.
Collapse
Affiliation(s)
- M Poon
- The Zena and Michael A. Wiener Cardiovascular Institute, Mount Sinai School of Medicine, New York, NY, 10029, USA
| | | | | | | | | | | | | |
Collapse
|
214
|
Affiliation(s)
- W S May
- University of Texas Medical Branch, Sealy Center for Oncology and Hematology, Galveston 77555-1048, USA
| |
Collapse
|
215
|
Diamond P, Shannon MF, Vadas MA, Coles LS. Cold shock domain factors activate the granulocyte-macrophage colony-stimulating factor promoter in stimulated Jurkat T cells. J Biol Chem 2001; 276:7943-51. [PMID: 11116154 DOI: 10.1074/jbc.m009836200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cold shock domain (CSD) family members have been shown to play roles in either transcriptional activation or repression of many genes in various cell types. We have previously shown that CSD proteins dbpAv and dbpB (also known as YB-1) act to repress granulocyte-macrophage colony-stimulating factor transcription in human embryonic lung (HEL) fibroblasts via binding to single-stranded DNA regions across the promoter. Here we show that the same CSD factors are involved in granulocyte-macrophage colony-stimulating factor transcriptional activation in Jurkat T cells. Unlike the mechanisms of CSD repression in HEL fibroblasts, CSD-mediated activation in Jurkat T cells is not mediated through DNA binding but presumably through protein-protein interactions via the C terminus of the CSD protein with transcription factors such as RelA/NF-kappaB p65. We demonstrate that Jurkat T cells lack truncated CSD factor subtypes present in HEL fibroblasts, which raises the possibility that the cellular content of CSD proteins may determine their final role as activators or repressors of transcription.
Collapse
Affiliation(s)
- P Diamond
- Division of Human Immunology, Hanson Centre for Cancer Research, Institute of Medical and Veterinary Science, Frome Road, Adelaide, South Australia, 5000, Australia.
| | | | | | | |
Collapse
|
216
|
Krishnaraju K, Hoffman B, Liebermann DA. Early growth response gene 1 stimulates development of hematopoietic progenitor cells along the macrophage lineage at the expense of the granulocyte and erythroid lineages. Blood 2001; 97:1298-305. [PMID: 11222373 DOI: 10.1182/blood.v97.5.1298] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Using a variety of differentiation-inducible myeloid cell lines, we previously showed that the zinc-finger transcription factor early growth response gene 1 (Egr-1) is a positive modulator of macrophage differentiation and negatively regulates granulocytic differentiation. In this study, high-efficiency retroviral transduction was used to ectopically express Egr-1 in myeloid-enriched or stem cell-enriched bone marrow cultures to explore its effect on the development of hematopoietic progenitors in vitro and in lethally irradiated mice. It was found that ectopic Egr-1 expression in normal hematopoietic progenitors stimulates development along the macrophage lineage at the expense of development along the granulocyte or erythroid lineages, regardless of the cytokine used. Moreover, Egr-1 accelerated macrophage development by suppressing the proliferative phase of the growth-to-macrophage developmental program. The remarkable ability of Egr-1 to dictate macrophage development at the expense of development along other lineages resulted in failure of Egr-1-infected hematopoietic progenitors to repopulate the bone marrow and spleen, and thereby prevent death, in lethally irradiated mice. These observations further highlight the role Egr-1 plays in monocytic differentiation and growth suppression.
Collapse
Affiliation(s)
- K Krishnaraju
- Fels Institute for Cancer Research and Molecular Biology and the Department of Biochemistry, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | |
Collapse
|
217
|
Elwood NJ, Smith CA. Current status of retroviral vector mediated gene transfer into human hematopoietic stem cells. Leuk Lymphoma 2001; 41:1-18. [PMID: 11342353 DOI: 10.3109/10428190109057950] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Genetic modification of hematopoietic stem cells (HSCs) has been proposed as a treatment strategy for a variety of hematologic diseases, tracking marked cells or conferring resistance to chemotherapeutic agents. Despite early enthusiasm, the results of clinical studies involving gene transfer into HSCs have not resulted in therapeutic benefits for the vast majority of treated patients. This review describes the limitations and advances that have been made in the areas of gene transfer vectors, identification of the appropriate HSCs to target for genetic modifications and the methods used to perform gene transfer.
Collapse
Affiliation(s)
- N J Elwood
- Center for Genetic and Cellular Therapies, Department of Medicine, Box 2601, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
218
|
Aikawa M, Rabkin E, Sugiyama S, Voglic SJ, Fukumoto Y, Furukawa Y, Shiomi M, Schoen FJ, Libby P. An HMG-CoA reductase inhibitor, cerivastatin, suppresses growth of macrophages expressing matrix metalloproteinases and tissue factor in vivo and in vitro. Circulation 2001; 103:276-83. [PMID: 11208689 DOI: 10.1161/01.cir.103.2.276] [Citation(s) in RCA: 372] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Unstable atherosclerotic plaques that cause acute coronary events usually contain abundant macrophages expressing matrix metalloproteinases (MMPs) and tissue factor (TF), molecules that probably contribute to plaque rupture and subsequent thrombus formation. Lipid lowering with HMG-CoA reductase inhibitors reduces acute coronary events. METHODS AND RESULTS To test whether lipid lowering with an HMG-CoA reductase inhibitor retards macrophage accumulation in rabbit atheroma, we administered cerivastatin to immature Watanabe heritable hyperlipidemic rabbits (cerivastatin group, n=10, cerivastatin 0.6 mg x kg(-1) x d(-1); control group, n=9, saline 0.6 mL x kg(-1) x d(-1)) for 32 weeks and measured macrophage accumulation and expression of MMPs and TF. Serum cholesterol levels after 32 weeks were 809+/-40 mg/dL (control group) and 481+/-24 mg/dL (treated group). Cerivastatin diminished accumulation of macrophages in aortic atheroma. Macrophage expression of MMP-1, MMP-3, MMP-9, and TF also decreased with cerivastatin treatment. Cerivastatin reduced the number of macrophages expressing histone mRNA (a sensitive marker of cell proliferation) detected by in situ hybridization but did not alter macrophages bearing a marker of death (TUNEL staining). Cerivastatin treatment (>or=0.01 micromol/L) also reduced growth, proteolytic activity due to MMP-9, and TF expression in cultured human monocyte/macrophages. CONCLUSIONS These results suggest that lipid lowering with HMG-CoA reductase inhibitors alters plaque biology by reducing proliferation and activation of macrophages, prominent sources of molecules responsible for plaque instability and thrombogenicity.
Collapse
Affiliation(s)
- M Aikawa
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
219
|
Wynn AA, Miyakawa K, Miyata E, Dranoff G, Takeya M, Takahashi K. Role of granulocyte/macrophage colony-stimulating factor in zymocel-induced hepatic granuloma formation. THE AMERICAN JOURNAL OF PATHOLOGY 2001; 158:131-45. [PMID: 11141486 PMCID: PMC1850246 DOI: 10.1016/s0002-9440(10)63951-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/13/2000] [Indexed: 12/20/2022]
Abstract
To examine the role of granulocyte/macrophage colony-stimulating factor (GM-CSF) in inflammatory granuloma formation, we injected GM-CSF-deficient (GM-CSF(-/-)) mice and wild-type (GM-CSF(+/+)) mice intravenously with 2 mg of zymocel, and mice were killed at various intervals for examination. In GM-CSF(-/-) mice, we demonstrated a marked delay of zymocel-induced hepatic granuloma formation until 5 days after zymocel injection with a rapid reduction in numbers of granulomas at 10 days until their disappearance. In the early phase of granuloma formation, monocyte infiltration and differentiation of monocytes into macrophages were impaired in GM-CSF(-/-) mice compared with GM-CSF(+/+) mice. The percentages of [(3)H]thymidine-labeled macrophages at 2 days after zymocel injection were lower in the GM-CSF(-/-) mice than in the GM-CSF(+/+) mice. The DNA nick-end-labeling method demonstrated increased numbers of apoptotic cells in and around hepatic granulomas of GM-CSF(-/-) mice from 8 days after zymocel injection, and electron microscopy detected apoptotic bodies. Granuloma macrophage digestion of glucan particles and activation of macrophages were similar in the two types of mice. In situ hybridization demonstrated expression of GM-CSF mRNA in the endothelial cells, hepatocytes, and some granuloma cells in the GM-CSF(+/+) mice but not in the GM-CSF(-/-) mice. These results provide evidence that GM-CSF is important for the influx of monocytes into hepatic granulomas, for differentiation of monocytes into macrophages, and for proliferation and survival of macrophages within hepatic granulomas.
