Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Nat Immunol ; 23(3): 458-468, 2022 03.
Article in English | MEDLINE | ID: mdl-35210623

ABSTRACT

Alveolar macrophages (AMs) are lung tissue-resident macrophages that can be expanded in culture, but it is unknown to what extent culture affects their in vivo identity. Here we show that mouse long-term ex vivo expanded AMs (exAMs) maintained a core AM gene expression program, but showed culture adaptations related to adhesion, metabolism and proliferation. Upon transplantation into the lung, exAMs reacquired full transcriptional and epigenetic AM identity, even after several months in culture and could self-maintain long-term in the alveolar niche. Changes in open chromatin regions observed in culture were fully reversible in transplanted exAMs and resulted in a gene expression profile indistinguishable from resident AMs. Our results indicate that long-term proliferation of AMs in culture did not compromise cellular identity in vivo. The robustness of exAM identity provides new opportunities for mechanistic analysis and highlights the therapeutic potential of exAMs.


Subject(s)
Lung , Macrophages, Alveolar , Animals , Chromatin/metabolism , Epigenesis, Genetic , Epigenomics , Lung/metabolism , Macrophages, Alveolar/metabolism , Mice
2.
Cell ; 167(5): 1264-1280.e18, 2016 11 17.
Article in English | MEDLINE | ID: mdl-28084216

ABSTRACT

Granulomas are immune cell aggregates formed in response to persistent inflammatory stimuli. Granuloma macrophage subsets are diverse and carry varying copy numbers of their genomic information. The molecular programs that control the differentiation of such macrophage populations in response to a chronic stimulus, though critical for disease outcome, have not been defined. Here, we delineate a macrophage differentiation pathway by which a persistent Toll-like receptor (TLR) 2 signal instructs polyploid macrophage fate by inducing replication stress and activating the DNA damage response. Polyploid granuloma-resident macrophages formed via modified cell divisions and mitotic defects and not, as previously thought, by cell-to-cell fusion. TLR2 signaling promoted macrophage polyploidy and suppressed genomic instability by regulating Myc and ATR. We propose that, in the presence of persistent inflammatory stimuli, pathways previously linked to oncogene-initiated carcinogenesis instruct a long-lived granuloma-resident macrophage differentiation program that regulates granulomatous tissue remodeling.


Subject(s)
DNA Damage , Granuloma/immunology , Macrophages/immunology , Mycobacterium tuberculosis/immunology , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Differentiation , Cell Proliferation , Humans , Inflammation/immunology , Lipoproteins/immunology , Mice , Mice, Inbred C57BL , Mitosis , Proto-Oncogene Proteins c-myc/metabolism , Toll-Like Receptor 2
4.
Cell ; 138(2): 300-13, 2009 Jul 23.
Article in English | MEDLINE | ID: mdl-19632180

ABSTRACT

While hematopoietic stem cell (HSC) self-renewal is well studied, it remains unknown whether distinct control mechanisms enable HSC divisions that generate progeny cells with specific lineage bias. Here, we report that the monocytic transcription factor MafB specifically restricts the ability of M-CSF to instruct myeloid commitment divisions in HSCs. MafB deficiency specifically enhanced sensitivity to M-CSF and caused activation of the myeloid master-regulator PU.1 in HSCs in vivo. Single-cell analysis revealed that reduced MafB levels enabled M-CSF to instruct divisions producing asymmetric daughter pairs with one PU.1(+) cell. As a consequence, MafB(-/-) HSCs showed a PU.1 and M-CSF receptor-dependent competitive repopulation advantage specifically in the myelomonocytic, but not T lymphoid or erythroid, compartment. Lineage-biased repopulation advantage was progressive, maintained long term, and serially transplantable. Together, this indicates that an integrated transcription factor/cytokine circuit can control the rate of specific HSC commitment divisions without compromising other lineages or self-renewal.


