1
|
Migueles RP, Shaw L, Rodrigues NP, May G, Henseleit K, Anderson KGV, Goker H, Jones CM, de Bruijn MFTR, Brickman JM, Enver T. Transcriptional regulation of Hhex in hematopoiesis and hematopoietic stem cell ontogeny. Dev Biol 2017; 424:236-245. [PMID: 28189604 DOI: 10.1016/j.ydbio.2016.12.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/25/2016] [Accepted: 12/19/2016] [Indexed: 11/28/2022]
Abstract
Hematopoietic stem cells (HSCs) emerge during development via an endothelial-to-hematopoietic transition from hemogenic endothelium of the dorsal aorta (DA). Using in situ hybridization and analysis of a knock-in RedStar reporter, we show that the transcriptional regulator Hhex is expressed in endothelium of the dorsal aorta (DA) and in clusters of putative HSCs as they are specified during murine development. We exploited this observation, using the Hhex locus to define cis regulatory elements, enhancers and interacting transcription factors that are both necessary and sufficient to support gene expression in the emerging HSC. We identify an evolutionarily conserved non-coding region (ECR) in the Hhex locus with the capacity to bind the hematopoietic-affiliated transcriptional regulators Gata2, SCL, Fli1, Pu.1 and Ets1/2. This region is sufficient to drive the expression of a transgenic GFP reporter in the DA endothelium and intra-aortic hematopoietic clusters. GFP-positive AGM cells co-expressed HSC-associated markers c-Kit, CD34, VE-Cadherin, and CD45, and were capable of multipotential differentiation and long term engraftment when transplanted into myelo-ablated recipients. The Hhex ECR was also sufficient to drive expression at additional blood sites including the yolk sac blood islands, fetal liver, vitelline and umbilical arteries and the adult bone marrow, suggesting a common mechanism for Hhex regulation throughout ontogenesis of the blood system. To explore the physiological requirement for the Hhex ECR region during hematoendothelial development, we deleted the ECR element from the endogenous locus in the context of a targeted Hhex-RedStar reporter allele. Results indicate a specific requirement for the ECR in blood-associated Hhex expression during development and further demonstrate a requirement for this region in the adult HSC compartment. Taken together, our results identified the ECR region as an enhancer both necessary and sufficient for gene expression in HSC development and homeostasis. The Hhex ECR thus appears to be a core node for the convergence of the transcription factor network that governs the emergence of HSCs.
Collapse
Affiliation(s)
- Rosa Portero Migueles
- MRC Centre for Regenerative Medicine - Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, EH16 4UU Edinburgh, UK
| | - Louise Shaw
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Neil P Rodrigues
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK; The European Cancer Stem Cell Research Institute, Cardiff University, School of Biosciences, Hadyn Ellis Building, Cardiff CF24 4HQ, UK
| | - Gillian May
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK; Stem Cell Laboratory, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Korinna Henseleit
- MRC Centre for Regenerative Medicine - Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, EH16 4UU Edinburgh, UK
| | - Kathryn G V Anderson
- MRC Centre for Regenerative Medicine - Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, EH16 4UU Edinburgh, UK; The Danish Stem Cell Centre - DanStem, University of Copenhagen, 3B Blegdamsvej, DK-2200 Copenhagen, Denmark
| | - Hakan Goker
- Institute for Cancer Research, Chester Beatty Laboratories, London SW3 6JB, UK
| | - C Michael Jones
- Institute for Cancer Research, Chester Beatty Laboratories, London SW3 6JB, UK
| | - Marella F T R de Bruijn
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | - Joshua M Brickman
- MRC Centre for Regenerative Medicine - Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, EH16 4UU Edinburgh, UK; The Danish Stem Cell Centre - DanStem, University of Copenhagen, 3B Blegdamsvej, DK-2200 Copenhagen, Denmark.
| | - Tariq Enver
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK; Stem Cell Laboratory, UCL Cancer Institute, University College London, London WC1E 6DD, UK.
| |
Collapse
|
2
|
Hoxa6 potentiates short-term hemopoietic cell proliferation and extended self-renewal. Exp Hematol 2009; 37:322-33.e3. [DOI: 10.1016/j.exphem.2008.10.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2008] [Revised: 10/10/2008] [Accepted: 10/28/2008] [Indexed: 11/23/2022]
|
3
|
Wang W, Horner DN, Chen WLK, Zandstra PW, Audet J. Synergy between erythropoietin and stem cell factor during erythropoiesis can be quantitatively described without co-signaling effects. Biotechnol Bioeng 2008; 99:1261-72. [DOI: 10.1002/bit.21677] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
4
|
Göttig S, Möbest D, Rüster B, Grace B, Winter S, Seifried E, Gille J, Wieland T, Henschler R. Role of the monomeric GTPase Rho in hematopoietic progenitor cell migration and transplantation. Eur J Immunol 2006; 36:180-9. [PMID: 16323242 DOI: 10.1002/eji.200525607] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
To investigate the role of the monomeric guanosine triphosphatase (GTPase) Rho on migration of hematopoietic progenitor cells (HPC), we employed different clostridial toxins which inhibit the Rho family of GTPases. Pretreatment with C2I-C3, a cell-accessible C3 transferase fusion protein that targets Rho, increased chemokinetic migration of the factor-dependent multipotent cell line Factor Dependent Cell Paterson with mixed lineage differentiation potential (FDCP-mix) and of primary lineage marker-depleted HPC in vitro. In contrast, treatment with lethal toxin (LT) from Clostridium sordellii, which predominantly inactivates Rac, and with toxin B from C. difficile, which inactivates Rho, Rac and Cdc42, decreased in vitro migration. When HPC pretreated with LT or toxin B were transplanted into mice, homing to the bone marrow was impaired, whereas C2I-C3 treatment did not alter HPC homing. However, in a competitive hematopoietic repopulation experiment in C57BL/6 mice, pretreatment of bone marrow cells with any of the inhibitors, including the Rho inhibitor C2I-C3, resulted in suppressed donor-type hematopoiesis. Our data indicate that whereas Rac supports HPC cell cycling, migration, short-term homing and hematopoietic regeneration, Rho coordinates down-regulation of HPC migration and is required for hematopoietic regeneration.
Collapse
Affiliation(s)
- Stephan Göttig
- Institute of Transfusion Medicine and Immune Hematology, German Red Cross Blood Center, Johann Wolfgang Goethe University, Frankfurt am Main, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Warwood S, Mohammed S, Cristea IM, Evans C, Whetton AD, Gaskell SJ. Guanidination chemistry for qualitative and quantitative proteomics. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2006; 20:3245-56. [PMID: 17019669 DOI: 10.1002/rcm.2691] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The application of guanidination chemistry, the conversion of lysine into homoarginine residues, is used to illustrate several important general considerations relating to the use of differential isotope labelling for relative quantification in proteomics. The derivatisation procedure has been optimised for automation using a liquid handling station designed for proteomics. Automated application of the procedure to the analysis of in-gel tryptic digests of multiple spots from the two-dimensional gel electrophoretic (2DE) analysis of proteins from the FDCP-mix cell line shows near-universal improvement in protein identification as a result of derivatisation. This chemistry has been extended for relative quantification, applicable to matrix-assisted laser desorption/ionisation mass spectrometry (MALDI-MS) and also tandem mass spectrometry (MS/MS). It provides a robust method for the quantitative comparison of two samples that have been separated by 2DE. A peptide pair may display poor detection during MS analysis, causing their reliable relative quantification to be difficult. In such circumstances, the additional selectivity of detection provided by MS/MS can substantiate identification and allow relative quantification of these species via product ion signal ratios.