Collapse
Affiliation(s)
- A A Wynn
- Second Department of Pathology, Kumamoto University School of Medicine, Kumamoto, Japan
| | | | | | | | | | | |
Collapse
|
220
|
Cook AD, Braine EL, Campbell IK, Rich MJ, Hamilton JA. Blockade of collagen-induced arthritis post-onset by antibody to granulocyte-macrophage colony-stimulating factor (GM-CSF): requirement for GM-CSF in the effector phase of disease. ARTHRITIS RESEARCH 2001; 3:293-8. [PMID: 11549370 PMCID: PMC64841 DOI: 10.1186/ar318] [Citation(s) in RCA: 148] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2000] [Revised: 05/04/2001] [Accepted: 05/11/2001] [Indexed: 12/01/2022]
Abstract
There is mounting evidence for a role of the growth factor granulocyte-macrophage colony-stimulating factor (GM-CSF) in inflammatory disease, including arthritis. In the present study, we examined the effectiveness of treatment of collagen-induced arthritis (CIA) with a neutralizing mAb to GM-CSF. DBA/1 mice were immunized for the development of CIA and treated at different times, and with different doses, with neutralizing mAb to GM-CSF or isotype control mAb. Anti-GM-CSF mAb treatment prior to the onset of arthritis, at the time of antigen challenge, was effective at ameliorating the ensuing disease. Modulation of arthritis was seen predominantly as a reduction in overall disease severity, both in terms of the number of limbs affected per mouse and the clinical score of affected limbs. Importantly, anti-GM-CSF mAb treatment ameliorated existing disease, seen both as a reduction in the number of initially affected limbs progressing and lower numbers of additional limbs becoming affected. By histology, both inflammation and cartilage destruction were reduced in anti-GM-CSF-treated mice, and the levels of tumor necrosis factor-a and IL-1beta were also reduced in joint tissue washouts of these mice. Neither humoral nor cellular immunity to type II collagen, however, was affected by anti-GM-CSF mAb treatment. These results suggest that the major effect of GM-CSF in CIA is on mediating the effector phase of the inflammatory reaction to type II collagen. The results also highlight the essential role of GM-CSF in the ongoing development of inflammation and arthritis in CIA, with possible therapeutic implications for rheumatoid arthritis.
Collapse
MESH Headings
- Animals
- Ankle Joint/drug effects
- Ankle Joint/metabolism
- Ankle Joint/pathology
- Antibodies, Blocking/administration & dosage
- Antibodies, Monoclonal/administration & dosage
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Arthritis, Experimental/prevention & control
- Cartilage, Articular/drug effects
- Cartilage, Articular/pathology
- Collagen/immunology
- Disease Models, Animal
- Dose-Response Relationship, Immunologic
- Enzyme-Linked Immunosorbent Assay
- Granulocyte-Macrophage Colony-Stimulating Factor/immunology
- Hindlimb/drug effects
- Hindlimb/pathology
- Immunization
- Interleukin-1/metabolism
- Local Lymph Node Assay
- Lymphocyte Activation/drug effects
- Male
- Mice
- Mice, Inbred DBA
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- A D Cook
- Arthritis and Inflammation Research Centre, Department of Medicine, University of Melbourne, Parkville, Victoria, Australia.
| | | | | | | | | |
Collapse
|
221
|
Abstract
Acetylcholine, catecholamines, serotonin, and histamine are classical neurotransmitters. These small molecules also play important roles in the endocrine and immune/inflammatory systems. Serotonin secreted from enterochromaffin cells of the gut epithelium regulates gut motility; histamine secreted from basophils and mast cells is a major regulator of vascular permeability and skin inflammatory responses; epinephrine is a classical hormone released from the adrenal medulla. Each of these molecules is released from neural, endocrine, or immune/inflammatory cells only in response to specific physiological stimuli. Regulated secretion is possible because amines are stored in secretory vesicles and released via a stimulus-dependent exocytotic event. Amine storage-at concentrations orders of magnitude higher than in the cytoplasm-is accomplished in turn by specific secretory vesicle transporters that recognize the amines and move them from the cytosol into the vesicle. Immunohistochemical visualization of specific vesicular amine transporters (VATs) in neuronal, endocrine, and inflammatory cells provides important new information about how amine-handling cell phenotypes arise during development and how vesicular transport is regulated during homeostatic response events. Comparison of the chemical neuroanatomy of VATs and amine biosynthetic enzymes has also revealed cell groups that express vesicular transporters but not enzymes for monoamine synthesis, and vice versa: their function and regulation is a new topic of investigation in mammalian neurobiology. The chemical neuroanatomy of the vesicular amine transporters is reviewed here. These and similar data emerging from the study of the localization of the recently characterized vesicular inhibitory and excitatory amino acid transporters will contribute to understanding chemically coded synaptic circuitry in the brain, and amine-handling neuroendocrine and immune/inflammatory cell regulation.
Collapse
Affiliation(s)
- E Weihe
- Department of Molecular Neuroimmunology, Institute of Anatomy and Cell Biology, Philipps University, Marburg, Germany.
| | | |
Collapse
|
222
|
Ziegler B, Testa U, Condorelli G, Vitelli L, Valtieri M, Peschle C. Unilineage hematopoietic differentiation in bulk and single cell culture. Stem Cells 2000; 16 Suppl 1:51-73. [PMID: 11012148 DOI: 10.1002/stem.5530160808] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The rarity of hematopoietic stem and progenitor cells (HSCs, HPCs) has hampered the analysis of cellular and molecular mechanisms underlying early hematopoiesis. Methodology for HPC purification has partially offset this limitation. A further hurdle has been represented by the heterogeneity of the analyzed HPC/precursor populations: recently, development of unilineage HPC differentiation cultures has provided homogeneous populations of hematopoietic cells, particularly in the early differentiation state, i.e., populations pertaining to a single lineage and a restricted stage of differentiation/maturation, but sufficiently large for cellular/molecular analysis. This report focuses on the development and characterization of the unilineage HPC differentiation culture systems. A section is devoted to selected cellular and molecular mechanisms underlying hematopoiesis, which have been investigated by the HPC unilineage culture approach. Finally, recent advances in the development of HPC unilineage cultures at single cell level are discussed.
Collapse
Affiliation(s)
- B Ziegler
- Department of Medicine, Eberhard-Karls University Tübingen, Germany
| | | | | | | | | | | |
Collapse
|
223
|
Cataldi A, Di Pietro R, Centurione L, Grilli A, Cutroneo G, Miscia S. Phosphatidylinositol-3-kinase activation and atypical protein kinase C zeta phosphorylation characterize the DMSO signalling in erythroleukemia cells. Cell Signal 2000; 12:667-72. [PMID: 11080619 DOI: 10.1016/s0898-6568(00)00109-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Here we provide evidence for a role of phosphatidylinositol-3-kinase (PI-3-kinase) and for its product phosphatidylinositol-3,4, 5-triphosphate (PI3,4,5P3) in the occurrence of the metabolic differentiation state induced by DMSO in murine Friend erythroleukemia cells. Of note, the activation of PI-3-kinase correlated with the modulation of the activation of another enzyme, the atypical protein kinase C zeta (aPKC zeta). In particular, the expression of PI-3-kinase was substantially unaffected by DMSO treatment while its phosphorylation and the production of PI3,4,5P3 was strongly increased within 24 h of DMSO. Such a result was paralleled by an evident phosphorylation of a PKC zeta. Treatment of the cells with the two unrelated PI-3-kinase inhibitors wortmannin and LY 294002 impaired the recovery of the number of differentiated cells, therefore indicating that PI-3-kinase might be involved in the induction of erythroid differentiation, possibly engaging a protein kinase C zeta as downstream effector.