Subject(s)
Cell Lineage , Hematopoietic Stem Cells/cytology , Macrophage Colony-Stimulating Factor/metabolism , MafB Transcription Factor/metabolism , Myeloid Cells/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins/metabolism , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Trans-Activators/metabolism
5.
Nat Immunol ; 17(6): 609-11, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27196510

Subject(s)
Eosinophils , Mast Cells , Humans
6.
Nature ; 497(7448): 239-43, 2013 May 09.
Article in English | MEDLINE | ID: mdl-23575636

ABSTRACT

Under stress conditions such as infection or inflammation the body rapidly needs to generate new blood cells that are adapted to the challenge. Haematopoietic cytokines are known to increase output of specific mature cells by affecting survival, expansion and differentiation of lineage-committed progenitors, but it has been debated whether long-term haematopoietic stem cells (HSCs) are susceptible to direct lineage-specifying effects of cytokines. Although genetic changes in transcription factor balance can sensitize HSCs to cytokine instruction, the initiation of HSC commitment is generally thought to be triggered by stochastic fluctuation in cell-intrinsic regulators such as lineage-specific transcription factors, leaving cytokines to ensure survival and proliferation of the progeny cells. Here we show that macrophage colony-stimulating factor (M-CSF, also called CSF1), a myeloid cytokine released during infection and inflammation, can directly induce the myeloid master regulator PU.1 and instruct myeloid cell-fate change in mouse HSCs, independently of selective survival or proliferation. Video imaging and single-cell gene expression analysis revealed that stimulation of highly purified HSCs with M-CSF in culture resulted in activation of the PU.1 promoter and an increased number of PU.1(+) cells with myeloid gene signature and differentiation potential. In vivo, high systemic levels of M-CSF directly stimulated M-CSF-receptor-dependent activation of endogenous PU.1 protein in single HSCs and induced a PU.1-dependent myeloid differentiation preference. Our data demonstrate that lineage-specific cytokines can act directly on HSCs in vitro and in vivo to instruct a change of cell identity. This fundamentally changes the current view of how HSCs respond to environmental challenge and implicates stress-induced cytokines as direct instructors of HSC fate.


Subject(s)
Cell Differentiation/drug effects , Cell Lineage/drug effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Macrophage Colony-Stimulating Factor/pharmacology , Myeloid Cells/cytology , Myeloid Cells/drug effects , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Single-Cell Analysis , Trans-Activators/biosynthesis , Trans-Activators/genetics , Trans-Activators/metabolism
7.
Semin Immunol ; 23(5): 326-34, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21937237

ABSTRACT

In the predominant model of hematopoietic stem cell differentiation lineage commitment is thought to be initiated by stochastic variation in the balance of lineage determining transcription factors, whereas cytokines have been seen in a purely permissive role of stimulating selective survival and proliferation of the down stream progeny. Recent observations, however, indicate that cytokines can also directly instruct cell fate change in uncommitted stem and progenitor cells by activating lineage determining transcription factors. We review the historic and recent evidence for instructive cytokine signaling and propose a model that integrates cytokine signaling and transcription factor activity in the initial decision making process, where the sensitivity to external instructive signals can be modulated by internal threshold setters. In contrast to a rigid stochastic explanation of lineage commitment this view allows for receptiveness of the hematopoietic stem cell to its environment and exposes lineage commitment as dependent on both instructive signals and cell intrinsically controlled sensitivity to external cues.


Subject(s)
Cytokines , Hematopoietic Stem Cells , Models, Biological , Transcription Factors , Cell Differentiation , Cell Lineage , Hematopoietic Stem Cells/cytology , Humans
8.
J Exp Med ; 220(7)2023 07 03.
Article in English | MEDLINE | ID: mdl-37067792

ABSTRACT

So far, hematopoietic stem cells (HSC) are considered the source of mature immune cells, the latter being the only ones capable of mounting an immune response. Recent evidence shows HSC can also directly sense cytokines released upon infection/inflammation and pathogen-associated molecular pattern interaction while keeping a long-term memory of previously encountered signals. Direct sensing of danger signals by HSC induces early myeloid commitment, increases myeloid effector cell numbers, and contributes to an efficient immune response. Here, by using specific genetic tools on both the host and pathogen sides, we show that HSC can directly sense B. abortus pathogenic bacteria within the bone marrow via the interaction of the cell surface protein CD150 with the bacterial outer membrane protein Omp25, inducing efficient functional commitment of HSC to the myeloid lineage. This is the first demonstration of direct recognition of a live pathogen by HSC via CD150, which attests to a very early contribution of HSC to immune response.