Collapse
Affiliation(s)
- Stacey Warwood
- Michael Barber Centre for Mass Spectrometry and Manchester Interdisciplinary Biocentre, University of Manchester, Manchester M1 7ND, UK
| | | | | | | | | | | |
Collapse
|
6
|
Heberlein C, Friel J, Itoh K, Medlock E, Li L, Nakayama N, Stocking C, Geldmacher M, Ostertag W. Involvement of CSF-1 in generating a stroma-independent hematopoietic stem cell line. J Cell Physiol 2005; 206:556-62. [PMID: 16250018 DOI: 10.1002/jcp.20505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The hematopoietic stem cell line, Myl-D7, is maintained by a self-renewing stem cell population that spontaneously generates myeloid, lymphoid, and erythroid progeny. MS-5 stromal cells are necessary for the growth of Myl-D7 cells. One component of the Myl-D7 cells proliferation activity released by MS-5 stromal cells was enriched by Q sepharose fractionation and shown to be colony stimulating factor-1 (CSF-1) by Western blotting, BAC1.2F5 cell bioassay and inhibition of Myl-D7 proliferation by CSF-1 antibody. The requirement of Myl-D7 cells for CSF-1 was also demonstrated independently by selecting for rare, stroma-independent Myl-D7 mutant clones able to grow without stroma and additional factors. Eighty-nine stroma-independent mutant clones were obtained and belonged to two classes. The majority of mutants did not secrete any growth promoting activity. The second, rarer class of mutants releases a factor that stimulates proliferation/survival for up to several months and approximately half of the secretors express high levels of CSF-1 mRNA. Wild type Myl-D7 grown with supernatants from the secretor cells retained the stem cell phenotype. These data suggest that CSF-1 may act as a key factor in stroma-regulated hematopoiesis and cell-cell interaction.
Collapse
|
7
|
Henschler R, Fehervizyova Z, Bistrian R, Seifried E. A mouse model to study organ homing behaviour of haemopoietic progenitor cells reveals high selectivity but low efficiency of multipotent progenitors to home into haemopoietic organs. Br J Haematol 2004; 126:111-9. [PMID: 15198741 DOI: 10.1111/j.1365-2141.2004.04995.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
To study the homing behaviour of an enriched multipotent primitive haemopoietic progenitor cell (HPC) population in mice, undifferentiated murine factor-dependent multipotent HPCs (FDCP-mix), stably transfected with the green fluorescence protein gene, were intravenously injected into congenic mice. After 2 or 24 h, cell suspensions were prepared from bone marrow, spleen, lung, liver, muscle, colon, kidney, brain or blood of the mice and analysed by flow cytometry. Using direct quantifiable determination of total HPC numbers homed per organ and a method to estimate the degree of organ contamination by HPC that were present in blood vessels within the organs before preparation, the highest absolute numbers of HPC were detected in the liver and lungs at 2 h but this was sharply decreased at 24 h, whereas HPC selectively accumulated in the bone marrow and spleen at 24 h after transplantation. Only a few HPC were detected in other organs. The seeding efficiency of homed FDCP-mix HPC to the bone marrow and spleen was approximately 1.5% and ranged between that of primary whole bone marrow cells and lineage-depleted freshly isolated bone marrow cells. Pretreatment of HPC with inhibitors of signal transduction indicated that short-term homing of multipotent HPC into haemopoietic organs is an active process requiring co-ordinated intracellular signalling through Rho family small GTPases and protein kinases. Thus, short-term homing of FDCP-mix HPC into haemopoietic organs is of low efficiency but high selectivity, and provides a system to analyse the mechanisms and manipulation of primitive HPC which saves large numbers of donor animals.
Collapse
Affiliation(s)
- R Henschler
- Institute of Transfusion Medicine and Immune Haematology, German Red Cross Blood Centre, Frankfurt, Germany.
| | | | | | | |
Collapse
|
8
|
Wandzioch E, Edling CE, Palmer RH, Carlsson L, Hallberg B. Activation of the MAP kinase pathway by c-Kit is PI-3 kinase dependent in hematopoietic progenitor/stem cell lines. Blood 2004; 104:51-7. [PMID: 14996702 DOI: 10.1182/blood-2003-07-2554] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The Steel factor (SF) and its receptor c-Kit play a critical role for various cell types at different levels in the hematopoietic hierarchy. Whether similar or distinct signaling pathways are used upon c-Kit activation in different cell types within the hematopoietic hierarchy is not known. To study c-Kit signaling pathways in the hematopoietic system we have compared c-Kit downstream signaling events in SF-dependent hematopoietic stem cell (HSC)-like cell lines to those of mast cells. Both Erk and protein kinase B (PKB)/Akt are activated by ligand-induced activation of the c-Kit receptor in the HSC-like cell lines. Surprisingly, phosphoinositide-3 (PI-3) kinase inhibitors block not only PKB/Akt activation but also activation of Raf and Erk. SF-induced activation of Ras is not affected by inhibition of PI-3 kinase. In mast cells and other more committed hematopoietic precursors, the activation of Erk by SF is not PI-3 kinase dependent. Our results suggest that a molecular signaling switch occurs during differentiation in the hematopoietic system whereby immature hematopoietic progenitor/stem cells use a PI-3 kinase-sensitive pathway in the activation of both Erk and PKB/Akt, which is then switched upon differentiation to the more commonly described PI-3 kinase-independent mitogen-activated protein (MAP) kinase pathway.
Collapse
Affiliation(s)
- Ewa Wandzioch
- Umeå Center for Molecualr Medicine, and Department of Medical Biosciences/Pathology, Umeå University, S-901 87 Umea, Sweden
| | | | | | | | | |
Collapse
|
9
|
Schroeder T, Kohlhof H, Rieber N, Just U. Notch signaling induces multilineage myeloid differentiation and up-regulates PU.1 expression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:5538-48. [PMID: 12759431 DOI: 10.4049/jimmunol.170.11.5538] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Hemopoietic commitment is initiated by and depends on activation of transcription factors. However, it is unclear whether activation of lineage-affiliated transcription factors is extrinsically regulated by to date unknown agents or is the result of a cell autonomous program. Here we show that signaling by the Notch1 transmembrane receptor instructively induces myeloid differentiation of multipotent hemopoietic progenitor cells and concomitantly up-regulates the expression of the transcription factor PU.1. Transient activation of Notch1 signaling is sufficient to irreversibly reduce self-renewal of multipotent progenitor cells accompanied by increased and accelerated differentiation along the granulocyte, macrophage, and dendritic cell lineages. Activated Notch1 has no direct influence on apoptosis of multipotent progenitor cells, shows a weak inhibition of proliferation, and does not substitute for survival and proliferation signals provided by cytokines. Activated Notch1 directly increases PU.1 RNA levels, leading to a high concentration of PU.1 protein, which has been shown to direct myeloid differentiation. These findings identify Notch as an extrinsic regulator of myeloid commitment, and the lineage-affiliated transcription factor PU.1 as a specific direct target gene of Notch.
Collapse
Affiliation(s)
- Timm Schroeder
- Institute of Clinical Molecular Biology and Tumor Genetics, GSF-National Research Center for Environment and Health, Munich, Germany
| | | | | | | |
Collapse
|
10
|
Abstract
The advent of 'genomics' technology, in particular transcript profiling, has already had a measurable impact on the drug discovery process in the areas of target identification and validation. This review is concerned with the potential application of this technology to toxicology and drug safety assessment, with particular emphasis on biomarker discovery and characterization. An advantage (or possibly a drawback!) of transcript profiling is that candidate biomarkers of toxicity can be speedily identified, with the caveat that a significant amount of subsequent experimental and bioinformatic effort needs to be expended in order to evaluate and validate them. Attention is also drawn to the critical need for robust experimental design with studies of this type and to issues associated with the analysis of large data sets. In summary, while genomics technology undoubtedly offers much that can assist drug safety assessment, its potential has yet to be realized fully in this area. However, a large amount of resource continues to be applied to 'toxicogenomics'. Tangible benefits, in terms of new biomarkers of toxicity and reduced numbers of adverse drug effects, remain realistic objectives.