Collapse
Affiliation(s)
- A Cataldi
- Istituto di Morfologia Umana Normale, Facoltà di Medicina e Chirurgia, Universita' G. D'Annunzio, Via dei Vestini,6, 66100, Chieti, Italy.
| | | | | | | | | | | |
Collapse
|
224
|
Carta C, Campisi S, Migliaccio G, Migliaccio AR. Erythropoietin-dependent suppression of the expression of the beta subunits of the interleukin-3 receptor during erythroid differentiation. Blood Cells Mol Dis 2000; 26:467-78. [PMID: 11112384 DOI: 10.1006/bcmd.2000.0327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To clarify how erythroid cells lose their response to interleukin-3 (IL-3), we analyzed the expression of the alpha (alpha(IL-3)) and beta (beta(IL-3)/beta(com)) subunits of its receptor in a panel of murine cell lines immortalized at different stages of hemopoietic differentiation. The panel was composed by the mast cell line 32D and by its granulo-monocytic (32D GM), granulocytic (32D G), and erythroid (32D Epo1.1 and Epo) subclones. The 32D Epo cells grow only in erythropoietin (EPO) while the Epo1.1 subclone grows in either EPO or IL-3. The phenotype of these cells is that of early (expression of globins and erythroid-specific carbonic anhydrase II) and late (also expression of the erythroid-specific band 4.1 mRNA) erythroblasts when they grow in IL-3 or EPO, respectively. All the cell lines expressed comparable levels of alpha(IL-3). In contrast, the expression of beta(IL-3)/beta(com) was restricted to cells growing in IL-3 and was barely detectable in 32D Epo and 32D Epo1.1 cells growing in EPO. When switched from EPO to IL-3, 32D Epo1.1 cells expressed 10 times more beta(IL-3)/beta(com) by rapidly activating (within 1 h) their transcription rate. When reexposed to EPO, 32D Epo1.1 cells first expressed (1-6 h) more beta(IL-3)/beta(com) (2 times) but suppressed such an expression at later time points (by 48 h). The beta(IL-3)/beta(com) mRNA half-life was also different when 32D Epo1.1 cells grew in EPO or IL-3 (2-3 h vs >5 h, respectively). These results indicate that EPO specifically induces transcriptional and posttranscriptional downmodulation of beta(IL-3)/beta(com) expression in late erythroid cells.
Collapse
Affiliation(s)
- C Carta
- Laboratory of Cell Biology, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | |
Collapse
|
225
|
Laker C, Friel J, Franz MJ, Hara T, Papadopoulos P, Ostertag W, Stocking C. Transcriptional activation of the granulocyte-macrophage colony-stimulating factor receptor gene in cell mutants. Exp Cell Res 2000; 259:1-11. [PMID: 10942574 DOI: 10.1006/excr.2000.4971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Retroviral insertional mutagenesis has proven to be a powerful in vivo approach for identifying genetic mutations involved in tumorigenesis or developmental abnormalities. Applying this approach to an in vitro system, where experimental design can be readily manipulated, would greatly increase its efficacy. In this study, we sought to determine whether retroviral insertional mutagenesis could be used to isolate cell mutants, in which the transcriptional activation of a receptor gene has occurred. Cells of the myeloid progenitor cell line FDC-P1(M), which do not express the alpha receptor subunit (GMRalpha) for granulocyte-macrophage colony-stimulating factor (GM-CSF), were infected and selected for growth in GM-CSF. Over 100 mutants were isolated at a frequency up to ninefold higher than that of uninfected controls. Expression of GMRalpha in these mutants was confirmed by blocking proliferation with GM-CSF antibodies, detection of high-affinity receptors, and Northern blot analysis. Significantly, in 7/18 mutants analyzed, gross DNA rearrangements had occurred in the GMRalpha locus. These rearrangements were demonstrated to be due to intergenic rearrangements, juxtaposing an active enhancer/promoter upstream of the GMRalpha gene. In one mutant it could be demonstrated that the wild-type allele was also expressed, providing evidence that secondary mutations had occurred. The implications of these results for retroviral insertional mutagenesis are discussed.
Collapse
Affiliation(s)
- C Laker
- Department of Cell and Virus Genetics, Heinrich-Pette-Institut für Experimentelle Virologie und Immunologie an der Universität Hamburg, Hamburg, D-20251, Germany
| | | | | | | | | | | | | |
Collapse
|
226
|
Hiroike T, Higo J, Jingami H, Toh H. Homology modeling of human leptin/leptin receptor complex. Biochem Biophys Res Commun 2000; 275:154-8. [PMID: 10944457 DOI: 10.1006/bbrc.2000.3275] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Leptin receptor mediates the weight regulatory signal carried by the adipocyte-secreted peptide hormone, leptin. It is important to understand the atomic interactions between leptin and the receptor for the therapeutic applications. However, the structure of leptin receptor has not yet been determined. Leptin shows structural similarity to G-CSF, while leptin receptor is similar in amino acid sequence to G-CSF receptor. Because of the similarity between leptin/leptin receptor complex and G-CSF/G-CSF receptor complex, we tried to build a model structure of leptin/leptin receptor complex with the crystal structure of the G-CSF/G-CSF receptor complex as the template. The obtained model for the complex was consistent with the results of the amino acid replacement and deletion experiments. The observation suggests that the model is useful to lead the experimental study on the interaction between leptin and the receptor.
Collapse
Affiliation(s)
- T Hiroike
- Biomolecular Engineering Research Institute, 6-2-3, Furuedai, Suita, 565-0874, Japan.
| | | | | | | |
Collapse
|
227
|
Tamura T, Nagamura-Inoue T, Shmeltzer Z, Kuwata T, Ozato K. ICSBP directs bipotential myeloid progenitor cells to differentiate into mature macrophages. Immunity 2000; 13:155-65. [PMID: 10981959 DOI: 10.1016/s1074-7613(00)00016-9] [Citation(s) in RCA: 247] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
During hematopoiesis, myeloid progenitor cells give rise to granulocytes and macrophages. To study the role for ICSBP, a hematopoietic cell-specific transcription factor in myeloid cell development, the gene was introduced into myeloid progenitor cells established from ICSBP-/- mice. ICSBP retrovirus-transduced cells differentiated into mature macrophages with phagocytic activity, which coincided with the induction of specific target DNA binding activity. Similar to macrophages in vivo, ICSBP-transduced cells were growth arrested, expressed many macrophage-specific genes, and responded to macrophage activation signals. Contrary to this, ICSBP transducion led to repression of granulocyte-specific genes and inhibited G-CSF-mediated granulocytic differentiation in these and other myeloid progenitor cells. Together, ICSBP has a key role in the myeloid cell lineage selection and macrophage maturation.
Collapse
Affiliation(s)
- T Tamura
- Laboratory of Molecular Growth Regulation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | |
Collapse
|
228
|
AML1-MTG8 leukemic protein induces the expression of granulocyte colony-stimulating factor (G-CSF) receptor through the up-regulation of CCAAT/enhancer binding protein epsilon. Blood 2000. [DOI: 10.1182/blood.v96.1.288] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractThe t(8;21) translocation is one of the most frequent chromosomal abnormalities associated with acute myeloid leukemia (AML). In this translocation, the AML1 (CBFA2/PEBP2aB) gene is disrupted and fused to the MTG8 (ETO) gene. The ectopic expression of the resulting AML1-MTG8 fusion gene product in L-G and 32Dcl3 murine myeloid precursor cells stimulates cell proliferation without inducing morphologic terminal differentiation into mature granulocytes in response to granulocyte-colony stimulating factor (G-CSF). This study found that the ectopic expression of AML1-MTG8 elevates the expression of the G-CSF receptor (G-CSFR). Analysis of the promoter region of the G-CSFR gene revealed that up-regulation of G-CSFR expression by AML1-MTG8 does not depend on the AML1-binding sequence, but on the C/EBP (CCAAT/enhancer binding protein) binding site. The results suggest that the overproduction of G-CSFR is at least partly mediated by C/EBPɛ, whose expression is activated by AML1-MTG8. The ectopic expression of G-CSFR in L-G cells induced cell proliferation in response to G-CSF, but did not inhibit cell differentiation into mature neutrophils. Overexpression of C/EBPɛ in L-G cells also stimulated G-CSF–dependent cell proliferation. High expression levels of G-CSFR were also found in the leukemic cells of AML patients with t(8;21). Therefore, G-CSF–dependent cell proliferation of myeloid precursor cells may be implicated in leukemogenesis.
Collapse
|
229
|
AML1-MTG8 leukemic protein induces the expression of granulocyte colony-stimulating factor (G-CSF) receptor through the up-regulation of CCAAT/enhancer binding protein epsilon. Blood 2000. [DOI: 10.1182/blood.v96.1.288.013k05_288_296] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The t(8;21) translocation is one of the most frequent chromosomal abnormalities associated with acute myeloid leukemia (AML). In this translocation, the AML1 (CBFA2/PEBP2aB) gene is disrupted and fused to the MTG8 (ETO) gene. The ectopic expression of the resulting AML1-MTG8 fusion gene product in L-G and 32Dcl3 murine myeloid precursor cells stimulates cell proliferation without inducing morphologic terminal differentiation into mature granulocytes in response to granulocyte-colony stimulating factor (G-CSF). This study found that the ectopic expression of AML1-MTG8 elevates the expression of the G-CSF receptor (G-CSFR). Analysis of the promoter region of the G-CSFR gene revealed that up-regulation of G-CSFR expression by AML1-MTG8 does not depend on the AML1-binding sequence, but on the C/EBP (CCAAT/enhancer binding protein) binding site. The results suggest that the overproduction of G-CSFR is at least partly mediated by C/EBPɛ, whose expression is activated by AML1-MTG8. The ectopic expression of G-CSFR in L-G cells induced cell proliferation in response to G-CSF, but did not inhibit cell differentiation into mature neutrophils. Overexpression of C/EBPɛ in L-G cells also stimulated G-CSF–dependent cell proliferation. High expression levels of G-CSFR were also found in the leukemic cells of AML patients with t(8;21). Therefore, G-CSF–dependent cell proliferation of myeloid precursor cells may be implicated in leukemogenesis.