Subject(s)
Brucella , Hematopoietic Stem Cells/metabolism , Bone Marrow , Bone Marrow Cells , Membrane Proteins/metabolism , Cell Differentiation
9.
EMBO Mol Med ; 15(11): e17694, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37635627

ABSTRACT

Therapies reconstituting autologous antiviral immunocompetence may represent an important prophylaxis and treatment for immunosuppressed individuals. Following hematopoietic cell transplantation (HCT), patients are susceptible to Herpesviridae including cytomegalovirus (CMV). We show in a murine model of HCT that macrophage colony-stimulating factor (M-CSF) promoted rapid antiviral activity and protection from viremia caused by murine CMV. M-CSF given at transplantation stimulated sequential myeloid and natural killer (NK) cell differentiation culminating in increased NK cell numbers, production of granzyme B and interferon-γ. This depended upon M-CSF-induced myelopoiesis leading to IL15Rα-mediated presentation of IL-15 on monocytes, augmented by type I interferons from plasmacytoid dendritic cells. Demonstrating relevance to human HCT, M-CSF induced myelomonocytic IL15Rα expression and numbers of functional NK cells in G-CSF-mobilized hematopoietic stem and progenitor cells. Together, M-CSF-induced myelopoiesis triggered an integrated differentiation of myeloid and NK cells to protect HCT recipients from CMV. Thus, our results identify a rationale for the therapeutic use of M-CSF to rapidly reconstitute antiviral activity in immunocompromised individuals, which may provide a general paradigm to boost innate antiviral immunocompetence using host-directed therapies.


Subject(s)
Cytomegalovirus Infections , Hematopoietic Stem Cell Transplantation , Humans , Mice , Animals , Cytomegalovirus , Macrophage Colony-Stimulating Factor , Hematopoietic Stem Cell Transplantation/methods , Cytomegalovirus Infections/prevention & control , Hematopoiesis , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cell Differentiation
10.
Mol Cell Biol ; 27(15): 5554-64, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17548468

ABSTRACT

During the execution of differentiation programs, lineage-specific transcription factors are in competition with antagonistic factors that drive progenitor proliferation. Thus, the myeloid transcription factor MafB promotes macrophage differentiation of myeloid progenitors, but a constitutively active Myb transcription factor (v-Myb) can maintain proliferation and block differentiation. Little is known, however, about the regulatory mechanisms that control such competing activities. Here we report that the small ubiquitin-like protein SUMO-1 can modify MafB in vitro and in vivo on lysines 32 and 297. The absence of MafB SUMO modification increased MafB-driven transactivation and macrophage differentiation potential but inhibited cell cycle progression and myeloid progenitor growth. Furthermore, we observed that direct repression of MafB transactivation by v-Myb was strictly dependent on MafB SUMO modification. Consequently, a SUMOylation-deficient MafB K32R K297R (K32,297R) mutant could specify macrophage fate even after activation of inducible Myb alleles and resist their differentiation-inhibiting activity. Our findings suggest that SUMO modification of MafB affects the balance between myeloid progenitor expansion and terminal macrophage differentiation by controlling MafB transactivation capacity and susceptibility to Myb repression. SUMO modification of lineage-specific transcription factors may thus modulate transcription factor antagonism to control tissue homeostasis in the hematopoietic system.