Collapse
Affiliation(s)
- Jonathan D Tugwood
- Molecular Toxicology Group Safety Assessment Department, AstraZeneca Pharmaceuticals Alderley Park Macclesfield.
| | | | | |
Collapse
|
11
|
Pinto do O P, Richter K, Carlsson L. Hematopoietic progenitor/stem cells immortalized by Lhx2 generate functional hematopoietic cells in vivo. Blood 2002; 99:3939-46. [PMID: 12010792 DOI: 10.1182/blood.v99.11.3939] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are unique in their capacity to maintain blood formation following transplantation into immunocompromised hosts. Expansion of HSCs in vitro is therefore important for many clinical applications but has met with limited success because the mechanisms regulating the self-renewal process are poorly defined. We have previously shown that expression of the LIM-homeobox gene Lhx2 in hematopoietic progenitor cells derived from embryonic stem cells differentiated in vitro generates immortalized multipotent hematopoietic progenitor cell lines. However, HSCs of early embryonic origin, including those derived from differentiated embryonic stem cells, are inefficient in engrafting adult recipients upon transplantation. To address whether Lhx2 can immortalize hematopoietic progenitor/stem cells that can engraft adult recipients, we expressed Lhx2 in hematopoietic progenitor/stem cells derived from adult bone marrow. This approach allowed for the generation of immortalized growth factor-dependent hematopoietic progenitor/stem cell lines that can generate erythroid, myeloid, and lymphoid cells upon transplantation into lethally irradiated mice. When transplanted into stem cell-deficient mice, these cell lines can generate a significant proportion of circulating erythrocytes in primary, secondary, and tertiary recipients for at least 18 months. Thus, Lhx2 immortalizes multipotent hematopoietic progenitor/stem cells that can generate functional progeny following transplantation into lethally irradiated hosts and can long-term repopulate stem cell-deficient hosts.
Collapse
|
12
|
Morris EC, Carrell RW, Coughlin PB. Intracellular serpins in haemopoietic and peripheral blood cells. Br J Haematol 2001; 115:758-66. [PMID: 11843806 DOI: 10.1046/j.1365-2141.2001.03157.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- E C Morris
- Department of Haematology, University College Hospital, London, UK
| | | | | |
Collapse
|
13
|
Pinto do O P, Wandzioch E, Kolterud A, Carlsson L. Multipotent hematopoietic progenitor cells immortalized by Lhx2 self-renew by a cell nonautonomous mechanism. Exp Hematol 2001; 29:1019-28. [PMID: 11495708 DOI: 10.1016/s0301-472x(01)00666-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Direct molecular and cellular studies of hematopoietic stem cells (HSCs) are hampered by the low levels of HSCs in hematopoietic tissues. To address these issues, we generated immortalized multipotent hematopoietic precursor cell (HPC) lines by expressing the LIM-homeobox gene Lhx2 (previously LH2) in hematopoietic progenitors derived from embryonic stem cells differentiated in vitro. MATERIALS AND METHODS To validate further the relevance of the HPC lines as a model for normal HSCs, we analyzed in detail the growth requirements of HPC lines in vitro. RESULTS Lhx2 immortalized the HPC lines by a putatively novel and cell nonautonomous mechanism. Self-renewal of the HPC lines is dependent on functional Lhx2 expression. Most early-acting hematopoiesis-related growth factors show synergistic effects on the HPC lines, whereas late-acting factors do not induce differentiation by themselves. Transforming growth factor-beta(1) is a potent inhibitor of proliferation of the HPC lines. HPC lines form cobblestone areas with high efficiency when seeded onto stromal cell lines, and the cobblestone area-forming cell can be maintained in these cultures for several months. CONCLUSIONS Our data show that, in many respects, HPC lines are similar to normal hematopoietic progenitor/stem cells on the cellular level, in contrast to most previously described multipotent hematopoietic cell lines. The cell nonautonomous mechanism for immortalization of the HPC lines suggests that Lhx2 regulates, directly or indirectly, soluble mediators involved in self-renewal of the HPC lines.
Collapse
Affiliation(s)
- P Pinto do O
- Department of Microbiology, University of Umeå, Umeå, Sweden
| | | | | | | |
Collapse
|
14
|
Baird JW, Ryan KM, Hayes I, Hampson L, Heyworth CM, Clark A, Wootton M, Ansell JD, Menzel U, Hole N, Graham GJ. Differentiating embryonal stem cells are a rich source of haemopoietic gene products and suggest erythroid preconditioning of primitive haemopoietic stem cells. J Biol Chem 2001; 276:9189-98. [PMID: 11106657 DOI: 10.1074/jbc.m008354200] [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: 11/06/2022] Open
Abstract
The difficulties associated with studying molecular mechanisms important in hemopoietic stem cell (HSC) function such as the problems of purifying homogeneous stem cell populations, have prompted us to adapt the murine ES cell system as an in vitro model of HSC generation and function. We now report that careful analysis of the time course of HSC generation in differentiating ES cells allows them to be used as a source of known and novel hemopoietic gene products. We have generated a subtracted library using cDNA from ES cells collected just prior to and just following the emergence of HSCs. Analysis of this library shows it to be a rich source of known hemopoietic and hemopoietic related gene products with 44% of identifiable cDNAs falling into these camps. We have demonstrated the value of this system as a source of novel genes of relevance to HSC function by characterizing a novel membrane protein encoding cDNA that is preferentially expressed in primitive hemopoietic cells. Intriguingly, further analysis of the known components of the subtracted library is suggestive of erythroid preconditioning of the ES cell-derived HSC. We have used dot-blot and in situ analysis to indicate that this erythroid preconditioning is probably restricted to primitive but not definitive HSC.
Collapse
Affiliation(s)
- J W Baird
- Beatson Institute for Cancer Research, Cancer Research Campaign Beatson Laboratories, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Just U, Boettiger D, Kan O, Dexter TM, Spooncer E. Insertional mutagenesis as a route to identifying genes involved in self renewal of haemopoietic stem cells. Curr Top Microbiol Immunol 2001; 251:27-34. [PMID: 11036755 DOI: 10.1007/978-3-642-57276-0_4] [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: 12/18/2022]
Abstract
The genes controlling self renewal in the haemopoietic system are still unknown. Using retroviral insertional mutagenesis we have established multipotent haemopoietic stem cell lines (FDCP-mix) that possess an increased self renewal capacity in vitro. To identify genes involved in the regulation of self renewal, proviral integration sites were cloned from FDCP-mix cells and used as probes to screen independently isolated FDCP-mix cell lines for a common proviral insertion site. So far, two common integration sites have been identified, A25 and M4. A25 is rearranged in 50% of the FDCP-mix cell lines and M4 in 10%. Genes located at or near these sites are likely candidates for the control of self renewal of haemopoietic stem cells.
Collapse
Affiliation(s)
- U Just
- Paterson Institute for Cancer Research, Manchester, UK
| | | | | | | | | |
Collapse
|
16
|
Henschler R, Appel KE, Heyworth CM, Glatt H. Proliferation and differentiation of murine haemopoietic progenitor cells in stroma-free culture in the presence of metabolites of chlorinated pesticides. Toxicol In Vitro 2001; 15:31-7. [PMID: 11259867 DOI: 10.1016/s0887-2333(00)00056-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We have studied the influence of metabolites of chlorinated pesticides (lindane, pentachlorophenol, hexachlorobenzene) on proliferation and differentiation in two stroma-free murine bone marrow culture models, a multipotent progenitor cell line (FDCP-mix) and primary lineage-depleted bone marrow cells. Tetrachlorohydroquinone (Cl(4)pHQ), tetrachloro-p-benzoquinone (Cl(4p)BQ), but not their positional isomers, tetrachlorocatechol (Cl(4)oHQ) and tetrachloro-o-benzoquinone (Cl(4)oBQ), nor 2,4,6-trichlorophenol (2,4,6-Cl(3)P), were much more toxic to FDCP-mix cells cultured under conditions which lead to self-renewal than under conditions which lead to granulocyte-macrophage differentiation. Under the latter conditions, Cl(4)pHQ and Cl(4p)BQ even stimulated growth at intermediate concentration levels. In the primary cell cultures, pronounced differences were observed in the sensitivity between individual developmental pathways and between the different compounds. The percent of cells differentiating into the granulocytic lineage was increased at high concentration levels of each test compound. However, stimulatory effects on the macrophage lineage were observed at intermediate concentration levels of Cl(4)pHQ, Cl(4p)BQ and 2,4,6-Cl(3)P, and differentiation into erythrocytes was stimulated at low concentrations of 2,4,6-Cl(3)P. It is concluded that chlorinated monocyclic pesticides, after biotransformation to quinoid metabolites, may interact directly with haemopoietic progenitor cells with differential effects on self-renewal and differentiation. These mechanisms could lead to myeloplastic disorders.