Collapse
|
230
|
Barreda DR, Neumann NF, Belosevic M. Flow cytometric analysis of PKH26-labeled goldfish kidney-derived macrophages. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2000; 24:395-406. [PMID: 10736523 DOI: 10.1016/s0145-305x(99)00059-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
We recently demonstrated that a goldfish macrophage cell line (GMCL) and primary in vitro-derived kidney macrophage (IVDKM) cultures contain three distinct macrophage subpopulations. Morphological, cytochemical, functional, and flow cytometric characterization of these sub-populations suggested that they may represent cells of the macrophage lineage temporally arrested at distinct differentiation junctures of fish macrophage development (putative early progenitors, monocytes, and macrophages). In this study, we examined the proliferation and differentiation events leading to the generation of mature macrophage-like cells from goldfish kidney hematopoietic tissues. The flow cytometric studies were done after labeling macrophages with PKH26 fluorescent dye and analysis of the data using the MODFIT software. Our results showed that IVDKM cultures proliferated non-synchronously, suggesting the presence of a temporal control mechanism regulating the number of cells entering the paths towards maturation. Such control is most evident during early progenitor proliferation and differentiation events. Our results showed that proliferation may not be a requirement for differentiation of early progenitors to putative monocyte and macrophage subsets. Detailed observation of the mature macrophage-like subpopulation indicated that: 1) they appear to develop from both, the differentiation of monocyte-like cells, and direct differentiation of early progenitors in the absence of a monocyte-like stage; and (2) mature macrophage-like cells appeared to be capable of self-proliferation. Our results suggest the presence of alternate pathways of fish macrophage development other than the classical hematopoietic pathway.
Collapse
Affiliation(s)
- D R Barreda
- Department of Biological Sciences, University of Alberta, Edmonton, Canada
| | | | | |
Collapse
|
231
|
Hiroyama T, Iwama A, Morita Y, Nakamura Y, Shibuya A, Nakauchi H. Molecular cloning and characterization of CRLM-2, a novel type I cytokine receptor preferentially expressed in hematopoietic cells. Biochem Biophys Res Commun 2000; 272:224-9. [PMID: 10872831 DOI: 10.1006/bbrc.2000.2764] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A murine expressed sequence tag (EST) showing homology with erythropoietin receptor (EPOR) was identified in the EST database. Cloning of the full-length cDNA revealed a 359 amino acid novel type I cytokine receptor, designated cytokine receptor like molecule-2 (CRLM-2). While CRLM-2 lacks typical WSXWS motif, it has a significant homology with EPOR, IL-2 receptor beta and gamma, and IL-9 receptor alpha. The murine CRLM-2 gene is composed of 8 exons, and an alternative mRNA splicing generates a variant transcript encoding a soluble CRLM-2. CRLM-2 is preferentially expressed in hematopoietic cells, particularly in hematopoietic progenitors and myeloid cells. Furthermore, CRLM-2 is constitutively associated with JAK2, a well-known tyrosine kinase that transmits signals from cytokine receptors. These data strongly suggest that CRLM-2 is a novel cytokine receptor involved in the regulation of hematopoietic system.
Collapse
Affiliation(s)
- T Hiroyama
- Department of Immunology, Institute of Basic Medical Sciences, University of Tsukuba, and CREST (JST), Ibaraki, Japan
| | | | | | | | | | | |
Collapse
|
232
|
Reddy EP, Korapati A, Chaturvedi P, Rane S. IL-3 signaling and the role of Src kinases, JAKs and STATs: a covert liaison unveiled. Oncogene 2000; 19:2532-47. [PMID: 10851052 DOI: 10.1038/sj.onc.1203594] [Citation(s) in RCA: 183] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Hematopoiesis is the cumulative result of intricately regulated signal transduction cascades that are mediated by cytokines and their cognate receptors. Proper culmination of these diverse signaling pathways forms the basis for an orderly generation of different cell types and aberrations in these pathways is an underlying cause for diseases such as cancer. Over the past several years, downstream events initiated upon cytokine/growth factor stimulation have been a major focus of biomedical research. As a result, several key concepts have emerged allowing a better understanding of the complex signaling processes. A group of novel transcription factors, termed signal transducers and activators of transcription (STATs) appear to orchestrate the downstream events propagated by cytokine/growth factor interactions with their cognate receptors. Until recently, the JAK proteins were considered to be the tyrosine kinases, which dictated the levels of phosphorylation and activation of STAT proteins, forming the basis of the JAK-STAT model. However, over the past few years, increasing evidence has accumulated which indicates that at least some of the STAT protein activation may be mediated by members of the Src gene family following cytokine/growth factor stimulation. Studies have demonstrated that the Src-family of tyrosine kinases can phosphorylate and activate certain STAT proteins, in lieu of JAK kinases. In such a scenario, JAK kinases may be more crucial to phosphorylation of the cytokine/growth factor receptors and in the process create docking sites on the receptors for binding of SH2-containing proteins such as STATs, Src-kinases and other signaling intermediates. Tyrosine phosphorylation and activation of STAT proteins can be achieved either by JAKs or Src-kinases depending on the nature of STAT that is being activated. This forms the basis for the JAK-Src-STAT model proposed in this review. The concerted action of JAK kinases, members of the Src-kinase family and STAT proteins, leads to cell proliferation and cell survival, the end-point of the cytokine/growth factor stimulus. Oncogene (2000).
Collapse
Affiliation(s)
- E P Reddy
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, 3307 N Broad Street, Philadelphia, Pennsylvania, PA 19140, USA
| | | | | | | |
Collapse
|
233
|
Bromberg J, Darnell JE. The role of STATs in transcriptional control and their impact on cellular function. Oncogene 2000; 19:2468-73. [PMID: 10851045 DOI: 10.1038/sj.onc.1203476] [Citation(s) in RCA: 946] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The STAT proteins (Signal Transducers and Activators of Transcription), were identified in the last decade as transcription factors which were critical in mediating virtually all cytokine driven signaling. These proteins are latent in the cytoplasm and become activated through tyrosine phosphorylation which typically occurs through cytokine receptor associated kinases (JAKs) or growth factor receptor tyrosine kinases. Recently a number of non-receptor tyrosine kinases (for example src and abl) have been found to cause STAT phosphorylation. Phosphorylated STATs form homo- or hetero-dimers, enter the nucleus and working coordinately with other transcriptional co-activators or transcription factors lead to increased transcriptional initiation. In normal cells and in animals, ligand dependent activation of the STATs is a transient process, lasting for several minutes to several hours. In contrast, in many cancerous cell lines and tumors, where growth factor dysregulation is frequently at the heart of cellular transformation, the STAT proteins (in particular Stats 1, 3 and 5) are persistently tyrosine phosphorylated or activated. The importance of STAT activation to growth control in experiments using anti-sense molecules or dominant negative STAT protein encoding constructs performed in cell lines or studies in animals lacking specific STATs strongly indicate that STATs play an important role in controlling cell cycle progression and apoptosis. Stat1 plays an important role in growth arrest, in promoting apoptosis and is implicated as a tumor suppressor; while Stats 3 and 5 are involved in promoting cell cycle progression and cellular transformation and preventing apoptosis. Many questions remain including: (1) a better understanding of how the STAT proteins through association with other factors increase transcription initiation; (2) a more complete definition of the sets of genes which are activated by different STATs and (3) how these sets of activated genes differ as a function of cell type. Finally, in the context of many cancers, where STATs are frequently persistently activated, an understanding of the mechanisms leading to their constitutive activation and defining the potential importance of persistent STAT activation in human tumorigenesis remains. Oncogene (2000).