Subject(s)
Cell Differentiation , Macrophages/cytology , MafB Transcription Factor/metabolism , Oncogene Proteins v-myb/metabolism , Repressor Proteins/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcription, Genetic , Animals , Cell Line , Cell Proliferation , Chickens , Humans , Mice , Models, Biological , Myeloid Cells/cytology , Protein Binding , Stem Cells/cytology , Transcriptional Activation/genetics
11.
Cell Stem Cell ; 26(5): 657-674.e8, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32169166

ABSTRACT

Hematopoietic stem cells (HSCs) maintain life-long production of immune cells and can directly respond to infection, but sustained effects on the immune response remain unclear. We show that acute immune stimulation with lipopolysaccharide (LPS) induced only transient changes in HSC abundance, composition, progeny, and gene expression, but persistent alterations in accessibility of specific myeloid lineage enhancers occurred, which increased responsiveness of associated immune genes to secondary stimulation. Functionally, this was associated with increased myelopoiesis of pre-exposed HSCs and improved innate immunity against the gram-negative bacterium P. aeruginosa. The accessible myeloid enhancers were enriched for C/EBPß targets, and C/EBPß deletion erased the long-term inscription of LPS-induced epigenetic marks and gene expression. Thus, short-term immune signaling can induce C/EBPß-dependent chromatin accessibility, resulting in HSC-trained immunity, during secondary infection. This establishes a mechanism for how infection history can be epigenetically inscribed in HSCs as an integral memory function of innate immunity.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta , Epigenesis, Genetic , Hematopoietic Stem Cells/immunology , Immunity, Innate , CCAAT-Enhancer-Binding Protein-beta/genetics , Epigenomics , Humans , Myelopoiesis
12.
Mol Cell Biol ; 26(18): 6808-18, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16943423

ABSTRACT

In the hematopoietic system the bZip transcription factor MafB is selectively expressed at high levels in monocytes and macrophages and promotes macrophage differentiation in myeloid progenitors, whereas a dominant-negative allele can inhibit this process. To analyze the requirement of MafB for macrophage development, we generated MafB-deficient mice and, due to their neonatal lethal phenotype, analyzed macrophage differentiation in vitro, in the embryo, and in reconstituted mice. Surprisingly we observed in vitro differentiation of macrophages from E14.5 fetal liver (FL) cells and E18.5 splenocytes. Furthermore we found normal numbers of F4/80(+)/Mac-1(+) macrophages and monocytes in fetal liver, spleen, and blood as well as in bone marrow, spleen, and peritoneum of adult MafB(-/-) FL reconstituted mice. MafB(-/-) macrophages showed intact basic macrophage functions such as phagocytosis of latex beads or Listeria monocytogenes and nitric oxide production in response to lipopolysaccharide. By contrast, MafB(-/-) macrophages expressed increased levels of multiple genes involved in actin organization. Consistent with this, phalloidin staining revealed an altered morphology involving increased numbers of branched protrusions of MafB(-/-) macrophages in response to macrophage colony-stimulating factor. Together these data point to an unexpected redundancy of MafB function in macrophage differentiation and a previously unknown role in actin-dependent macrophage morphology.


Subject(s)
Actins/metabolism , Macrophages/cytology , MafB Transcription Factor/deficiency , Animals , Animals, Newborn , Cell Differentiation , Embryo, Mammalian/cytology , Fetus/cytology , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation , Hematopoietic System/cytology , Liver/cytology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-maf/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spleen/cytology , Whole-Body Irradiation
15.
Mol Cell Biol ; 23(4): 1390-402, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12556498