Collapse
Affiliation(s)
- R Henschler
- Institute of Transfusion Medicine and Immune Haematology, Sandhofstrasse 1, D-60528 Frankfurt, Germany.
| | | | | | | |
Collapse
|
17
|
Schroeder T, Lange C, Strehl J, Just U. Generation of functionally mature dendritic cells from the multipotential stem cell line FDCP-mix. Br J Haematol 2000. [DOI: 10.1046/j.1365-2141.2000.02421.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
18
|
Schroeder T, Lange C, Strehl J, Just U. Generation of functionally mature dendritic cells from the multipotential stem cell line FDCP-mix. Br J Haematol 2000. [DOI: 10.1111/j.1365-2141.2000.02421.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
19
|
Baird JW, Nibbs RJ, Komai-Koma M, Connolly JA, Ottersbach K, Clark-Lewis I, Liew FY, Graham GJ. ESkine, a novel beta-chemokine, is differentially spliced to produce secretable and nuclear targeted isoforms. J Biol Chem 1999; 274:33496-503. [PMID: 10559234 DOI: 10.1074/jbc.274.47.33496] [Citation(s) in RCA: 39] [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
Using the murine embryonal stem cell system, we have identified a novel gene encoding a highly divergent member of the beta-chemokine family of proinflammatory mediators and have called this protein ESkine. Much of the coding sequence for ESkine overlaps with the 3'-end of a novel interleukin 11 receptor alpha-like sequence on murine chromosome 4. ESkine is produced as two splice variants. One of these variants encodes a classical chemokine with an associated signal peptide, while the other variant (PESKY) possesses the main body of the chemokine but has replaced the signal peptide with an alternative stretch of amino acids that allows for nuclear targeting of this isoform. This differential splicing arises as a result of alternative 5' exon usage. These differentially spliced forms are expressed at discrete tissue loci. Thus, while ESkine is highly expressed in the placenta, PESKY is mainly expressed in the Testes and brain and weakly in the developing embryo. Studies on the proinflammatory properties of ESkine reveal it to be active in inducing polarization of CD4(+) T cells but to be inactive on other hemopoietic cellular populations.
Collapse
Affiliation(s)
- J W Baird
- Beatson Institute for Cancer Research, Cancer Research Campaign Beatson Laboratories, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Thomson AM, Roberts NA, Sloane Stanley JA, Souyri M, Wood WG. Haemopoietic progenitor cell lines generated by the myeloproliferative leukaemia virus: a model system to analyse murine and human lineage-affiliated genes. Br J Haematol 1999; 107:33-48. [PMID: 10520023 DOI: 10.1046/j.1365-2141.1999.01688.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Multipotential progenitor and stem cells occur with a low frequency in haemopoietic tissue. As a result, it is often difficult to obtain sufficient numbers of cells to undertake many of the assays that would be informative about the molecular events involved in the regulation of lineage-affiliated genes within these multipotent cells. To circumvent this problem, we have used the myeloproliferative leukaemia virus (MPLV) to generate a phenotypically diverse array of haemopoietic progenitors from adult mouse bone marrow and embryonic blood. These cells could be expanded to perform a variety of analyses that would not previously have been possible using analogous primary cells. The validity of these assays was supported by the observation that the phenotype of several MPLV-infected lines was very similar to previously described primary haemopoietic progenitor cells. By using mice transgenic for the human alpha and beta globin gene clusters, we have shown that human genes may also be investigated. In addition, this strategy has a wide potential applicability including the rescue of haemopoietic progenitors from mouse embryos lacking genes critical for their survival as well as the study of any haemopoietic gene for which an appropriate transgenic mouse is available.
Collapse
Affiliation(s)
- A M Thomson
- MRC Molecular Haematology Unit, Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, U.K
| | | | | | | | | |
Collapse
|
21
|
Mcl-1 in Transgenic Mice Promotes Survival in a Spectrum of Hematopoietic Cell Types and Immortalization in the Myeloid Lineage. Blood 1998. [DOI: 10.1182/blood.v92.9.3226.421k49_3226_3239] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mcl-1 is a member of the Bcl-2 family that is expressed in early monocyte differentiation and that can promote viability on transfection into immature myeloid cells. However, the effects of Mcl-1 are generally short lived compared with those of Bcl-2 and are not obvious in some transfectants. To further explore the effects of this gene, mice were produced that expressed Mcl-1 as a transgene in hematolymphoid tissues. The Mcl-1 transgene was found to cause moderate viability enhancement in a wide range of hematopoietic cell types, including lymphoid (B and T) as well as myeloid cells at both immature and mature stages of differentiation. However, enhanced hematopoietic capacity in transgenic bone marrow and spleen was not reflected in any change in pool sizes in the peripheral blood. In addition, among transgenic cells, mature T cells remained long lived compared with B cells and macrophages could live longer than either of these. Interestingly, when hematopoietic cells were maintained in tissue culture in the presence of interleukin-3, Mcl-1 enhanced the probability of outgrowth of continuously proliferating myeloid cell lines. Thus, Mcl-1 transgenic cells remained subject to normal in vivo homeostatic mechanisms controlling viable cell number, but these constraints could be overridden under specific conditions in vitro. Within the organism, Bcl-2 family members may act at “viability gates” along the differentiation continuum, functioning as part of a system for controlled hematopoietic cell amplification. Enforced expression of even a moderate viability-promoting member of this family such as Mcl-1, within a conducive intra- and extracellular environment in isolation from normal homeostatic constraints, can substantially increase the probability of cell immortalization.
© 1998 by The American Society of Hematology.
Collapse
|
22
|
Mcl-1 in Transgenic Mice Promotes Survival in a Spectrum of Hematopoietic Cell Types and Immortalization in the Myeloid Lineage. Blood 1998. [DOI: 10.1182/blood.v92.9.3226] [Citation(s) in RCA: 118] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Mcl-1 is a member of the Bcl-2 family that is expressed in early monocyte differentiation and that can promote viability on transfection into immature myeloid cells. However, the effects of Mcl-1 are generally short lived compared with those of Bcl-2 and are not obvious in some transfectants. To further explore the effects of this gene, mice were produced that expressed Mcl-1 as a transgene in hematolymphoid tissues. The Mcl-1 transgene was found to cause moderate viability enhancement in a wide range of hematopoietic cell types, including lymphoid (B and T) as well as myeloid cells at both immature and mature stages of differentiation. However, enhanced hematopoietic capacity in transgenic bone marrow and spleen was not reflected in any change in pool sizes in the peripheral blood. In addition, among transgenic cells, mature T cells remained long lived compared with B cells and macrophages could live longer than either of these. Interestingly, when hematopoietic cells were maintained in tissue culture in the presence of interleukin-3, Mcl-1 enhanced the probability of outgrowth of continuously proliferating myeloid cell lines. Thus, Mcl-1 transgenic cells remained subject to normal in vivo homeostatic mechanisms controlling viable cell number, but these constraints could be overridden under specific conditions in vitro. Within the organism, Bcl-2 family members may act at “viability gates” along the differentiation continuum, functioning as part of a system for controlled hematopoietic cell amplification. Enforced expression of even a moderate viability-promoting member of this family such as Mcl-1, within a conducive intra- and extracellular environment in isolation from normal homeostatic constraints, can substantially increase the probability of cell immortalization.
© 1998 by The American Society of Hematology.
Collapse
|
23
|
Proceedings of the British Toxicology Society Annual Congress University of Surrey, Guildford 19 - 22 April 1998. Hum Exp Toxicol 1998. [DOI: 10.1177/096032719801700901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
24
|
Partington GA, Patient RK. Factor binding to the human gamma-globin gene distal CCAAT site: candidates for repression of the normal gene or activation of HPFH mutants. Br J Haematol 1998; 102:940-51. [PMID: 9734644 DOI: 10.1046/j.1365-2141.1998.00849.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We have examined factor binding to the distal human gamma-globin CCAAT site and three naturally occurring hereditary persistence of fetal haemoglobin (HPFH) mutations of this site. Factor binding was examined using nuclear extracts from the erythroleukaemic cell lines K562 and MEL, and from A4 cells, a non-transformed mouse bone marrow stem cell line, using the electrophoretic mobility shift assay. Under standard binding conditions, in addition to the previously reported binding by a CCAAT factor (CP1) and GATA-1, the wild-type (wt) sequence bound high mobility factors which appeared to be GATA-2 isoforms. However, when the non-specific competitor conditions were varied, the binding profile with K562, but not MEL nuclear extract, was substantially altered. CP1 and GATA-1 were absent, and two new factors were detected, one of which bound preferentially to the Greek and Japanese non-deletion HPFH mutants. However, binding by the GATA-2 isoforms to the wt sequence was maintained with both cell types, as it was using the A4 cell line. With modified binding conditions, in A4 cells the two non-deletion and the Black deletion HPFH mutants each had a different protein binding profile which was lost on erythroid induction of the cells. We discuss the possibility that the GATA-2 isoforms bound to the wt sequence may function to suppress wt gamma gene expression in the bone marrow. Additionally, those factors which bind preferentially either to the deletion or non-deletion HPFH mutants may play positive roles in establishing an active chromatin structure.
Collapse
Affiliation(s)
- G A Partington
- Developmental Biology Research Centre, The Randall Institute, Division of Biomedical Sciences, King's College London
| | | |
Collapse
|
25
|
Hu M, Krause D, Greaves M, Sharkis S, Dexter M, Heyworth C, Enver T. Multilineage gene expression precedes commitment in the hemopoietic system. Genes Dev 1997; 11:774-85. [PMID: 9087431 DOI: 10.1101/gad.11.6.774] [Citation(s) in RCA: 596] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We have tested the hypothesis that multipotential hemopoietic stem and progenitor cells prime several different lineage-affiliated programs of gene activity prior to unilineage commitment and differentiation. Using single cell RT-PCR we show that erythroid (beta-globin) and myeloid (myeloperoxidase) gene expression programs can be initiated by the same cell prior to exclusive commitment to the erythroid or granulocytic lineages. Furthermore, the multipotential state is characterized by the coexpression of several lineage-affiliated cytokine receptors. These data support a model of hemopoietic lineage specification in which unilineage commitment is prefaced by a "promiscuous" phase of multilineage locus activation.
Collapse
Affiliation(s)
- M Hu
- The Leukaemia Research Fund Centre at the Institute of Cancer Research, Chester Beatty Laboratories, London, UK
| | | | | | | | | | | | | |
Collapse
|
26
|
Identification of a Serpin Specifically Expressed in Multipotent and Bipotent Hematopoietic Progenitor Cells and in Activated T Cells. Blood 1997. [DOI: 10.1182/blood.v89.1.108] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractWe have identified a gene that has a high level of mRNA expression in undifferentiated, multipotential hematopoietic cells (FDCP-Mix) and that downregulates both transcript and protein, as these cells are induced to differentiate into mature myeloid cells. Sequence analysis of this gene has identified it as a serine protease inhibitor EB22/3 (serpin 2A). Constitutive expression of serpin 2A in FDCP-Mix cells was associated with an increase in the clonogenic potential of the cells and with a delay in the appearance of fully mature cells in cultures undergoing granulocyte macrophage differentiation when compared with control cells. Serpin 2A was also found to be expressed in bone marrow-derived bipotent granulocyte macrophage progenitor cells (GM-colony forming cell [CFC]), but not in erythrocyte progenitor cells from day 15 fetal liver. Expression of serpin 2A also showed a marked up regulation during the activation of cytotoxic suppressor CD8+ T cells, with a clear lag between the appearance of transcript and detection of protein.
Collapse
|
27
|
Identification of a Serpin Specifically Expressed in Multipotent and Bipotent Hematopoietic Progenitor Cells and in Activated T Cells. Blood 1997. [DOI: 10.1182/blood.v89.1.108.108_108_118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We have identified a gene that has a high level of mRNA expression in undifferentiated, multipotential hematopoietic cells (FDCP-Mix) and that downregulates both transcript and protein, as these cells are induced to differentiate into mature myeloid cells. Sequence analysis of this gene has identified it as a serine protease inhibitor EB22/3 (serpin 2A). Constitutive expression of serpin 2A in FDCP-Mix cells was associated with an increase in the clonogenic potential of the cells and with a delay in the appearance of fully mature cells in cultures undergoing granulocyte macrophage differentiation when compared with control cells. Serpin 2A was also found to be expressed in bone marrow-derived bipotent granulocyte macrophage progenitor cells (GM-colony forming cell [CFC]), but not in erythrocyte progenitor cells from day 15 fetal liver. Expression of serpin 2A also showed a marked up regulation during the activation of cytotoxic suppressor CD8+ T cells, with a clear lag between the appearance of transcript and detection of protein.
Collapse
|
28
|
Ford AM, Bennett CA, Healy LE, Towatari M, Greaves MF, Enver T. Regulation of the myeloperoxidase enhancer binding proteins Pu1, C-EBP alpha, -beta, and -delta during granulocyte-lineage specification. Proc Natl Acad Sci U S A 1996; 93:10838-43. [PMID: 8855267 PMCID: PMC38242 DOI: 10.1073/pnas.93.20.10838] [Citation(s) in RCA: 114] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
We have compared the molecular architecture and function of the myeloperoxidase upstream enhancer in multipotential versus granulocyte-committed hematopoietic progenitor cells. We show that the enhancer is accessible in multipotential cell chromatin but functionally incompetent before granulocyte commitment. Multipotential cells contain both Pu1 and C-EBP alpha as enhancer-binding activities. Pu1 is unphosphorylated in both multipotential and granulocyte-committed cells but is phosphorylated in B lymphocytes, raising the possibility that differential phosphorylation may play a role in specifying its lymphoid versus myeloid functions. C-EBP alpha exists as multiple phosphorylated forms in the nucleus of both multipotential and granulocyte-committed cells. C-EBP beta is unphosphorylated and cytoplasmically localized in multipotential cells but exists as a phosphorylated nuclear enhancer-binding activity in granulocyte-committed cells. Granulocyte colony-stimulating factor-induced granulocytic differentiation of multipotential progenitor cells results in activation of C-EBP delta expression and functional recruitment of C-EBP delta and C-EBP beta to the nucleus. Our results implicate Pu1 and the C-EBP family as critical regulators of myeloperoxidase gene expression and are consistent with a model in which a temporal exchange of C-EBP isoforms at the myeloperoxidase enhancer mediates the transition from a primed state in multipotential cells to a transcriptionally active configuration in promyelocytes.
Collapse
Affiliation(s)
- A M Ford
- Leukaemia Research Fund Centre, Chester Beatty Laboratories, London, United Kingdom
| | | | | | | | | | | |
Collapse
|
29
|
Huber MC, Graf T, Sippel AE, Bonifer C. Dynamic changes in the chromatin of the chicken lysozyme gene domain during differentiation of multipotent progenitors to macrophages. DNA Cell Biol 1995; 14:397-402. [PMID: 7748489 DOI: 10.1089/dna.1995.14.397] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The chicken lysozyme locus is regulated in oviduct and macrophages by a complex set of well-characterized cis-regulatory DNA elements. We determined the DNase I hypersensitive chromatin site pattern of the chromatin of the lysozyme locus in retrovirally transformed cell lines representing different stages of myelomonocytic cell differentiation. In the transformed multipotent progenitor stage and in erythroblasts, only a DNase I hypersensitive chromatin site at a silencer element located -2.4 kb upstream of the transcriptional start site is present. At the myeloblast stage DNase I hypersensitive chromatin sites are formed both at the distal enhancer located at -6.1 kb and at the promoter. Later in differentiation, at the monocytic stage, a second DNase I hypersensitive chromatin site appears at the medial enhancer located at -2.7 kb. Parallel with DNase I hypersensitive chromatin site formation at the medial enhancer, the DNase I hypersensitive chromatin site at the silencer element disappears. These chromatin rearrangements correlate with the mRNA expression of the gene that is undetectable in multipotent progenitors and maximal in a lipopolysaccharide-stimulated monocyte cell line. Our results show that the chromatin structure and the transcriptional activity of the gene are tightly coupled during commitment and maturation of the myelomonocytic lineage.
Collapse
Affiliation(s)
- M C Huber
- Institut für Biologie III der Universität Freiburg, Germany
| | | | | | | |
Collapse
|
30
|
Towatari M, May GE, Marais R, Perkins GR, Marshall CJ, Cowley S, Enver T. Regulation of GATA-2 phosphorylation by mitogen-activated protein kinase and interleukin-3. J Biol Chem 1995; 270:4101-7. [PMID: 7876160 DOI: 10.1074/jbc.270.8.4101] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
GATA-2 is a member of a family of transcription factors which bind a common DNA sequence motif (WGA-TAR) through an evolutionarily conserved zinc finger domain. An essential role for GATA-2 in the development of hematopoietic stem cells has recently been shown in gene targeting experiments in mice. Here we show that GATA-2 exists in hematopoietic progenitor cells as a phosphoprotein. Stimulation of progenitors with interleukin-3 (IL-3) results in enhanced phosphorylation of GATA-2 which occurs within 5 min. IL-3 is known to signal in part through mitogen-activated protein (MAP) kinase, and evidence for MAP kinase signaling in the control of GATA-2 phosphorylation was obtained by genetically manipulating the MAP kinase pathway in COS cells using either constitutively activating or interfering mutants of MAP kinase kinase. Furthermore, using an interfering mutant of MAP kinase kinase, we directly demonstrated a critical role for the MAP kinase pathway in the IL-3-dependent phosphorylation of GATA-2 in hematopoietic progenitor cells. Finally, in vitro phosphorylation experiments using recombinant GATA-2 raise the possibility that MAP kinase itself may phosphorylate GATA-2. Our results provide evidence for phosphorylation via the MAP kinase pathway constituting a cytoplasmic link between GATA-2 and growth factor receptors and are consistent with the hypothesis that GATA-2 is involved in the growth factor responsiveness and proliferation control of hematopoietic progenitor cells.
Collapse
Affiliation(s)
- M Towatari
- Leukaemia Research Fund Centre, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
31
|
Just U, Kan O, Fennelly J, Dexter TM, Spooncer E. The Hox-2.4 gene is not involved in the generation of IL-3 dependent multipotent FDCP-mix cell lines. Growth Factors 1995; 12:173-7. [PMID: 8619923 DOI: 10.3109/08977199509036877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The establishment of IL-3-dependent multipotent progenitor cell lines from Hox-2.4-expressing bone marrow cells suggests that homeobox genes may contribute to immortalization of early myeloid cells. A survey of 20 independently derived multipotent IL-3-dependent cell lines established from either src-virus-infected long-term bone marrow cultures (FDCP-mix) or Multi-CSF-virus (M3MuV)-infected bone marrow revealed that Hox-2.4 was not expressed in any of these cell lines. In addition DNA rearrangements were not observed. We conclude that activation of Hox-2.4 is not an obligatory event in the immortalization of early myeloid cells.
Collapse
Affiliation(s)
- U Just
- Department of Experimental Haematology, Paterson Institute for Cancer Research, Manchester, UK
| | | | | | | | | |
Collapse
|
32
|
Ahlers N, Hunt N, Just U, Laker C, Ostertag W, Nowock J. Selectable retrovirus vectors encoding Friend virus gp55 or erythropoietin induce polycythemia with different phenotypic expression and disease progression. J Virol 1994; 68:7235-43. [PMID: 7933106 PMCID: PMC237163 DOI: 10.1128/jvi.68.11.7235-7243.1994] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The Friend spleen focus-forming virus induces a massive expansion of erythroid progenitor cells resulting in polycythemia and splenomegaly. The pathogenic agent is the membrane glycoprotein gp55, encoded by the env gene. Recent evidence indicates that gp55 binds to and activates the erythropoietin (Epo) receptor. It is not clear, however, whether gp55 completely mimics the natural receptor ligand (Epo). To directly compare both effectors, we constructed selectable retroviral vectors which carry either the env or the Epo gene. The selection marker allowed for clonal analysis of infected cells. After infection of DBA/2J mice, the spleen weight, hematological indices, and Epo titer of peripheral blood were monitored. Although both viruses induced an acute erythrocytosis, there were significant differences in disease phenotype and progression. The Epo virus caused an enhanced increase of hematocrit and erythrocytes, whereas with the env virus the pool of late progenitors (CFU-erythroid) was dramatically expanded, resulting in a more severe splenomegaly. The distribution of cytologically recognizable erythroid precursors was shifted towards immature cell types by the env vector compared with Epo. These data suggest that Epo and gp55 differentially affect proliferation and differentiation. Gp55 appears to promote proliferation over differentiation, whereas Epo preferentially drives differentiation.
Collapse
Affiliation(s)
- N Ahlers
- Heinrich-Pette-Institut für Experimentelle Virologie und Immunologie, Universität Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
33
|
Kraut N, Frampton J, McNagny KM, Graf T. A functional Ets DNA-binding domain is required to maintain multipotency of hematopoietic progenitors transformed by Myb-Ets. Genes Dev 1994; 8:33-44. [PMID: 8288126 DOI: 10.1101/gad.8.1.33] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Earlier work demonstrated that the Myb-Ets fusion protein of E26 avian leukemia virus induces the proliferation of multipotent hematopoietic progenitors (MEPs). These progenitors differentiate spontaneously at low frequencies along the erythroid lineage, and following the introduction of kinase/ras-type oncogenes or treatment with TPA, they are induced to differentiate along the myelomonocytic and eosinophilic lineages. Here, we show that the ts1.1 mutant of E26 encodes an Ets DNA-binding domain that is both defective and thermolabile for binding of specific DNA sequences. Correlating with this, ts1.1 MEP colonies transformed at the permissive temperature exhibit elevated levels of erythroid cells and eosinophils, whereas at the nonpermissive temperature they are induced to differentiate along the erythroid and myelomonocytic lineages and, to a lesser extent, along the eosinophil lineage. Induction of the former two lineages cannot be separated by pulse shift experiments and is essentially completed 2.5 days after temperature shift. Our results indicate that the Ets portion of the Myb-Ets fusion protein inhibits the lineage commitment of multipotent hematopoietic progenitors, probably via binding to regulatory DNA sequences of specific target genes.
Collapse
Affiliation(s)
- N Kraut
- Differentiation Programme, European Molecular Biology Laboratory, Heidelberg, Germany
| | | | | | | |
Collapse
|
34
|
Tsai S, Collins SJ. A dominant negative retinoic acid receptor blocks neutrophil differentiation at the promyelocyte stage. Proc Natl Acad Sci U S A 1993; 90:7153-7. [PMID: 8394011 PMCID: PMC47094 DOI: 10.1073/pnas.90.15.7153] [Citation(s) in RCA: 180] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
We have investigated the roles of retinoic acid receptors in the development of neutrophils by using an interleukin 3-dependent multipotent hematopoietic cell line (FDCP mix A4) as well as normal mouse bone marrow cells. Treatment of the FDCP mix A4 cells with murine granulocyte/macrophage-colony-stimulating factor (GM-CSF) induced these cells to differentiate into neutrophils and macrophages. When the endogenous retinoic acid receptor activity in FDCP mix A4 cells was suppressed by a dominant negative retinoic acid receptor construct, this GM-CSF-induced neutrophil differentiation was blocked at the promyelocyte stage. The blocked promyelocytes proliferated continuously as a GM-CSF-dependent cell line but could be induced to terminally differentiate into neutrophils with supraphysiological concentrations of all-trans-retinoic acid (1-10 microM). The ability of the dominant negative retinoic acid receptor to block neutrophil differentiation at the promyelocyte stage was also demonstrated in normal, primary mouse bone marrow cells. Our results indicate that retinoic acid receptors in conjunction with hematopoietic growth factors play a crucial role in the terminal differentiation of normal neutrophil precursors. The system described here may also serve as a model for studying the pathogenesis of human acute promyelocytic leukemia.
Collapse
Affiliation(s)
- S Tsai
- Program in Molecular Medicine, Fred Hutchinson Cancer Research Center, Seattle, WA 98104
| | | |
Collapse
|
35
|
Jiménez G, Griffiths SD, Ford AM, Greaves MF, Enver T. Activation of the beta-globin locus control region precedes commitment to the erythroid lineage. Proc Natl Acad Sci U S A 1992; 89:10618-22. [PMID: 1438257 PMCID: PMC50392 DOI: 10.1073/pnas.89.22.10618] [Citation(s) in RCA: 137] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The beta-globin locus control region (LCR) is characterized by erythroid-specific DNase I hypersensitive sites and is involved in the chromatin organization, transcriptional potentiation, developmental regulation, and replication timing of the entire beta-globin gene cluster. When and how the LCR is first activated during erythropoiesis is not known. Here we analyze the chromatin structure of the LCR during early hematopoietic differentiation using nontransformed, multipotential, growth factor-dependent, murine hematopoietic progenitor cells. We show that LCR hypersensitive sites characteristic of erythroid cells are present in three independent multilineage progenitors [FDCP (factor-dependent cell, Paterson)-mix A4, B6SUtA, and LyD9] under conditions of self-renewal. Induction of differentiation down a nonerythroid pathway causes a progressive loss of hypersensitivity in the LCR. These results show that the beta-globin LCR is in an active chromatin configuration prior to erythroid commitment and indicate a significant role for selective gene repression in lineage specification.