Collapse
Affiliation(s)
- J Bromberg
- Laboratory of Molecular Cell Biology, Rockefeller University, New York, NY 10021, USA
| | | |
Collapse
|
234
|
Mitani H, Katayama N, Araki H, Ohishi K, Kobayashi K, Suzuki H, Nishii K, Masuya M, Yasukawa K, Minami N, Shiku H. Activity of interleukin 6 in the differentiation of monocytes to macrophages and dendritic cells. Br J Haematol 2000; 109:288-95. [PMID: 10848814 DOI: 10.1046/j.1365-2141.2000.02020.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Peripheral blood monocytes are common precursor cells of dendritic cells (DCs) and macrophages. We have searched for factors with the potential to regulate the differentiation of monocytes to DCs and macrophages. When CD14+ monocytes are cultured with granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin (IL) 4, the CD14+CD1a- population, which consists of macrophages, was found in the serum-containing cultures but not in the serum-free cultures. Addition of IL-6 receptor-neutralizing monoclonal antibody (mAb) or gp130-neutralizing mAb to the serum-containing cultures resulted in a decreased population of CD14+CD1a- cells. An increase in the CD14+CD1a- population with reduction in CD14-CD1a+ DCs was observed with the addition of IL-6 to cultures, whereas IL-11, leukaemia inhibitory factor, oncostatin M or macrophage colony-stimulating factor did not affect the differentiation of monocytes in the presence of GM-CSF plus IL-4. This effect of IL-6 was blocked by tumour necrosis factor alpha (TNF-alpha), lipopolysaccharide (LPS), IL-1beta, CD40 ligand (CD40L) and transforming growth factor beta1 (TGF-beta1). Among these factors, TNF-alpha was most potent in interfering with the action of IL-6. These results suggest that IL-6 inhibits the differentiation of monocytes to DCs by promoting their differentiation toward macrophages, which is modulated by factors such as TNF-alpha, LPS, IL-1beta, CD40L and TGF-beta1.
Collapse
Affiliation(s)
- H Mitani
- The Second Department of Internal Medicine, Mie University School of Medicine, Tsu, Mie 514-8507, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
235
|
Douer D, Ramezani L, Parker J, Levine AM. All-trans-retinoic acid effects the growth, differentiation and apoptosis of normal human myeloid progenitors derived from purified CD34+ bone marrow cells. Leukemia 2000; 14:874-81. [PMID: 10803520 DOI: 10.1038/sj.leu.2401772] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We have previously shown that all-trans retinoic acid (ATRA) increases the number of CFU-GM colonies grown from unseparated human bone marrow cells with crude sources of colony stimulating factors. In this study, we further characterized the effect of ATRA on the growth of CFU-GM stimulated by individual cytokines from multiple samples of CD34+ enriched or purified human bone marrow cells. The number of IL-3- or GM-CSF-induced CFU-GM with 3 x 10(-7) M ATRA was 3.25+/-1.13, and 2.17+/-0.8-fold greater respectively, compared to controls without ATRA, while G-CSF had no effect and the ratio of colony-induced with or without ATRA was 1.06+/-0.17 (P = 0.00012). No colonies grew with ATRA + IL-6 or ATRA without a cytokine. Maximum enhancing effect on IL-3-induced CFU-GM occurred when ATRA was added on day 2, gradually diminished when delaying ATRA, and in cultures of day 9 or older adding ATRA had no effect. In 14 days liquid cultures of purified CD34+ cells with IL-3, ATRA increased the number of myeloid differentiated cells to 91-95%, compared to 37-70% with IL-3 alone. In addition, the number of apoptotic cells using the annexin V method increased after 14 days from 5.1% with IL-3 to 17.1% with IL-3 + ATRA and by the TUNEL in situ method from 10-26% to 60-95%, respectively. This study demonstrates that ATRA consistently enhances the growth of myeloid progenitors from CD34+ cells. This effect is dependent on the stimulating cytokine, suggesting the myeloid cells responding to ATRA are the less mature CFU-GMs that are targets of IL-3 and GM-CSF and not the G-CSF-responding mature progenitors. The growth stimulation by ATRA and IL-3 is also associated with granulocyte differentiation and increased apoptosis. These studies further suggest a potential role of pharmacological doses of ATRA on the development of normal human hematopoietic cells.
Collapse
Affiliation(s)
- D Douer
- Department of Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles 90033, USA
| | | | | | | |
Collapse
|
236
|
Ebihara Y, Xu MJ, Manabe A, Kikuchi A, Tanaka R, Kato S, Nakahata T, Tsuji K. Exclusive expression of G-CSF receptor on myeloid progenitors in bone marrow CD34+ cells. Br J Haematol 2000; 109:153-61. [PMID: 10848795 DOI: 10.1046/j.1365-2141.2000.01952.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Although granulocyte colony-stimulating factor (G-CSF) has been reported to act on cells of neutrophilic lineage, the administration of G-CSF to induce the mobilization of various haematopoietic progenitors into the circulation. We analysed the expression of receptors for G-CSF (G-CSFR) on human bone marrow and G-CSF-mobilized peripheral blood CD34+ cells, and examined the proliferation and differentiation capabilities of sorted CD34+G-CSFR+ and CD34+G-CSFR- cells using methylcellulose clonal culture. Flow cytometric analysis showed that G-CSFR was expressed on 14.9 +/- 4.9% of bone marrow CD34+ cells, most of which were included in CD34+CD33+ and CD34+CD38+ cell fractions. In clonal cultures, CD34+G-CSFR+ cells produced only myeloid colonies, whereas CD34+G-CSFR- cells produced erythroid bursts, megakaryocyte and multilineage colonies. When incubated with the cytokine cocktail for 5 d, CD34+G-CSFR- cells generated CD34+G-CSFR+ myeloid progenitors. In G-CSF-mobilized peripheral blood, CD34+ cells contained 10.8 +/- 5.8% of G-CSFR+ cells, most of which were also myeloid progenitors, although CD34+G-CSFR- cells contained a substantial number of myeloid progenitors. These results indicated that the expression of G-CSFR on CD34+ cells is restricted to myeloid progenitors, suggesting that the specific activity of G-CSF on myelopoiesis depends on the exclusive expression of its receptor on myeloid progenitors, and that the mobilization of various haematopoietic progenitors is not a direct effect of G-CSF in humans.
Collapse
Affiliation(s)
- Y Ebihara
- Department of Clinical Oncology, The Institute of Medical Science, The University of Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
237
|
Regulation of Jak2 tyrosine kinase by protein kinase C during macrophage differentiation of IL-3–dependent myeloid progenitor cells. Blood 2000. [DOI: 10.1182/blood.v95.5.1626.005k21_1626_1632] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Differentiation of macrophages from myeloid progenitor cells depends on a discrete balance between cell growth, survival, and differentiation signals. Interleukin-3 (IL-3) supports the growth and survival of myeloid progenitor cells through the activation of Jak2 tyrosine kinase, and macrophage differentiation has been shown to be regulated by protein kinase C (PKC). During terminal differentiation of macrophages, the cells lose their mitogenic response to IL-3 and undergo growth arrest, but the underlying signaling mechanisms have remained elusive. Here we show that in IL-3–dependent 32D myeloid progenitor cells, the differentiation-inducing PKC isoforms PKC- and PKC-δ specifically caused rapid inhibition of IL-3–induced tyrosine phosphorylation. The target for this inhibition was Jak2, and the activation of PKC by 12-O-tetradecanoyl-phorbol-13-acetate treatment also abrogated IL-3–induced tyrosine phosphorylation of Jak2 in Ba/F3 cells. The mechanism of this regulation was investigated in 32D and COS7 cells, and the inhibition of Jak2 required catalytic activity of PKC-δ and involved the phosphorylation of Jak2 on serine and threonine residues by the associated PKC-δ. Furthermore, PKC-δ inhibited the in vitro catalytic activity of Jak2, indicating that Jak2 was a direct target for PKC-δ. In 32D cells, the inhibition of Jak2 either by PKC-δ, tyrosine kinase inhibitor AG490, or IL-3 deprivation caused a similar growth arrest. Reversal of PKC-δ–mediated inhibition by the overexpression of Jak2 promoted apoptosis in differentiating 32D cells. These results demonstrate a PKC-mediated negative regulatory mechanism of cytokine signaling and Jak2, and they suggest that it serves to integrate growth-promoting and differentiation signals during macrophage differentiation.
Collapse
|
238
|
Zermati Y, Varet B, Hermine O. TGF-beta1 drives and accelerates erythroid differentiation in the epo-dependent UT-7 cell line even in the absence of erythropoietin. Exp Hematol 2000; 28:256-66. [PMID: 10720690 DOI: 10.1016/s0301-472x(99)00155-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE TGF-beta1 is a powerful inhibitor of erythropoiesis. However, its mechanisms of action are not fully elucidated yet at the cellular level. In this work we have studied the effects of TGF-beta on UT-7 cell survival, proliferation and differentiation. MATERIALS AND METHODS UT-7 cell line is strictly dependent on growth factors for cell survival, growth, and differentiation. Epo (2 U/mL) induces erythroid differentiation as assessed by up regulation of glycophorin A and the presence of 5%-10% benzidine positive cells (BPC). In contrast, even in the presence of Epo (2 U/mL), GM-CSF (1 ng/mL) inhibits erythroid differentiation. RESULTS When UT-7 cells were switched from GM-CSF to Epo, TGF-beta1 (2 ng/mL) induced a rapid (3 days [Epo+TGF-beta1] vs 8 days [Epo]) and marked erythroid differentiation (80% [Epo+TGF-beta1] vs 10% [Epo] BPC) including Hemoglobin A synthesis (HbA/HbF ratio of 1 [Epo] vs 4 [Epo+TGF-beta1]). In the presence of GM-CSF, although to a lesser extent, TGF-beta1 induced erythroid differentiation (40% BPC). This effect was not a consequence of TGF-beta1-induced apoptosis because, in the presence of Epo or GM-CSF, apoptosis occurred only at day 8 or 10, respectively. Moreover, although SCF inhibited apoptotic effect of TGF-beta1, SCF+TGF-beta1+Epo was the best combination to give rise to the highest number of hemoglobinized cells. We further demonstrated that induction of erythroid differentiation by TGF-beta1 was not due to an autocrine loop involving Epo/Epo-R or to a prolongation of the G1 phase of the cell cycle. CONCLUSION Taken together, these data suggest that TGF-beta1 is an inducer of erythroid differentiation, even stronger than Epo at the cellular level.
Collapse
Affiliation(s)
- Y Zermati
- CNRS UMR 8603 and IFR Necker, Université René Descartes (Paris V), Paris, France
| | | | | |
Collapse
|
239
|
Prevost JM, Farrell PJ, Iatrou K, Brown CB. Determinants of the functional interaction between the soluble GM-CSF receptor and the GM-CSF receptor beta-subunit. Cytokine 2000; 12:187-97. [PMID: 10704245 DOI: 10.1006/cyto.1999.0527] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The GM-CSF receptor consists of a GM-CSF specific low affinity alpha-subunit (GMRalpha) and a beta-subunit (betac) that associates with GMRalpha in the presence of GM-CSF to form a high-affinity complex. A splice variant soluble isoform of GMRalpha (solalpha) consists of the extracellular domain of GMRalpha and a unique 16-amino acid C-terminal domain. Exogenously administered solalpha is unable to associate with betac on the cell surface either in the presence or absence of GM-CSF. However, paradoxically, co-expression of solalpha with betac results in the ligand-independent association of solalpha with betac on the cell surface via the C-terminal domain of solalpha. To study the interaction and functional characteristics of the solalpha-betac complex we engineered a soluble betac-subunit (ECDbeta) and expressed it alone and with solalpha. Co-expressed but not independent sources of solalpha and ECDbeta could be co-precipitated in the absence of ligand demonstrating the extracellular domain of betac was sufficient for association with solalpha upon co-expression. However, independent sources of solalpha could associate with ECDbeta in the presence of GM-CSF as could a C-terminal deficient solalpha mutant (ECDalpha) and the addition of ECDbeta to ECDalpha and GM-CSF was associated with a conversion from a low- to high-affinity ligand-receptor complex.
Collapse
Affiliation(s)
- J M Prevost
- Alberta Bone Marrow Transplant Program, Cancer Biology Research Group, Department of Medicine and Oncology, The University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | | | | | | |
Collapse
|
240
|
Akashi K, Eto T, Shibuya T, Shimoda K, Harada M, Niho Y. Aclarubicin induces differentiation of leukemic progenitors in myelodysplastic syndrome cooperating with granulocyte colony-stimulating factor. Leuk Res 2000; 24:243-8. [PMID: 10739006 DOI: 10.1016/s0145-2126(99)00175-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We have reported that low-dose aclarubicin (ACR) therapy is effective in some patients with myelodysplastic syndrome (MDS). Here, we demonstrate that a low concentration of ACR induces the in vitro differentiation of leukemic progenitor cells from patients with MDS. ACR (0.1 ng/ml) significantly increased the number of granulocyte colony-stimulating factor (G-CSF)-dependent colonies from circulating blast cells in vitro in six out of seven MDS patients with refractory anemia with excess of blast in transformation or chronic myelomonocytic leukemia, but not in all four patients with primary acute myelogenous leukemia. In these MDS patients, the effect of ACR gradually disappeared along with the progression of MDS. Interestingly, the majority of G-CSF/ACR-dependent colonies consisted of rather differentiated myeloid cells such as myelocytes and metamyelocytes, whereas colonies formed by G-CSF alone were composed mainly of immature blastic cells. The number of G-CSF-responding progenitors significantly increased during a 24-48 h incubation with ACR alone. The circulating blasts in MDS patients expressed G-CSF receptors at unchanged levels before and after the incubation with ACR. It is suggested that ACR might increase clonogenic progenitor responsiveness to G-CSF in MDS, probably through modulating downstream signaling cascades associated with G-CSF receptors, and induce these progenitors to differentiate in response to G-CSF.
Collapse
Affiliation(s)
- K Akashi
- First Department of Internal Medicine, Faculty of Medicine, Kyushu University, Fukuoka, Japan.
| | | | | | | | | | | |
Collapse
|
241
|
Siena S, Schiavo R, Pedrazzoli P, Carlo-Stella C. Therapeutic relevance of CD34 cell dose in blood cell transplantation for cancer therapy. J Clin Oncol 2000; 18:1360-77. [PMID: 10715309 DOI: 10.1200/jco.2000.18.6.1360] [Citation(s) in RCA: 263] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To review recent advances in peripheral-blood progenitor-cell (PBPC) transplantation in order to define the optimal cell dose required for autologous and allogeneic transplantation. MATERIALS AND METHODS A search of MEDLINE was conducted to identify relevant publications. Their bibliographies were also used to identify further articles and abstracts for critical review. RESULTS The CD34(+) cell content of a graft is regarded as an accurate predictor of engraftment success. Postchemotherapy autologous PBPC transplantation with >/= 5 x 10(6) CD34(+) cells/kg body weight leads to more rapid engraftment than does transplantation of lower cell doses. Further increases in transplant cell dose further accelerate platelet but not neutrophil engraftment. Evidence that long-term hematopoietic recovery may be more accurately predicted by the subpopulation of primitive progenitors transplanted suggests that the content of CD34(+)CD33(-) and long-term culture-initiating cells in cell collection samples may be important for predicting successful engraftment, particularly in patients with poor mobilization. Allogeneic transplantation has been limited by concerns regarding graft-versus-host disease and the use of hematopoietic growth factors in donors. The risk of graft rejection and engraftment failure after HLA-mismatched allogeneic transplantation may be overcome by intensive chemoradiotherapy and the infusion of large numbers of T cell-depleted hematopoietic stem cells. CONCLUSION An optimal cell dose of >/= 8 x 10(6) CD34(+) cells/kg seems to be recommended for autologous PBPC transplantation. This dose facilitates the administration of scheduled chemotherapy on time and reduces the demand for other supportive therapies. A combination of growth factors may enable patients with poor mobilization to achieve a collection sufficient to allow transplantation. The optimum PBPC dose for allogeneic transplantation remains to be defined.
Collapse
Affiliation(s)
- S Siena
- Falck Division of Medical Oncology, Department of Hematology and Oncology, Ospedale Niguarda Ca' Granda, Milan, Italy.
| | | | | | | |
Collapse
|
242
|
Abstract
In hematopoiesis, self-renewal, proliferation, differentiation and apoptosis represent opposing decisions made by stem cells and progenitor cells, which when dysregulated can result in leukaemia. Here, we have investigated the function of Myb proteins in regulating these key cellular decisions, using the cell line FDCP-mix A4 as a model of early hematopoiesis. High concentrations of IL-3 in these cells favour self-renewal over differentiation and apoptosis. However when endogenous Myb activity was inhibited with an inducible dominant interfering protein, self-renewal was replaced by apoptosis and differentiation. Differentiation was to granulocytes and monocyte/macrophages and was closely associated with a G1-S phase block in the cell cycle. As for normal hematopoiesis, cytokine-induced terminal differentiation of FDCP-mix cells is associated with concomitant proliferation prior to its completion. However, when Myb activity was inhibited during this process, proliferation and survival were both reduced, resulting in a much lower yield of mature cells. These results indicate multiple cellular roles of Myb proteins during normal hematopoiesis.
Collapse
Affiliation(s)
- J R White
- CRC Centre for Cell and Molecular Biology, Institute of Cancer Research, London SW3 6JB, UK
| | | |
Collapse
|
243
|
Bischof RJ, Zafiropoulos D, Hamilton JA, Campbell IK. Exacerbation of acute inflammatory arthritis by the colony-stimulating factors CSF-1 and granulocyte macrophage (GM)-CSF: evidence of macrophage infiltration and local proliferation. Clin Exp Immunol 2000; 119:361-7. [PMID: 10632676 PMCID: PMC1905504 DOI: 10.1046/j.1365-2249.2000.01125.x] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CSF-1 and GM-CSF have been implicated in the pathogenesis of rheumatoid arthritis. We report the effects of CSF-1 and GM-CSF in the development of an acute methylated bovine serum albumin (mBSA)-induced murine arthritis model. Examination of histopathological features revealed that the systemic administration of CSF-1 or GM-CSF following mBSA administration into the knee resulted in the exacerbation of arthritis. This included synovial hyperplasia and joint inflammation, most evident at 7 and 14 days post-mBSA administration, and the appearance of erosive pannus tissue. The exacerbation by CSF-1 and GM-CSF was not sustained but declined in incidence and severity by 21 days post-mBSA administration, similar to the effects of IL-1beta in this model, reported here and previously. Macrophages expressing Mac-2 and F4/80 were a prominent feature of the pathology observed, particularly the infiltration of Mac-2+ macrophages seen in all mice administered CSF-1, GM-CSF or IL-1beta. Present in inflamed knees was a locally dividing population of cells which included Mac-2+ and F4/80+ macrophages. These studies demonstrate that CSF-1 and GM-CSF can exacerbate and prolong the histopathology of acute inflammatory arthritis and lend support to monocytes/macrophages being a driving influence in the pathogenesis of inflammatory arthritis.
Collapse
Affiliation(s)
- R J Bischof
- Inflammation Research Centre, The University of Melbourne, Department of Medicine at The Royal Melbourne Hospital, Parkville, Victoria, Australia.
| | | | | | | |
Collapse
|
244
|
Molnár L, Berki T, Hussain A, Németh P, Losonczy H. Detection of TNFalpha expression in the bone marrow and determination of TNFalpha production of peripheral blood mononuclear cells in myelodysplastic syndrome. Pathol Oncol Res 2000; 6:18-23. [PMID: 10749583 DOI: 10.1007/bf03032653] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
TNFalpha is a highly active cytokine which plays an important role in the regulation of apoptotic cell death, a mechanism involved in the pathophysiology of myelodysplastic syndrome (MDS). In this study we investigated the expression of TNFalpha of the bone marrow trephine biopsies by immunohistochemical method and the TNFalpha production of peripheral blood mononuclear cells by ELISA method in 15 patients affected by MDS. Five of seven patients without excess of blasts showed high or intermediate TNFalpha expression in the bone marrow biopsies, whereas two patients with excess of blasts were negative and one had low expression. The five CMML patients revealed low or intermediate expression. The production of TNFalpha by the PBMC was analysed in 10 patients, four patients with RA and two with CMML produced higher level of TNFalpha which increased after stimulation with phorbol myristic acetate, but none of the RAEB patients revealed increase in TNFalpha production. In conclusion we suppose that increased TNFalpha expression and production by PBMC may be an indirect evidence of the role of increased apoptosis in low risk MDS patients.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Anemia, Refractory/blood
- Anemia, Refractory/genetics
- Anemia, Refractory/metabolism
- Anemia, Refractory, with Excess of Blasts/blood
- Anemia, Refractory, with Excess of Blasts/genetics
- Anemia, Refractory, with Excess of Blasts/metabolism
- Bone Marrow/metabolism
- Enzyme-Linked Immunosorbent Assay
- Female
- Gene Expression Profiling
- Humans
- Immunoenzyme Techniques
- Leukemia, Myelomonocytic, Chronic/blood
- Leukemia, Myelomonocytic, Chronic/genetics
- Leukemia, Myelomonocytic, Chronic/metabolism
- Male
- Middle Aged
- Monocytes/metabolism
- Myelodysplastic Syndromes/blood
- Myelodysplastic Syndromes/genetics
- Myelodysplastic Syndromes/metabolism
- Organ Specificity
- Tumor Necrosis Factor-alpha/biosynthesis
- Tumor Necrosis Factor-alpha/genetics
Collapse
Affiliation(s)
- L Molnár
- University Medical School of Pécs, 1st Department of Internal Medicine Ifjúság str. 13, Pécs, 7624, Pécs, Hungary
| | | | | | | | | |
Collapse
|
245
|
Iwasaki H, Shimoda K, Okamura S, Otsuka T, Nagafuji K, Harada N, Ohno Y, Miyamoto T, Akashi K, Harada M, Niho Y. Production of Soluble Granulocyte Colony-Stimulating Factor Receptors from Myelomonocytic Cells. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.12.6907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Abstract
It has been speculated that a soluble form of G-CSFR might be physiologically present in humans, since G-CSFR mRNA that lacks a transmembrane domain has been identified from a human myelomonocytic cell line. Here, we demonstrate human soluble G-CSFR (sG-CSFR) of two different molecular sizes (80 and 85 kDa) on an immunoblot analysis using Abs generated against the amino-terminal, extracellular domain of the full-length G-CSFR. Both isoforms of sG-CSFR were able to bind recombinant human G-CSF (rhG-CSF). RT-PCR analysis with primers targeted outside of the transmenbrane region revealed that membrane-anchored G-CSFR is expressed at all maturation stages of purified myeloid cells, including CD34+CD13+ cells (blasts), CD11b−CD15+ cells (promyelocytes or myelocytes), CD11b+CD15+ cells (metamyelocytes and mature neutrophils), and CD14+ cells (monocytes). On the other hand, sG-CSFR mRNA was detectable in CD11b−CD15+, CD11b+CD15+, and CD14+ cells, but not in the CD34+CD13+ blast population. The serum concentration of both isoforms of sG-CSFR appeared to be correlated with the numbers of neutrophils/monocytes before and after rhG-CSF treatment in normal individuals. Thus, two isoforms of sG-CSFR are physiologically secreted from relatively mature myeloid cells and might play an important role in myelopoiesis through their binding to serum G-CSF.
Collapse
Affiliation(s)
- Hiromi Iwasaki
- *First Department of Internal Medicine, Faculty of Medicine, Kyushu University, Fukuoka, Japan; and
| | - Kazuya Shimoda
- *First Department of Internal Medicine, Faculty of Medicine, Kyushu University, Fukuoka, Japan; and
| | - Seiichi Okamura
- *First Department of Internal Medicine, Faculty of Medicine, Kyushu University, Fukuoka, Japan; and
| | - Teruhisa Otsuka
- *First Department of Internal Medicine, Faculty of Medicine, Kyushu University, Fukuoka, Japan; and
| | - Koji Nagafuji
- *First Department of Internal Medicine, Faculty of Medicine, Kyushu University, Fukuoka, Japan; and
| | - Naoki Harada
- *First Department of Internal Medicine, Faculty of Medicine, Kyushu University, Fukuoka, Japan; and
| | - Yuju Ohno
- *First Department of Internal Medicine, Faculty of Medicine, Kyushu University, Fukuoka, Japan; and
| | - Toshihiro Miyamoto
- *First Department of Internal Medicine, Faculty of Medicine, Kyushu University, Fukuoka, Japan; and
| | - Koichi Akashi
- *First Department of Internal Medicine, Faculty of Medicine, Kyushu University, Fukuoka, Japan; and
| | - Mine Harada
- †Second Department of Internal Medicine, Okayama University School of Medicine, Okayama, Japan
| | - Yoshiyuki Niho
- *First Department of Internal Medicine, Faculty of Medicine, Kyushu University, Fukuoka, Japan; and
| |
Collapse
|
246
|
Kafert S, Luther S, Böll I, Wagner K, Ganser A, Eder M. Functional analysis of a single chain chimeric alpha/beta-granulocyte-macrophage colony-stimulating factor receptor. Importance of a glutamate residue in the transmembrane region. J Biol Chem 1999; 274:33064-71. [PMID: 10551876 DOI: 10.1074/jbc.274.46.33064] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To analyze the function of each subunit of the receptor for granulocyte-macrophage colony-stimulating factor (GM-CSF), GMR, we previously generated a single-chain chimeric receptor by fusion of the extracellular and transmembrane domain from the alpha-subunit (alpha-GMR) to the intracellular part of the beta-subunit (beta-GMR) introducing an additional glutamate residue at the fusion site (alpha/beta-GMR). We demonstrated the capacity of alpha/beta-GMR to bind GM-CSF with low affinity and to induce GM-CSF-dependent activation of tyrosine kinase activity and proliferation in transfected Ba/F3 cells. To further compare the functions of wild type and chimeric receptors, we now report that this alpha/beta-GMR is sufficient to mediate morphological changes, expression of alpha(4)- and beta(1)-integrin receptor subunits, and serine-phosphorylation of Akt kinase. To analyze the function of the glutamate residue at the fusion region of alpha/beta-GMR various point mutants changing this amino acid and its position were expressed in Ba/F3 cells. None of these mutants was capable of supporting GM-CSF-dependent proliferation; however, when beta-GMR was coexpressed, GM-CSF mediated short and long term proliferation. Interestingly, some mutants but not alpha/beta-GMR can induce proliferation in the presence of an anti-alpha-GMR antibody. These data demonstrate the significance of a glutamate residue in the transmembrane region of alpha/beta-GMR for ligand-induced receptor activation.
Collapse
Affiliation(s)
- S Kafert
- Department of Hematology, Hannover Medical School, D-30625 Hannover, Germany
| | | | | | | | | | | |
Collapse
|
247
|
Nichogiannopoulou A, Trevisan M, Neben S, Friedrich C, Georgopoulos K. Defects in hemopoietic stem cell activity in Ikaros mutant mice. J Exp Med 1999; 190:1201-14. [PMID: 10544193 PMCID: PMC2195677 DOI: 10.1084/jem.190.9.1201] [Citation(s) in RCA: 177] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/1999] [Accepted: 08/19/1999] [Indexed: 11/18/2022] Open
Abstract
Here we provide evidence that the Ikaros family of DNA binding factors is critical for the activity of hemopoietic stem cells (HSCs) in the mouse. Mice homozygous for an Ikaros null mutation display a >30-fold reduction in long-term repopulation units, whereas mice homozygous for an Ikaros dominant negative mutation have no measurable activity. The defect in HSC activity is also illustrated by the ability of wild-type marrow to repopulate unconditioned Ikaros mutants. A progressive reduction in multipotent CFU-S(14) (colony-forming unit-spleen) progenitors and the earliest erythroid-restricted precursors (BFU-E [burst-forming unit-erythroid]) is also detected in the Ikaros mutant strains consistent with the reduction in HSCs. Nonetheless, the more mature clonogenic erythroid and myeloid precursors are less affected, indicating either the action of a compensatory mechanism to provide more progeny or a negative role of Ikaros at later stages of erythromyeloid differentiation. In Ikaros mutant mice, a decrease in expression of the tyrosine kinase receptors flk-2 and c-kit is observed in the lineage-depleted c-kit(+)Sca-1(+) population that is normally enriched for HSCs and may in part contribute to the early hemopoietic phenotypes manifested in the absence of Ikaros.
Collapse
Affiliation(s)
- Aliki Nichogiannopoulou
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129
| | - Maryanne Trevisan
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129
| | - Steve Neben
- Bayer Corporation, Biotechnology Division, Berkeley, California 94710
| | - Christoph Friedrich
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129
| | - Katia Georgopoulos
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129
| |
Collapse
|
248
|
Dannaeus K, Johannisson A, Nilsson K, Jönsson JI. Flt3 ligand induces the outgrowth of Mac-1+B220+ mouse bone marrow progenitor cells restricted to macrophage differentiation that coexpress early B cell-associated genes. Exp Hematol 1999; 27:1646-54. [PMID: 10560912 DOI: 10.1016/s0301-472x(99)00106-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Flt3 ligand (FL) is an important cytokine that affects the proliferation of hematopoietic stem cells and multipotent progenitors. In addition, FL seems to be strongly involved in the differentiation of B cells and macrophages. These two cell types are derived from separate hematopoietic lineages and display distinct surface markers, for instance, the pan-B cell marker B220 (CD45R) and the myelo/monocytic marker Mac-1 (CD11b), respectively. However, reports during several years have shown that some lineage markers can be coexpressed on factor-dependent progenitor cells as well as on some malignant leukemic clones. In the present study, we describe the ability of FL to induce the development and growth of Mac-1+ progenitor cells coexpressing B220 from c-kit+Lin- mouse bone marrow cells. FL was shown to be necessary but not sufficient for the development of Mac-1(-)B220+ cells, because certain other cytokines, in particular IL-6, had to be added to the cultures. An extended characterization of the cells revealed coexpression of other early B-cell markers, i.e., CD24, CD43, and c-kit. They expressed transcripts for c-fms, the receptor for macrophage-colony stimulating factor (M-CSF), and were able to develop into macrophages at high numbers, but not to other myeloid cells. By RT-PCR analysis we could also demonstrate expression of the B-cell associated genes Pax-5, Rag-2, and TdT. In contrast, Mac-1(+)B220- cells from the same cultures did not express any of the B-cell genes, and retained a broader myeloid differentiation capacity. Despite these B-cell associated features, Mac-1(+)B220- cells could not be induced towards B-cell progenitors. Our data suggest that FL triggers the activation of some B-cell associated genes in progenitor cells predestined to macrophage differentiation.
Collapse
Affiliation(s)
- K Dannaeus
- Department of Laboratory Medicine, Lund University, University Hospital MAS, Malmö, Sweden
| | | | | | | |
Collapse
|
249
|
Aritomi M, Kunishima N, Okamoto T, Kuroki R, Ota Y, Morikawa K. Atomic structure of the GCSF-receptor complex showing a new cytokine-receptor recognition scheme. Nature 1999; 401:713-7. [PMID: 10537111 DOI: 10.1038/44394] [Citation(s) in RCA: 102] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Granulocyte colony-stimulating factor (GCSF) is the principal growth factor regulating the maturation, proliferation and differentiation of the precursor cells of neutrophilic granulocytes and is used to treat neutropenia. GCSF is a member of the long-chain subtype of the class 1 cytokine superfamily, which includes growth hormone, erythropoietin, interleukin 6 and oncostatin M. Here we have determined the crystal structure of GCSF complexed to the BN-BC domains, the principal ligand-binding region of the GCSF receptor (GCSFR). The two receptor domains form a complex in a 2:2 ratio with the ligand, with a non-crystallographic pseudo-twofold axis through primarily the interdomain region and secondarily the BC domain. This structural view of a gp130-type receptor-ligand complex presents a new molecular basis for cytokine-receptor recognition.
Collapse
Affiliation(s)
- M Aritomi
- Biomolecular Engineering Research Institute, Suita-city, Osaka, Japan
| | | | | | | | | | | |
Collapse
|
250
|
Whitehead RH, Demmler K, Rockman SP, Watson NK. Clonogenic growth of epithelial cells from normal colonic mucosa from both mice and humans. Gastroenterology 1999; 117:858-65. [PMID: 10500068 DOI: 10.1016/s0016-5085(99)70344-6] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS The factors controlling the proliferation and differentiation of the colonic mucosa are unknown and have proved difficult to identify mainly because of a lack of in vitro methods for studying the proliferative cells of the mucosa. METHODS We have developed a novel method of preparing a viable single-cell suspension from isolated crypts and cloning these single cells. RESULTS We have obtained clonogenic growth from this single-cell suspension with an average of 1 colony per 10(5) cells in control cultures. Addition of conditioned medium from the LIM1863 colon carcinoma cell line increased the mean colony number to 11 +/- 3 per 10(5) cells. The cells forming the colonies are still viable after 4 weeks in culture. The epithelial nature of the cells was confirmed by ultrastructural and immunohistochemical methods with staining for keratin 8 and 18 and anti-human epithelial membrane-specific antigen and a positive result on polymerase chain reaction for keratin 19. CONCLUSIONS We have successfully cloned single cells from disaggregated colonic crypts from both human and murine colonic mucosa. We have also demonstrated the presence of an active clonogenic factor in the conditioned medium of a colon carcinoma cell line. Assays show that the clonogenic activity in the conditioned medium is not caused by the presence of any of the epidermal growth factor family of growth factors. This is the first report of a clonogenic assay for epithelial cells of normal colonic mucosa.
Collapse
Affiliation(s)
- R H Whitehead
- Ludwig Institute for Cancer Research, Melbourne Tumour Biology Branch, Royal Melbourne Hospital, Melbourne, Australia.
| | | | | | | |
Collapse
|