ABSTRACT

FLI-1 is an ETS family transcription factor which is overexpressed in Friend erythroleukemia and contributes to the blockage of differentiation of erythroleukemic cells. We show here that FLI-1 represses the transcriptional activity of the beta-globin gene promoter in MEL cells and interacts with two of its critical transactivators, GATA-1 and EKLF. Unexpectedly, FLI-1 enhances the stimulating activity of GATA-1 on a GATA-1-responsive promoter but represses that of EKLF on beta-globin and an EKLF-responsive artificial promoters. This repressive effect of FLI-1 requires the ETS DNA binding domain and its association with either the N- or C-terminal domain, which themselves interact with EKLF but not with GATA-1. Furthermore, the FLI-1 ETS domain alone behaves as an autonomous repression domain when linked to the Gal4 DNA binding domain. Taken together, these data indicate that FLI-1 represses EKLF-dependent transcription due to the repression activity of its ETS domain and its indirect recruitment to erythroid promoters by protein-protein interaction with EKLF. Reciprocally, we also show that EKLF itself represses the FLI-1-dependent megakaryocytic GPIX gene promoter, thus further suggesting that functional cross-antagonism between FLI-1 and EKLF might be involved in the control of the erythrocytic versus megakaryocytic differentiation of bipotential progenitors.


Subject(s)
DNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Acetamides/pharmacology , Animals , Base Sequence , Cell Differentiation/physiology , Cells, Cultured , DNA/metabolism , DNA-Binding Proteins/genetics , Erythrocytes/cytology , Erythrocytes/physiology , Erythroid-Specific DNA-Binding Factors , GATA1 Transcription Factor , Globins/drug effects , Globins/genetics , Kruppel-Like Transcription Factors , Mice , Molecular Sequence Data , Platelet Glycoprotein GPIb-IX Complex/genetics , Promoter Regions, Genetic , Protein Structure, Tertiary , Proto-Oncogene Protein c-fli-1 , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Trans-Activators/genetics , Transcription Factors/genetics , Transcription, Genetic
16.
J Exp Med ; 213(11): 2269-2279, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27811055

ABSTRACT

Myeloablative treatment preceding hematopoietic stem cell (HSC) and progenitor cell (HS/PC) transplantation results in severe myeloid cytopenia and susceptibility to infections in the lag period before hematopoietic recovery. We have previously shown that macrophage colony-stimulating factor (CSF-1; M-CSF) directly instructed myeloid commitment in HSCs. In this study, we tested whether this effect had therapeutic benefit in improving protection against pathogens after HS/PC transplantation. M-CSF treatment resulted in an increased production of mature myeloid donor cells and an increased survival of recipient mice infected with lethal doses of clinically relevant opportunistic pathogens, namely the bacteria Pseudomonas aeruginosa and the fungus Aspergillus fumigatus M-CSF treatment during engraftment or after infection efficiently protected from these pathogens as early as 3 days after transplantation and was effective as a single dose. It was more efficient than granulocyte CSF (G-CSF), a common treatment of severe neutropenia, which showed no protective effect under the tested conditions. M-CSF treatment showed no adverse effect on long-term lineage contribution or stem cell activity and, unlike G-CSF, did not impede recovery of HS/PCs, thrombocyte numbers, or glucose metabolism. These results encourage potential clinical applications of M-CSF to prevent severe infections after HS/PC transplantation.


Subject(s)
Aspergillosis/drug therapy , Aspergillosis/prevention & control , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Macrophage Colony-Stimulating Factor/therapeutic use , Pseudomonas Infections/drug therapy , Pseudomonas Infections/prevention & control , Animals , Aspergillosis/blood , Aspergillosis/microbiology , Aspergillus/drug effects , Aspergillus/physiology , Blood Glucose/metabolism , Blood Platelets/drug effects , Blood Platelets/metabolism , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Self Renewal/drug effects , Hematopoietic Stem Cells/drug effects , Humans , Macrophage Colony-Stimulating Factor/pharmacology , Mice, Inbred C57BL , Myelopoiesis/drug effects , Pseudomonas Infections/blood , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology
17.
Science ; 353(6301): aad8670, 2016 Aug 19.
Article in English | MEDLINE | ID: mdl-27338705

ABSTRACT

Microglia, the resident myeloid cells of the central nervous system, play important roles in life-long brain maintenance and in pathology. Despite their importance, their regulatory dynamics during brain development have not been fully elucidated. Using genome-wide chromatin and expression profiling coupled with single-cell transcriptomic analysis throughout development, we found that microglia undergo three temporal stages of development in synchrony with the brain--early, pre-, and adult microglia--which are under distinct regulatory circuits. Knockout of the gene encoding the adult microglia transcription factor MAFB and environmental perturbations, such as those affecting the microbiome or prenatal immune activation, led to disruption of developmental genes and immune response pathways. Together, our work identifies a stepwise microglia developmental program integrating immune response pathways that may be associated with several neurodevelopmental disorders.