Collapse
Affiliation(s)
- G Jiménez
- Leukaemia Research Fund Centre, Chester Beatty Laboratories, London, United Kingdom
| | | | | | | | | |
Collapse
|
36
|
Graf T, McNagny K, Brady G, Frampton J. Chicken "erythroid" cells transformed by the Gag-Myb-Ets-encoding E26 leukemia virus are multipotent. Cell 1992; 70:201-13. [PMID: 1322247 DOI: 10.1016/0092-8674(92)90096-u] [Citation(s) in RCA: 98] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The E26 avian leukemia virus encodes a transcriptional activator-type oncoprotein consisting of Gag, Myb, and Ets domains, and transforms early erythroid cells as well as myeloblasts. Surprisingly, we have found that "early erythroid" transformants obtained in culture are multipotent, since they can be induced to differentiate into myeloblasts and eosinophils after superinfection with retroviruses containing kinase-type or ras oncogenes. In addition, TPA is an efficient inducer that generates predominantly eosinophils at low concentrations and myeloblasts at high concentrations. The determination process involves the complete extinction of erythroid/thrombocytic markers and the subsequent activation of myelomonocytic/eosinophilic properties, including the acquisition of specific growth factor requirements. "Erythroleukemic" cells from virus-infected animals were likewise found to be multipotent, making this a unique system to study the genesis of stem cell leukemias and the molecular basis of lineage commitment during hematopoiesis.
Collapse
Affiliation(s)
- T Graf
- Differentiation Programme, European Molecular Biology Laboratory, Heidelberg, Germany
| | | | | | | |
Collapse
|
37
|
Ford AM, Bennett CA, Healy LE, Navarro E, Spooncer E, Greaves MF. Immunoglobulin heavy-chain and CD3 delta-chain gene enhancers are DNase I-hypersensitive in hemopoietic progenitor cells. Proc Natl Acad Sci U S A 1992; 89:3424-8. [PMID: 1533043 PMCID: PMC48880 DOI: 10.1073/pnas.89.8.3424] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Multipotential interleukin 3-dependent non-immortalized murine hemopoietic progenitor cells have DNase I-hypersensitive sites in the immunoglobulin heavy-chain and CD3 delta enhancers and transcribe germ-line T-cell antigen receptor gamma-chain (TCR gamma), but not IgM or TCR beta, genes. Induction of myeloid differentiation in these cells clones down expression and/or transcriptional accessibility of the immunoglobulin heavy-chain and TCR gamma genes. The CD3 delta enhancer region remains DNase I-hypersensitive but closes down in B cells. In embryonic stem cells and pan-mesodermal cells, these genes or enhancer regions are neither expressed nor DNase I-hypersensitive. These data suggest that lineage potential may be programmed, at least in part, by alterations in the accessibility or conformation of regulatory regions of genes and that some promiscuity of gene expression and/or accessibility can precede lineage commitment and maturation in progenitor cells induced to self-renew by interleukin 3.
Collapse
Affiliation(s)
- A M Ford
- Leukaemia Research Fund Centre, Chester Beatty Laboratories, London, United Kingdom
| | | | | | | | | | | |
Collapse
|
38
|
Just U, Stocking C, Spooncer E, Dexter TM, Ostertag W. Expression of the GM-CSF gene after retroviral transfer in hematopoietic stem cell lines induces synchronous granulocyte-macrophage differentiation. Cell 1991; 64:1163-73. [PMID: 2004422 DOI: 10.1016/0092-8674(91)90271-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Multipotent murine stem cell lines (FDC-Pmix) depend on IL-3 for self-renewal and proliferation and can be induced to differentiate into multiple hematopoietic lineages. Single FDC-Pmix cells infected with retroviral vectors expressing GM-CSF are induced to differentiate into granulocytes and macrophages. This results in a complete loss of clonogenic cells if IL-3 is not exogenously supplied; however, multipotent variants can be selected that do not terminally differentiate if cells are kept in the presence of IL-3. Unidirectional and synchronous granulocyte and macrophage differentiation accompanied with loss of self-renewal capacity is induced when IL-3 is removed. Our data indicate that activation of the GM-CSF receptor induces differentiation of stem cells by an instructive mechanism that can be blocked by the activated IL-3 receptor. A model of how receptors can induce proliferation and cell-specific differentiation by two separate pathways is discussed.
Collapse
Affiliation(s)
- U Just
- Heinrich-Pette-Institut für Experimentelle Virologie und Immunologie, Universität Hamburg, Federal Republic of Germany
| | | | | | | | | |
Collapse
|
39
|
Bahnson AB, Boggs SS. Addition of serum to electroporated cells enhances survival and transfection efficiency. Biochem Biophys Res Commun 1990; 171:752-7. [PMID: 2403361 DOI: 10.1016/0006-291x(90)91210-j] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Optimal electroporation efficiency of many cell types is associated with poor survival. We show that serum rapidly reseals the membranes of electroporated cells and that timely addition of serum following electroporation can improve cell survival and transfection efficiency.
Collapse
Affiliation(s)
- A B Bahnson
- Department of Radiation Oncology, University of Pittsburgh School of Medicine, PA 15261
| | | |
Collapse
|
40
|
Abstract
Fresh bone marrow (BM)-derived cells infected with the J2 recombinant retrovirus (carrying v-myc and v-raf/mil oncogenes) grow as immortal cell lines belonging to the monocytic lineage. BM cells cultured for 24 h in conventional medium are no longer able to grow following infection with the J2 virus. We investigated whether specific growth factors affected the proliferative response of BM cells to the J2 virus. If the BM cells were cultured for 24 h in the presence of concanavalin A or CSF-1 and then infected with the J2 virus, immortalization of BM cells was observed. Under these conditions, the cell lines that we obtained were shown to belong to the monocytic lineage. We investigated whether target cells for the J2 virus existed in other hematopoietic organs. We observed J2-induced proliferation in fetal liver (FL) but not in spleen or thymus. The cells proliferating in the FL had macrophage characteristics during the early passages. However, some macrophage markers were lost upon extensive in vitro culture. We conclude that we have identified conditions in which J2 virus consistently and selectively stimulates the growth of macrophages from murine bone marrow and a wider range of hematopoietic cells from fetal liver.
Collapse
Affiliation(s)
- L Gandino
- Dipartimento di Scienze Biomediche ed Oncologia Umana, Università di Torino, Italy
| | | |
Collapse
|
41
|
Keller G, Wagner EF. Expression of v-src induces a myeloproliferative disease in bone-marrow-reconstituted mice. Genes Dev 1989; 3:827-37. [PMID: 2568314 DOI: 10.1101/gad.3.6.827] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A recombinant retrovirus, N-TK-src, was used to introduce the v-src oncogene into mouse hematopoietic cells. This vector efficiently expresses both the neo and v-src genes in different hematopoietic lineages in culture as well as in mice reconstituted with infected bone marrow cells. Expression of v-src had no dramatic effect on the proliferative and differentiative capacity of hematopoietic precursors when assayed in methyl cellulose cultures. However, in mice reconstituted with N-TK-src-infected bone marrow cells, expression of v-src leads to the rapid development of a severe myeloproliferative disease, characterized by splenomegaly, anemia, and a shift of hematopoiesis from the bone marrow to the spleen.
Collapse
Affiliation(s)
- G Keller
- Basel Institute for Immunology, Switzerland
| | | |
Collapse
|
42
|
Morris AJ, Dexter TM, Gallagher JT. Metabolic properties of a homogeneous proteoglycan of a haemopoietic stem cell line, FDCP-mix. Biochem J 1989; 260:479-86. [PMID: 2504144 PMCID: PMC1138693 DOI: 10.1042/bj2600479] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A biochemical analysis has been carried out of metabolically labelled proteoglycans and glycosaminoglycans synthesized by a haemopoietic multipotential stem cell line, FDCP-mix. The only proteoglycan identified in these multipotential cells was a homogeneous component that contained chondroitin 4-sulphate chains (Mr approximately 10,000) arranged in close proximity in a proteinase-resistant domain of the protein core. Small quantities of free chondroitin 4-sulphate were also detected. Following a 48 h incubation with Na2 35SO4 the majority of the 35S-radiolabelled proteoglycans (approximately 80%) were associated with the cells, mainly in an intracellular compartment, and the remaining 20% were in the culture medium. Pulse-chase studies demonstrated two turnover pathways for the newly synthesized cellular proteoglycans. In the minor pathway, the proteoglycans were secreted rapidly into the medium without any discernable structural modification. In the major pathway the proteoglycans seemed to be transferred into a storage compartment from which the intact macromolecules were not secreted. Eventually, these proteoglycans were degraded to yield free polysaccharide chains and these chains were then released into the medium, but only at a relatively slow rate. There was very little intracellular degradation of chondroitin sulphate chains. The pathway to polysaccharide secretion was a slow stepwise process with a time-lag of about 5 h between proteoglycan synthesis and the appearance of free chondroitin sulphate and a second time-lag, also of about 5 h, before these chains began to be secreted. The existence of separate secretory pathways for proteoglycans and chondroitin sulphate chains is an interesting characteristic that seems to distinguish proteoglycan metabolism in primitive multipotent stem cells from related metabolic processes in mature haemopoietic cells.
Collapse
Affiliation(s)
- A J Morris
- Department of Medical Oncology, Paterson Institute for Cancer Research, Christie Hospital, Manchester, U.K
| | | | | |
Collapse
|
43
|
Spooncer E, Katsuno M, Hampson I, Dexter TM, Just U, Stocking C, Kluge N, Ostertag W. Biological effects of retroviral transfection of the murine interleukin-3 gene into FDCP-Mix cells. Curr Top Microbiol Immunol 1989; 149:109-16. [PMID: 2786463 DOI: 10.1007/978-3-642-74623-9_10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
44
|
Ford AM, Healy LE, Watt SM, Spooncer E, Dexter TM, Molgaard HV, Greaves MF. Cell lineage specificity of chromatin configuration and gene expression in haemopoietic progenitor cells. HAEMATOLOGY AND BLOOD TRANSFUSION 1989; 32:352-6. [PMID: 2516497 DOI: 10.1007/978-3-642-74621-5_61] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- A M Ford
- Leukaemia Research Fund Centre, Institute of Cancer Research, Chester Beatty Laboratories, London, UK
| | | | | | | | | | | | | |
Collapse
|
45
|
Palmieri S. Oncogene requirements for tumorigenicity: cooperative effects between retroviral oncogenes. Curr Top Microbiol Immunol 1989; 148:43-91. [PMID: 2684549 DOI: 10.1007/978-3-642-74700-7_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
46
|
|
47
|
Daniel CP, Ponting IL, Dexter TM. Growth and development of haemopoietic cells: a deterministic process? HAEMATOLOGY AND BLOOD TRANSFUSION 1989; 32:172-7. [PMID: 2696678 DOI: 10.1007/978-3-642-74621-5_29] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The in vitro methods used to study haemopoiesis fall into two distinct categories. Short-term colony forming assays have identified a number of potent soluble factors capable of maintaining survival, proliferation and differentiation of haemopoietic cells but not their self-renewal. In contrast, in long-term bone marrow culture, extensive self-renewal occurs in the absence of exogenous factors and direct physical contact between haemopoietic cells and cells of the adherent stromal layer seems to be important. Obviously, LTBMC more closely resembles the situation in haemopoietic tissues but the potency of growth factors imply that they too play a role. Our data suggest that this may be at an earlier stage of haemopoietic development than previously appreciated. Primitive multipotent cells have the potential to respond to CSFs which were previously thought to stimulate only committed progenitor cells. This response is only seen, however, when the cells are exposed to a combination of factors which include either IL-1 or G-CSF. Thus, a combination of factors is able to recruit cells which are not already committed and determine the lineage along which they will differentiate. While it remains to be conclusively demonstrated that growth factors regulate normal "steady state" haemopoiesis in vivo it is clear that contact with stromal cells is important. The mechanisms by which the adherent layer influences haemopoietic development, however, are less obvious. We have shown that a component of stroma, heparan sulphate, is able to bind growth factors and present them to haemopoietic cells in a way that stimulates haemopoiesis.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- C P Daniel
- Department of Experimental Haematology, Paterson Institute for Cancer Research, Manchester, UK
| | | | | |
Collapse
|
48
|
Peppoloni S, Blasi E, Ortaldo JR, Rapp UR, Riccardi C, Varesio L. In vitro proliferation of human large granular lymphocytes with v-raf/v-myc recombinant retrovirus. EXPERIENTIA 1988; 44:1013-5. [PMID: 2461868 DOI: 10.1007/bf01939906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The effect of infection with a retrovirus carrying v-raf/v-myc oncogenes (J2 virus) on the in vitro proliferation of human large granular lymphocytes (LGL) was investigated. LGL infected with J2 virus (J2LGL), unlike uninfected cells, grew with a proliferation peak eight days after infection. Such cells retained the morphology and functional properties typical of LGL. Furthermore, 5% of J2LGL produced virus the day after infection, whereas non-virus production was detectable five days later. These data indicate that J2 virus provides a transient mitogenic signal for LGL.
Collapse
Affiliation(s)
- S Peppoloni
- Biological Response Modifiers Program, NCI-FCRF, Frederick, Maryland
| | | | | | | | | | | |
Collapse
|
49
|
Preisler HD. A hypothesis regarding the development of acute myeloid leukemia from preleukemic disorders. The role of protooncogenes. CANCER GENETICS AND CYTOGENETICS 1988; 32:133-42. [PMID: 3162703 DOI: 10.1016/0165-4608(88)90320-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Acute nonlymphocytic leukemia is often preceded by a preleukemic phase that can be characterized by a reduction in hematopoiesis, by ineffective hematopoiesis, or by a myeloproliferative/myelodysplastic state. The time between onset of the preleukemic state and the appearance of leukemia is quite variable, and at times such evolution does not occur. These clinical observations are compatible with the multistep hypothesis of tumor development. In studies underway in our laboratory, chronic myelogenous leukemia (CML) is being used as a model system to study the steps in and possible mechanisms underlying the development of preleukemia and its evolution to acute leukemia. Chronic myelogenous leukemia is particularly suited for this role because the chronic phase of the disease is an easily identifiable myeloproliferative state that invariably evolves into acute leukemia. In the discussion that follows, this clinical entity is used to develop a general model for the preleukemias and their evolution to acute leukemia.
Collapse
Affiliation(s)
- H D Preisler
- Department of Hematologic Oncology, Roswell Park Memorial Institute, Buffalo, NY 14263
| |
Collapse
|
50
|
Harel-Bellan A, Farrar WL. Modulation of proto-oncogene expression by colony stimulating factors. Biochem Biophys Res Commun 1987; 148:1001-8. [PMID: 2446603 DOI: 10.1016/s0006-291x(87)80231-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Bone marrow stem cells are dependent upon Colony Stimulating Factors for proliferation in vitro. Using murine myeloid cell lines, which are able to proliferate in culture in the presence of three different murine Colony Stimulating Factors, we have investigated the effect of Colony Stimulating Factors on c-fos and c-myc mRNA induction. Cells were blocked in G1 phase by Colony Stimulating Factor and serum deprivation and allowed to proceed into the cell cycle by addition of Colony Stimulating Factor. Northern analysis of RNA showed that c-fos and c-myc message were induced by the three different Colony Stimulating Factors tested, Interleukin 3, Granulocyte Monocyte-Colony Stimulating Factor and Granulocyte-Colony Stimulating Factor, with similar kinetics and levels of induction. Induction of c-fos mRNA was rapid, transient (with a peak 30 min after induction) and very faint. Accumulation of c-myc (15- to 30-fold) was more stable and quantitatively more important. These results suggests that c-fos and c-myc gene activation is a "consensus" and obligatory event in cell proliferation induction.
Collapse
Affiliation(s)
- A Harel-Bellan
- Laboratory of Molecular Immunoregulation, National Cancer Institute, Frederick, MD 21701
| | | |
Collapse
|