Subject(s)
Brain/embryology , Homeostasis/physiology , Microglia/cytology , Neurogenesis/immunology , Animals , Blood-Brain Barrier/embryology , Blood-Brain Barrier/immunology , Brain/immunology , Chromatin/metabolism , Epigenesis, Genetic , Female , Gene Expression Profiling , Gene Knockout Techniques , Histone Code , Homeostasis/genetics , Immunity/genetics , MafB Transcription Factor/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Myeloid Cells/cytology , Neurogenesis/genetics , Single-Cell Analysis
18.
Biochim Biophys Acta ; 1592(2): 123-7, 2002 Oct 21.
Article in English | MEDLINE | ID: mdl-12379474

ABSTRACT

Programmed cell death (apoptosis) is a complex phenomenon that is mediated in mammals mainly via the selective cleavage of intracellular proteins by the large family of cysteine aspartate protease caspases. Apoptosis is tightly regulated by the competitive effect of numerous proteins displaying either pro-apoptotic or anti-apoptotic activity. The ETS-family transcription factor FLI-1, frequently associated with malignant transformation, has been shown to display anti-apoptotic activity in several cell types including avian erythroblasts, mouse fibroblasts or lymphoid cells. We show here that apoptosis of murine preB leukemic cells is accompanied with the specific cleavage of FLI-1 by a caspase-like activity. We also demonstrate that the two isoforms of FLI-1 are indeed cleaved at three conserved sites by caspase 3 in vitro. The conservation of these cleavage sites among species suggests that the caspase cleavage of the anti-apoptotic transcription factor FLI-1 may represent a critical step to ensure irreversible cell death.


Subject(s)
Apoptosis , Caspases/metabolism , DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins , Trans-Activators/metabolism , Animals , Binding Sites , Blotting, Western , Caspase 3 , Electrophoresis, Polyacrylamide Gel , Mice , Molecular Weight , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Proto-Oncogene Protein c-fli-1 , Tumor Cells, Cultured
20.
Science ; 326(5954): 867-71, 2009 Nov 06.
Article in English | MEDLINE | ID: mdl-19892988

ABSTRACT

In metazoan organisms, terminal differentiation is generally tightly linked to cell cycle exit, whereas the undifferentiated state of pluripotent stem cells is associated with unlimited self-renewal. Here, we report that combined deficiency for the transcription factors MafB and c-Maf enables extended expansion of mature monocytes and macrophages in culture without loss of differentiated phenotype and function. Upon transplantation, the expanded cells are nontumorigenic and contribute to functional macrophage populations in vivo. Small hairpin RNA inactivation shows that continuous proliferation of MafB/c-Maf deficient macrophages requires concomitant up-regulation of two pluripotent stem cell-inducing factors, KLF4 and c-Myc. Our results indicate that MafB/c-MafB deficiency renders self-renewal compatible with terminal differentiation. It thus appears possible to amplify functional differentiated cells without malignant transformation or stem cell intermediates.


Subject(s)
Cell Proliferation , Macrophages/physiology , MafB Transcription Factor/deficiency , Proto-Oncogene Proteins c-maf/deficiency , Animals , Cell Differentiation , Cell Transformation, Neoplastic , Cells, Cultured , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/physiology , Macrophage Colony-Stimulating Factor/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/cytology , Macrophages/transplantation , MafB Transcription Factor/genetics , MafB Transcription Factor/physiology , Mice , Mice, Knockout , Monocytes/cytology , Monocytes/physiology , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/physiology , Phagocytosis , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/physiology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/physiology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL