The proto-oncogene c-fos was transiently upregulated in primitive hematopoietic stem (LinSca-1+) cells stimulated with stem cell factor, interleukin-3 (IL-3), and IL-6. To investigate a role of the c-fos in hematopoietic stem cells, we used bone marrow (BM) cells from transgenic mice carrying the c-fos gene under the control of the interferon-/β–inducible Mx-promoter (Mx–c-fos), and fetal liver cells from c-fos–deficient mice. Prolonged expression of the c-fos in LinSca-1+ BM cells inhibited factor-dependent colony formation and hematopoiesis on a stromal cell layer by keeping them at G0/G1 phase of the cell cycle. These LinSca-1+ BM cells on a stromal layer entered into the cell cycle whenever exogenous c-fos was downregulated. However, ectopic c-fos did not perturb colony formation by LinSca-1+ BM cells after they entered the cell cycle. Furthermore, endogenous c-fos is not essential to cell cycle progression of hematopoietic stem cells because the factor-dependent and the stroma-dependent hematopoiesis by LinSca-1+ fetal liver cells from c-fos–deficient mice was not impaired. These results suggest that the c-fos induced in primitive hematopoietic stem cells negatively controls cell cycle progression and maintains them in a dormant state.

IT IS WIDELY CONSIDERED that pluripotent hematopoietic stem cells (PHSC) are highly quiescent in normal steady state bone marrow (BM), and that dormancy plays an important role in their preservation.1 However, mechanisms involved in retaining PHSC at a dormant state are not well understood. Several cytokines are involved in initiation of cell cycle progression of the dormant PHSC.1 Those cytokines activate signal transduction pathways via their own receptors in the PHSC to induce expression of immediate early genes. Those early gene products function as transcription factors that regulate expression of target genes. Many transcription factors control proliferation and differentiation of PHSC.2,3 Although lineage-restricted transcription factors such as tal-1/SCL and rtbn2/LMO2 may be candidates as key regulator for differentiation of PHSC,4-9 widely or even ubiquitously expressed transcription factors may have a special role in maintaining PHSC in a dormant state.2,3 

The c-fos proto-oncogene, one of the immediate early genes, is transiently expressed on stimulation by external stimuli leading to cell cycle progression.10 Its product (c-Fos) forms a complex with the product of another proto-oncogene c-jun (AP-1) that regulates expression of AP-1–binding genes at their transcriptional level.10-12 Thus, c-Fos may play a key role in the transduction of signals induced by external stimuli.12-14 c-Fos is known to be critical for the G0/G1 transition and cell cycle progression in fibroblasts.13,14The overexpression of c-fos in transgenic mice leads to a deregulated bone growth and results in sarcomas,15,16 and the overexpression in several cell lines leads to acceleration of cell cycle progression.17,18 On the contrary, overexpression of c-Fos negatively regulates cell cycle progression in some cell types.19 Thus, functions of c-Fos in cell cycle progression have remained open to question.

c-Fos is thought to be important in various differentiation processes as well as in development.2,10-12 Although the developmental capacity of PHSC lacking the c-fos gene appears to be fairly normal,20-23 functions of c-Fos in controlling proliferation and differentiation of PHSC are unknown. Inducible type transgenic mice are powerful tools to investigate the gene function at certain stages of development.24 Using the transgenic mice carrying the c-fos gene under the control of the interferon (IFN)-α/β–inducible Mx-promoter (Mx–c-fos mice),25 we have shown that c-Fos interferes cell cycle progression of mature B cells at the G1/S transition of the cell cycle.26Furthermore, c-Fos induces apoptosis in pro–B cells27 and germinal-center B cells28 from Mx–c-fos mice. Thus, functions of c-Fos in regulating proliferation and differentiation of PHSC can be investigated using PHSC from Mx–c-fos mice.

Recent progress in the stem-cell biology revealed that PHSC and progenitors can be identified in BM cells, based on their surface marker profile.29 They lack lineage-specific antigens (Lin) and express c-kit, H-2K, low level of Thy-1, and a high affinity to WGA.30-32 PHSC can be further isolated from committed progenitors by cell-surface staining with monoclonal antibody against Sca-1 or CD34, or by nuclear staining with supravital staining dye (rhodamine-123, Hoechst-33342).29,33-36 This method has made feasible studies on the nature of PHSC at the clonal level. We investigated the role of c-Fos in cell cycle progression of PHSC using primitive hematopoietic stem (LinSca-1+) cells isolated from Mx–c-fos mice and also c-fos–deficient mice. We show here that the c-fos gene is transiently expressed in LinSca-1+ BM cells stimulated with stem cell factor (SCF), interleukin-3 (IL-3), and IL-6. The prolonged expression of c-fos inhibits LinSca-1+ BM cells from entering the cell cycle. The role of c-Fos in maintenance of PHSC in a dormant state is discussed.

Mice.

C57BL/6CrSlc mice were purchased from Japan SLC Co, Ltd (Hamamatsu, Japan). Transgenic mice carrying the mouse c-fos gene under the control of the Mx gene promoter (Mx–c-fos)25 and c-fos–deficient mice,20 provided by Dr E.F. Wagner (IMP, Vienna, Austria), were maintained by heterozygous mating in our animal facility.

Reverse-transcribed polymerase chain reaction (RT-PCR) analysis.

Total RNA was extracted from 1 × 104 of LinSca-1+ BM cells using an ISOGEN total RNA isolating kit (Waco, Tokyo, Japan). RNAs were reverse-transcribed using Superscript (Life Technologies, Grand Island, NY) and oligo(dT) (Pharmacia, Piscataway, NJ), in a final volume of 20 μL, and 1 μL of cDNA was used for PCR. After an initial 7-minute incubation at 95°C, 22 cycles of PCR were performed using the following conditions: c-fos cDNA, denaturation at 95°C for 1.5 minutes, annealing at 55°C for 1.5 minutes, and polymerization at 72°C for 1.5 minutes; G3PDH cDNA, denaturation at 95°C for 1 minute, annealing at 60°C for 1 minute, and polymerization at 72°C for 1 minute. PCR primers for the cDNA amplification were as follows: the c-fos primers,375′-TTCTCGGGTTTCAACGCC-3′ and 5′-GGCGTTGAAACCCGAGAA-3′; and the G3PDH primers,38 5′-TGAAGGTCGGTGTGAACGGATTTGGC-3′ and 5′-CATGTAGGCCATGAGGTCCACCAC-3′.

PCR products were separated on a 1.5% agarose gel, transferred onto a nylon membrane (Boehringer Mannheim, Mannheim, Germany) and fixed by cross-linking with ultraviolet irradiation and by baking at 80°C for 3 hours. The filter was hybridized overnight with the digoxigenin (DIG)-labeled probe at 42°C. Following hybridization, the filter was washed twice with 0.1× standard saline citrate/0.1% sodium dodecyl sulfate at 68°C for 15 minutes. The probe on the filter was detected by sheep anti-DIG antibody conjugated with alkaline phosphatase. The antibody detection reaction was performed using the enhanced chemiluminescent detection system (Boehringer Mannheim GmbH, Mannheim, Germany). The probes were the full-length murine c-fos cDNA and murine G3PDH cDNA that were subcloned into pGEM vectors and labeled by DIG using PCR with T7 and SP6 primers.

Hematopoietic growth factors.

Purified recombinant human IL-6, purified recombinant murine IL-3, and conditioned medium (CM) of Chinese hamster ovary (COS) cells that had been transfected with an expression plasmid containing the murine SCF cDNA, were provided by Dr Tetsuo Sudo (Toray Industries, Kamakura, Japan). Concentration of SCF CM was assessed as 3 μg/mL in the proliferation assay by BaF cells transfected with mouse c-kit receptor. Recombinant murine SCF was purchased from Pepro Tech (Rocky Hill, NJ). Unless specified otherwise, SCF CM was used as a source of SCF. Recombinant human erythropoietin (Epo) was provided by Kirin Brewery (Tokyo, Japan). Concentration of the cytokines used here was as follows: IL-3, 200 U/mL; IL-6, 20 ng/mL; SCF CM, 5%; SCF, 100 ng/mL; Epo, 2 U/mL.

Antibodies.

Biotinylated monoclonal antibodies against B220 (RA3-6B2), Mac-1 (M1/70), Gr-1 (RB6-8C5), CD4 (GK1.5), CD8 (53-6.72), and TER119 were purchased from PharMingen (San Diego, CA) and used to detect lineage markers. Fluorescein isothiocyanate (FITC)-conjugated anti–Sca-1 (Ly6A/E) antibody was purchased from PharMingen. Biotinylated antibodies were visualized using Streptavidin-phycoerythrin (PE; PharMingen).

Isolation of LinSca-1+ cells from BM cells or fetal liver (FL) cells.

Total BM cells from Mx–c-fos mice and their littermates were stained with a cocktail of biotinylated monoclonal antibodies against lineage markers for 20 minutes at 4°C. FL cells isolated from c-fos–deficient embryos and their littermates on day 14.5 postcoitus were stained with a cocktail of biotinylated monoclonal antibodies against lineage markers except for anti–Mac-1 antibody, as described.39 After washing the cells 3 times with staining medium (phosphate-buffered saline with 3% fetal calf serum [FCS] and 0.1% sodium azide), the cells were treated with streptavidin-conjugated immunomagnetic beads (BioMag; Perceptive Diagnostics, Cambridge, MA) for 30 minutes to remove lineage marker highly positive cells. The remaining cells were collected and stained with FITC–anti–Sca-1 antibody and Streptavidin-PE at 4°C for 20 minutes. After washing, the cells were resuspended in staining medium supplemented with propidium iodide (PI; 1 μg/mL). Stained cells were analyzed by FACS Vantage (Becton Dickinson, San Jose, CA), and the LinSca-1+ cells were sorted and used as a primitive hematopoietic stem-cell fraction.40 

In vitro colony assay.

Methylcellulose culture was performed using the modified method32 described by Iscove.41 Briefly, 1 mL of culture medium contained an adequate number of total BM cells or sorted LinSca-1+ cells, 1.2% methylcellulose (Shin-etsu Chemical Co, Tokyo, Japan), alpha-medium (Flow Laboratories, North Ryde, Australia), 30% FCS (Flow Laboratories), 1% deionized bovine serum albumin (Sigma Chemical, St Louis, MO), 0.1 mmol/L β-2-mercaptoethanol (Eastman Organic Chemical, Rochester, NY), and appropriate concentrations of growth factors in the presence or absence of IFN-α/β (Sigma Chemical). The cultures were prepared in 35-mm nontissue culture dishes (Beckton Dickinson Labware, Lincoln Park, NJ) and incubated at 37°C in a humidified atmosphere of 5% CO2. The number of colonies was counted using an inverted microscope. When IFN-α/β was added in the culture on day 4, 100 μL of alpha-medium with 200 U of IFN-α/β was added to the methylcellulose culture dish.

Short-term liquid culture.

Total BM cells or sorted LinSca-1+ cells were cultured in a 24-well plate (Beckton Dickinson Labware) with 1 mL alpha-medium containing 20% FCS and appropriate concentrations of growth factors in the presence (200 U/mL) or absence of IFN-α/β under a humidified 5% CO2 atmosphere at 37°C. After 7 days of culture, cells were obtained and nucleated cells were counted using Trypan blue (Life Technologies).

Spleen colony assay.

The spleen colony assay of Till and McCulloch42 was used. Freshly isolated or cultured LinSca-1+cells were injected into lethally irradiated mice (9.0 Gy total body irradiation). The spleens were removed on day 8 or day 12 after transplantation, fixed in Bouin’s solution, and macroscopically visible colonies were counted and scored as colony forming unit in spleen (CFU-S).

Coculture of LinSca-1+ cells with stromal cells.

PA-6 stromal cells support myelopoiesis.43 PA-6 cells (3 × 105/well) were seeded in a 6-well plate (PRIMARIA, Beckton Dickinson Labware) 1 day before coculture. Five hundred sorted LinSca-1+ cells were cultured on the PA-6 stromal layer with 3 mL of alpha-medium containing 10% FCS in the presence (200 U/mL) or absence of IFN-α/β.

Cell cycle analysis.

Cell cycle analysis was performed as described by Nicoletti.44 Briefly, sorted LinSca-1+ cells were cultured for 24 hours with SCF, IL-3, and IL-6 in the presence or absence of IFN-α/β. Those cells were incubated in hypotonic lysing buffer (0.1% sodium citrate, 0.01% Triton X, 0.1 mg/mL RNase, and 0.1 mg/mL PI). DNA content in each nuclei was analyzed on FACScalibur (Becton Dickinson, Mountain View, CA) using Cell Quest software (Becton Dickinson) for Macintosh (Apple Computet Inc, Cupertino, CA).

5-bromo-2′-deoxyuridine (BrdU) incorporation assay.

Sorted LinSca-1+ cells were cultured with SCF, IL-3, and IL-6 in the presence or absence of IFN-α/β for 36 hours and pulsed with 10 μmol/L BrdU (Sigma) for 3 hours. Those cells were spread on a slide glass by Cytospin (Shandon Southern Instruments Inc, Sewickley, PA) and fixed with cold acetone for 10 minutes. The cells were then incubated with 2N HCl for 1 hour, followed by reaction with mouse monoclonal antibody to BrdU (Boehringer Mannheim, Indianapolis, IN). These cells were further incubated with F(ab′)2 fragment of anti-mouse Ig labeled with horseradish peroxidase (Nycomed Amersham plc, Buckinghamshire, UK). The DAB kit (Nichirei, Tokyo, Japan) was used to visualize peroxidase, and counter staining was done using hematoxylin.45 

Expression of the c-fos gene in primitive hematopoietic stem cells stimulated with SCF, IL-3, and IL-6.

To examine expression of the c-fos gene in primitive hematopoietic stem cells after stimulation, LinSca-1+ BM cells from Mx–c-fos mice and their control littermates were stimulated with SCF, IL-3, and IL-6 in the presence or absence of IFN-α/β. Expression of c-fos was analyzed by RT-PCR with Southern blotting (Fig 1A). c-fos RNA was detected in the LinSca-1+ cells from both Mx–c-fos and control mice at 1 hour but not at 7 hours after stimulation in the absence of IFN-α/β, thereby indicating transient expression induced in these hematopoietic stem cells. When the cells were stimulated with SCF, IL-3, and IL-6 in the presence of IFN-α/β (200 U/mL), a large amount of c-fos RNA was evident in the Mx–c-fos but not in control stem cells, even 7 hours after stimulation. When IFN-α/β was added to those cultures on day 0 and day 4, c-fos mRNA was continuously detected until day 7 of culture (Fig 1B). Because IFN-α/β receptor is present in almost every cell type, including hematopoietic stem cells,46 we concluded that all of the cells derived from Mx–c-fos LinSca-1+cells in the culture express the exogenous c-fos in the presence of IFN-α/β.

Fig. 1.

Prolonged expression of c-fos mRNA in primitive hematopoietic stem cells from Mx–c-fos mice. LinSca-1+ BM cells were cultured with SCF, IL-3, and IL-6 in the presence (200 U/mL) or absence of IFN-/β for 7 hours (A), or by the addition of IFN-/β (200 U/mL) on day 0 and day 4 (B). Levels of c-fos mRNA were measured by RT-PCR analysis followed by Southern blotting, as described in Materials and Methods. G3PDH mRNA served as an internal control for the amount of RNA.

Fig. 1.

Prolonged expression of c-fos mRNA in primitive hematopoietic stem cells from Mx–c-fos mice. LinSca-1+ BM cells were cultured with SCF, IL-3, and IL-6 in the presence (200 U/mL) or absence of IFN-/β for 7 hours (A), or by the addition of IFN-/β (200 U/mL) on day 0 and day 4 (B). Levels of c-fos mRNA were measured by RT-PCR analysis followed by Southern blotting, as described in Materials and Methods. G3PDH mRNA served as an internal control for the amount of RNA.

Close modal
Effect of the prolonged expression of c-fos on colony formation by primitive hematopoietic stem cells stimulated with SCF, IL-3, and IL-6.

Total BM cells or LinSca-1+ BM cells from Mx–c-fos mice and their control littermates were cultured with SCF, IL-3, and IL-6 in the presence of various concentrations of IFN-α/β, and the number of colonies was counted in those cultures on day 8. As shown in Fig 2A, colony formation by BM cells from Mx–c-fos mice was suppressed in the presence of IFN-α/β at more than 200 U/mL. However, the suppression was not so significant in control cultures even at 2,000 U/mL of IFN-α/β, although size of each colony was reduced at more than 1,000 U/mL of IFN-α/β (data not shown). The suppression was more evident in the LinSca-1+ cell cultures from Mx–c-fos mice in the presence of IFN-α/β (Fig 2B), which means that c-fos suppressed on onset of colony formation from primitive hematopoietic stem cells but not from progenitors.

Fig. 2.

Inhibitory effect of c-fos on colony formation by primitive hematopoietic stem cells. Total BM cells or LinSca-1+ BM cells from Mx–c-fos mice or control littermates were cultured with SCF, IL-3, and IL-6 with various concentrations of IFN-/β. On day 8 of culture, the number of colonies was scored. Results represent mean and SD of four dishes. The data presented are representative of two independent experiments.

Fig. 2.

Inhibitory effect of c-fos on colony formation by primitive hematopoietic stem cells. Total BM cells or LinSca-1+ BM cells from Mx–c-fos mice or control littermates were cultured with SCF, IL-3, and IL-6 with various concentrations of IFN-/β. On day 8 of culture, the number of colonies was scored. Results represent mean and SD of four dishes. The data presented are representative of two independent experiments.

Close modal

Kinetics of colony formation by LinSca-1+ cells was further examined by adding IFN-α/β to the cultures on day 0 and day 4 (Fig 3). The number of colonies increased and reached a plateau in both Mx–c-fos and control cultures on day 8 of culture in the absence of IFN-α/β. When IFN-α/β (200 U/mL) was added to the culture on day 0, colony formation was delayed and the plateau level was slightly reduced in the Mx–c-fos but not in the control cultures. Furthermore, the addition of IFN-α/β to those cultures on day 0 and day 4 slowed down the colony formation and lowered the plateau level in the Mx–c-fos cultures but not in the control cultures. These observations suggest that prolonged expression of c-fos suppresses the onset of colony formation by primitive hematopoietic stem cells stimulated with SCF, IL-3, and IL-6.

Fig. 3.

Kinetics of colony formation by primitive hematopoietic stem cells. LinSca-1+ BM cells (3 × 102) from Mx–c-fos mice or control littermates were cultured with SCF, IL-3, and IL-6 (□). IFN-/β (200 U/mL) was added on day 0 (•), or on day 0 and day 4 (○). The number of colonies was scored every 4 days. Results represent mean and SD of four dishes. The data presented are representative of two independent experiments.

Fig. 3.

Kinetics of colony formation by primitive hematopoietic stem cells. LinSca-1+ BM cells (3 × 102) from Mx–c-fos mice or control littermates were cultured with SCF, IL-3, and IL-6 (□). IFN-/β (200 U/mL) was added on day 0 (•), or on day 0 and day 4 (○). The number of colonies was scored every 4 days. Results represent mean and SD of four dishes. The data presented are representative of two independent experiments.

Close modal

To further analyze the inhibitory effect of c-fos on growth of the colony, we serially plotted growth rate of each colony through colony mapping study, as described by Ikebuchi.47 One hundred and fifty of LinSca-1+ cells per dish were cultured with SCF, IL-3, IL-6, and Epo, and emergence of a new colony later showed the CFU-Mix and its subsequent rate of proliferation were analyzed (Fig 4). Although the number of CFU-Mix was about 13 to 16 per dish in both Mx–c-fos and control cultures in the presence (200 U/mL) or absence of IFN-α/β, the onset of each colony was delayed in Mx–c-fos cultures but not in control cultures when IFN-α/β was added on day 0 of culture. When IFN-α/β was added to those cultures on day 0 and day 4, the onset of each colony was further delayed in the Mx–c-fos cultures. However, the proliferation rate of each colony developed in the Mx–c-fos cultures did not differ significantly from that in the control cultures in the presence of IFN-α/β. Becuase 200 U/mL of IFN-α/β can induce expression of the exogenous c-fos gene in BM cells from Mx–c-fos mice for more than 2 days,26,28 these results suggest that after entering the cell cycle c-fos does not inhibit growth of colonies derived from primitive hematopoietic stem cells.

Fig. 4.

Mapping study of colony formation by primitive hematopoietic stem cells. LinSca-1+ BM cells (1 × 102) from Mx–c-fos mice or control littermates were cultured with SCF, IL-3, IL-6, and Epo. IFN-/β (200 U/mL) was added on day 0, or on day 0 and day 4. Graphic presentation indicates cell number changes in individual colonies that later became a mixed colony (CFU-Mix). The data represent colonies identified in two plates. The data presented are representative of two independent experiments.

Fig. 4.

Mapping study of colony formation by primitive hematopoietic stem cells. LinSca-1+ BM cells (1 × 102) from Mx–c-fos mice or control littermates were cultured with SCF, IL-3, IL-6, and Epo. IFN-/β (200 U/mL) was added on day 0, or on day 0 and day 4. Graphic presentation indicates cell number changes in individual colonies that later became a mixed colony (CFU-Mix). The data represent colonies identified in two plates. The data presented are representative of two independent experiments.

Close modal
Effect of c-fos on cell growth of primitive hematopoietic stem cells stimulated with various combinations of cytokines in the liquid culture.

The combination of SCF, IL-3, and IL-6 provides one of the strongest signals for differentiation of stem cells and progenitors.1,36 Stimulation of SCF and IL-6 is required for expansion of immature cells48 and that of IL-3 alone supports differentiation of stem cells and progenitors in a cycling state.1,47 To examine which cytokine signals were inhibited by c-fos, we did short-term liquid cultures of stem cells from Mx–c-fos mice, using various combinations of cytokines by adding IFN-α/β (200 U/mL) on day 0 and day 4 of culture, and the number of nucleated cells was counted on day 7 of culture (Fig 5). When total BM cells were cultured with combinations of cytokines, cell growth in the Mx–c-fos cultures with SCF+IL-6 stimulation was suppressed in the presence of IFN-α/β. However, cell growth in Mx–c-fos cultures with SCF+IL-3+IL-6 or IL-3 alone was not suppressed, suggesting that the prolonged expression of c-fos inhibits cell growth of primitive hematopoietic stem cells. Indeed, cell growth in cultures of LinSca-1+ BM cells from Mx–c-fosmice but not from control littermates with all of the combinations of cytokines used was inhibited in the presence of IFN-α/β.

Fig. 5.

Inhibitory effect of c-fos on cell proliferation by primitive hematopoietic stem cells. LinSca-1+ BM cells from Mx–c-fos or control littermates were cultured with SCF+IL-3+IL-6, SCF+IL-6, or IL-3. IFN-/β (200 U/mL) was added to the culture on day 0 and day 4. On day 7 of culture, the number of viable cells was counted by Trypan blue dye exclusion. Results represent mean and SD of four dishes. The data presented are representative of three independent experiments. Purified recombinant SCF was used in this experiment.

Fig. 5.

Inhibitory effect of c-fos on cell proliferation by primitive hematopoietic stem cells. LinSca-1+ BM cells from Mx–c-fos or control littermates were cultured with SCF+IL-3+IL-6, SCF+IL-6, or IL-3. IFN-/β (200 U/mL) was added to the culture on day 0 and day 4. On day 7 of culture, the number of viable cells was counted by Trypan blue dye exclusion. Results represent mean and SD of four dishes. The data presented are representative of three independent experiments. Purified recombinant SCF was used in this experiment.

Close modal

To confirm the suppressive effect of c-fos on cell growth of primitive hematopoietic stem cells, cell cycle analysis of LinSca-1+ BM cells stimulated with SCF, IL-3, and IL-6 in the liquid culture was done using fluorescence-activated cell sorter (FACS). As shown in Fig 6A, the percentage of cells in the S/G2/M phase was approximately 15% in Mx–c-fos and control cultures in the absence of IFN-α/β and in control cultures in the presence of IFN-α/β (200 U/mL) 24 hours after stimulation. In contrast, only 3.3% of the LinSca-1+ cells from Mx–c-fos mice were in the S/G2/M phase in the presence of IFN-α/β. Similar results were obtained in the case of BrdU incorporation assay (Fig 6B). Approximately 30% of cells were BrdU positive in cultures from Mx–c-fos and from control mice in the absence of IFN-α/β and in control cultures in the presence of IFN-α/β 36 hours after stimulation. However, only 5% of cells were positive for BrdU in Mx–c-fos cultures in the presence of IFN-α/β.

Fig. 6.

Cell cycle analysis of primitive hematopoietic stem cells after stimulation. LinSca-1+ BM cells from Mx–c-fos or control littermates were cultured with SCF, IL-3, and IL-6 in the presence (200 U/mL) or absence of IFN-/β. (A) After 24 hours, the cells were lysed and the nuclei were stained with propidium iodide. DNA content in the nuclei was determined by FACS. Percentage of PI-labeled nuclei in S/G2/M phase of the cell cycle is indicated. (B) After 36 hours, cells were pulsed with BrdU for 3 hours. Incorporated BrdU was detected by anti-BrdU monoclonal antibody and positive cells were counted. Results represent mean and SD of four dishes. The data presented are representative of three independent experiments.

Fig. 6.

Cell cycle analysis of primitive hematopoietic stem cells after stimulation. LinSca-1+ BM cells from Mx–c-fos or control littermates were cultured with SCF, IL-3, and IL-6 in the presence (200 U/mL) or absence of IFN-/β. (A) After 24 hours, the cells were lysed and the nuclei were stained with propidium iodide. DNA content in the nuclei was determined by FACS. Percentage of PI-labeled nuclei in S/G2/M phase of the cell cycle is indicated. (B) After 36 hours, cells were pulsed with BrdU for 3 hours. Incorporated BrdU was detected by anti-BrdU monoclonal antibody and positive cells were counted. Results represent mean and SD of four dishes. The data presented are representative of three independent experiments.

Close modal

To examine the suppressive effect of c-fos on differentiation of hematopoietic stem cells, we performed the spleen colony assay.42 Five hundred LinSca-1+ BM cells were cultured with SCF+IL-3+IL-6 in the presence (200 U/mL) or absence of IFN-α/β. On day 4 of culture, cells were collected and injected into lethally irradiated mice. As shown in Table 1, generation of day-8 CFU-S but not day-12 CFU-S was suppressed in mice injected with these cultured cells from Mx–c-fos mice in the presence of IFN-α/β. The number of day-8 CFU-S and that of day-12 CFU-S reflect the number of hematopoietic progenitor cells and that of more primitive hematopoietic stem cells,42,49 respectively. Therefore, these results indicate that prolonged expression of c-fos suppresses differentiation of hematopoietic stem cells.

Table 1.

Spleen Colony Formation by LinSca-1+ BM Cells Following Prolonged Expression of c-fos

Mice IFN-α/β Day-8 CFU-S Day-12 CFU-S Day-8/Day 12 CFU-S Ratio
Control  −  5.3 ± 1.5 11.0 ± 3.7  0.48  
 +  7.5 ± 2.9 10.8 ± 2.9  0.69  
Mx–c-fos − 6.4 ± 2.5  10.3 ± 4.2  0.62  
 1.0 ± 1.4  13.7 ± 5.1  0.07 
Mice IFN-α/β Day-8 CFU-S Day-12 CFU-S Day-8/Day 12 CFU-S Ratio
Control  −  5.3 ± 1.5 11.0 ± 3.7  0.48  
 +  7.5 ± 2.9 10.8 ± 2.9  0.69  
Mx–c-fos − 6.4 ± 2.5  10.3 ± 4.2  0.62  
 1.0 ± 1.4  13.7 ± 5.1  0.07 

Five hundred LinSca-1+ BM cells from Mx–c-fos or control littermates were cultured with SCF + IL-3 + IL-6 in the presence (200 U/mL) or absence of IFN-α/β. On day 4 of culture, those cells were collected and injected into lethally irradiated mice. Spleens from the mice were removed on day 8 or day 12 after the injection, fixed in Bouin’s solution, and the number of macroscopically visible spleen colonies was determined. Results represent mean and SD of 5 mice. The data presented are representative of two independent experiments. Purified recombinant SCF was used in this experiment.

Effect of c-fos on hematopoiesis by primitive hematopoietic stem cells cultured on a stromal cell layer.

To investigate the suppressive effect of c-fos on hematopoiesis by primitive hematopoietic stem cells on a stromal cell layer, LinSca-1+ BM cells were cultured on a layer of PA-6 stromal cells. Medium in the cultures was changed twice a week, and the number of nonadherent cells was counted by Trypan blue exclusion. As shown in Fig 7, the number of cells in both Mx–c-fos and control cultures exponentially increased and reached a plateau after day 12 of culture. When IFN-α/β (200 U/mL) was added to medium from the start of culture, the number of cells in Mx–c-fos cultures did not clearly increase. When addition of IFN-α/β in the medium was ceased after day 12 of culture, the number of cells increased and caught up to the control level in the Mx–c-fos cultures within 1 week. These data suggest that prolonged expression of c-fos to primitive hematopoietic stem cells also inhibits stroma-dependent hematopoiesis.

Fig. 7.

Inhibitory effect of c-fos on the stroma-dependent hematopoiesis by primitive hematopoietic stem cells. LinSca-1+ BM cells (3 × 102 cells/well in 6-well tissue culture plate) were cultured on a PA-6 stromal-cell layer in the presence (closed square; 200 U/mL) or absence (□) of IFN-/β. On day 12 of culture, the cells from Mx–c-fos culture with IFN-/β were obtained and washed with medium, and continued to culture on the PA-6 stromal layer with (▪) or without (○) IFN-/β. Nonadherent cells obtained from at least four dishes by gentle pipetting at every medium change were rinsed once with medium, pooled, and counted. The data presented are representative of three independent experiments.

Fig. 7.

Inhibitory effect of c-fos on the stroma-dependent hematopoiesis by primitive hematopoietic stem cells. LinSca-1+ BM cells (3 × 102 cells/well in 6-well tissue culture plate) were cultured on a PA-6 stromal-cell layer in the presence (closed square; 200 U/mL) or absence (□) of IFN-/β. On day 12 of culture, the cells from Mx–c-fos culture with IFN-/β were obtained and washed with medium, and continued to culture on the PA-6 stromal layer with (▪) or without (○) IFN-/β. Nonadherent cells obtained from at least four dishes by gentle pipetting at every medium change were rinsed once with medium, pooled, and counted. The data presented are representative of three independent experiments.

Close modal
Factor-dependent and stroma-dependent hematopoiesis by primitive hematopoietic stem cells from c-fos–deficient mice.

FL cells from c-fos–deficient mice was used as the source of primitive hematopoietic stem cells because of the osteopetrosis in c-fos–deficient mice.20,21 Kinetics of colony formation by LinSca-1+ cells was examined in the cultures with SCF+IL-3+IL-6 or IL-3 alone. As shown in Fig 8, kinetics of colony formation did not significantly differ between c-fos–deficient and control littermates. In addition, stromal-dependent hematopoiesis by LinSca-1+ cells from c-fos–deficient mice was normal because kinetics of cell growth on a PA-6 stromal layer by the LinSca-1+ cells from c-fos–deficient mice showed similar features to those seen in case of control littermates (Fig9). These results suggest that endogenous c-fos is not required for cell cycle progression of primitive hematopoietic stem cells.

Fig. 8.

Kinetics of colony formation by primitive hematopoietic stem cells from c-fos–deficient mice. LinSca-1+ cells (3 × 102) from FL of c-fos–deficient mice or control littermates were cultured with SCF, IL-3, and IL-6 (□), or IL-3 alone (▪). The number of colonies was scored and plotted. Results represent mean and SD of four dishes. The data presented are representative of three independent experiments.

Fig. 8.

Kinetics of colony formation by primitive hematopoietic stem cells from c-fos–deficient mice. LinSca-1+ cells (3 × 102) from FL of c-fos–deficient mice or control littermates were cultured with SCF, IL-3, and IL-6 (□), or IL-3 alone (▪). The number of colonies was scored and plotted. Results represent mean and SD of four dishes. The data presented are representative of three independent experiments.

Close modal
Fig. 9.

Kinetics of the stroma-dependent hematopoiesis by primitive hematopoietic stem cells from c-fos–deficient mice. LinSca-1+ cells (3 × 102cells/well in 6-well tissue culture plate) from FL of c-fos–deficient mice (○) or control littermates (c-fos+/+, □; c-fos+/−, ▪) were cultured on a PA-6 stromal cell layer. The number of nonadherent cells was counted (see Fig 7 legend). The data presented are representative of three independent experiments.

Fig. 9.

Kinetics of the stroma-dependent hematopoiesis by primitive hematopoietic stem cells from c-fos–deficient mice. LinSca-1+ cells (3 × 102cells/well in 6-well tissue culture plate) from FL of c-fos–deficient mice (○) or control littermates (c-fos+/+, □; c-fos+/−, ▪) were cultured on a PA-6 stromal cell layer. The number of nonadherent cells was counted (see Fig 7 legend). The data presented are representative of three independent experiments.

Close modal

In the steady state, the majority of hematopoietic stem cells reside in BM at a dormant state,1,49,50 and only a few stem cells supply all of the hematopoietic cells at a given time.1,51Because hematopoietic stem cells in BM are always exposed to various forms of stimuli, mechanisms to maintain the stem cells in a dormant state no doubt exist. We found that c-fos negatively controls cell cycle progression of primitive hematopoietic stem cells. Initiation of cell cycling and subsequent proliferation of stem cells appear to require collaboration of early acting cytokines. Ogawa proposed that cytokines regulating proliferation of primitive hematopoietic progenitors may be separated into three arbitrary groups.1 The combination among these three groups such as SCF, IL-3, and IL-6 is one of the most effective stimuli supporting the early process of hematopoiesis.1,36 These stimulations transiently induce expression of c-fos in primitive hematopoietic stem cells (Fig 1). Because prolonged expression of c-fos inhibits G0/G1 transition of dormant hematopoietic stem cells in both cytokine-dependent (Figs 2 to 5) and stroma-dependent (Fig 8) hematopoiesis, downregulation of the c-fos may initiate G0/G1 transition. Indeed, cell proliferation began in the stem cell culture from Mx–c-fos mice whenever addition of IFN-α/β to the culture was stopped (Fig 8). Therefore, c-Fos may be a gate keeper for cell cycle entry of dormant hematopoietic stem cells.

c-Fos is an important positive regulator of cell growth and notably of the G0/G1 transition.2,10-12,14 Expression of c-fosantisense RNA or injection of anti-Fos antibodies inhibit serum-stimulated cells to enter into S phase of the cell cycle and to reduce the growth rate of asynchronously growing cells.52In addition, overexpression of c-fos in fibroblasts and myeloid cell lines accelerates growth rate.17,18 On the other hand, Balsalobre and Jolicoeur19 showed that G0-S progression of rat-1 fibroblasts was delayed by overexpression of c-fos. We reported that prolonged expression of c-fos also perturbs cell cycle progression of mature B cells by surface Ig cross-linking.26 The c-fos gene is transiently induced in B cells, and prolonged expression inhibits B cells from passing into S phase of the cell cycle. This perturbation of cell cycle progression is due to poor degradation of the cyclin kinase inhibitor p27kip1 in the G1 phase. However, prolonged expression of c-fos accelerates cell cycle progression of mature B cells stimulated with LPS.53 Taken together, c-Fos may act as a negative and as a positive regulator of cell growth that is dependent on cell types, stimulation signals, differentiation stages, and cell cycle states.

The inhibitory effect of c-fos on cell cycle progression of dormant hematopoietic stem cells was also shown in a mapping study (Fig4). However, growth rate was not significantly affected by the expression after colony formation began. This is supported by the finding that colony formation by total BM cells was affected by c-fos much less than that by purified LinSca-1+ BM cells because the majority of colony-forming cells in total BM are committed progenitors. These results suggest that endogenous c-fos negatively regulates cell cycle progression of dormant hematopoietic stem cells.

Embryonal stem cells and 3T3-type fibroblasts lacking the c-fosgene divide at a normal rate.54,55 Furthermore, c-fos–deficient mice are viable but do have osteopetrosis, as a primary pathology.20,21 Although they have extramedullary hematopoiesis in the spleen, B lymphopenia, and thymic atrophy,20,21 we have found that hematopoietic stem cells can appear normal,22 except for failure differentiation into functional osteoclasts.56 In the present study, we found that both factor-dependent and stroma-dependent hematopoiesis by primitive hematopoietic stem cells from c-fos–deficient mice are normal (Fig 8 and 9). Colony formation by stem cells stimulated with IL-3+IL-6+SCF or IL-3 alone was almost the same as that seen in control littermates (Fig 8). These results indicate that c-fos is not essential for cell proliferation in several cell types and that redundancy with other members of the fosgene family may exist.11,12 These results also support the notion that c-Fos is a gate keeper for cell cycle entry of primitive hematopoietic stem cells.

c-Fos is also implicated in apoptosis.11,57 Treatment of cells with antisense oligonucleotide against c-fos increased survival of growth factor–derived lymphoid cells,37suggesting that expression of c-fos may represent an early event in activating programmed cell death. High levels of c-fosexpression were also observed in mouse tissues in which apoptosis is part of normal development.57 We also reported that overexpression of c-fos induced apoptosis of CD43+pro–B cells.27 On the other hand, c-fos plays a protective function from apoptosis of fibroblasts in response to stress such as ultraviolet irradiation.58 Mapping study (Fig 4) and spleen colony assay (Table 1) showed that significant number of primitive hematopoietic stem cells could survive despite the prolonged expression of c-fos. Furthermore, hematopoietic stem cells with long-term repopulating ability remain after the liquid culture for 7 days with SCF, IL-3, and IL-6 in the presence of IFN-α/β (data not shown). These results indicate that c-fos is not related to apoptosis in the dormant-state hematopoietic stem cells. Alternatively, as SCF, IL-3, and IL-6 are survival factors for primitive hematopoietic stem cells,59,60 these factors may contribute to survival of hematopoietic stem cells with overexpression of c-fos.

In summary, the c-fos was transiently induced in primitive hematopoietic stem cells stimulated with SCF, IL-3, and IL-6. The prolonged expression of c-fos inhibited cell-cycle entry of primitive hematopoietic stem cells stimulated with SCF, IL-3, and IL-6 as well as in case of cultures on a stromal cell layer. Hematopoietic stem cells with the c-fos expression in culture survived in a dormant state and entered the cell cycle after c-fos was downregulated. We propose that c-Fos plays the role of gate keeper in cell cycle progression of dormant hematopoietic stem cells.

We thank Drs T. Suda and M. Hatano for helpful discussions, Dr E.F. Wagner for c-fos–deficint mice, Dr T. Sudo for reagents, E. Furusawa and N. Fujita for secretarial services, and M. Ohara for comments on the manuscript.

Supported in part by Grants-in-Aid from the Ministry of Education, Science, Sports and Culture of Japan and a Research Grant from the Inohana Foundation.

The publication costs of this article were defrayed in part by page charge payment. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. section 1734 solely to indicate this fact.

1
Ogawa
M
Differentiation and proliferation of hematopoietic stem cells.
Blood
81
1993
2844
2
Shivdasani
RA
Orkin
SH
The transcriptional control of hematopoiesis.
Blood
87
1996
4025
3
Orkin
SH
Transcriptional factors and hematopoietic development.
J Biol Chem
270
1995
4955
4
Shivdasani
RA
Mayer
EL
Orkin
SH
Absence of blood formation in mice lacking the T-cell leukemia oncoprotein tal-1/SCL.
Nature
373
1995
43
5
Robb
L
Elwood
NJ
Elefanty
AG
Kontgen
F
Li
R
Barnett
LD
Begley
CG
The scl gene product is required for the generation of all hematopoietic lineages in the adult mouse.
EMBO J
15
1996
4123
6
Robb
L
Lyons
I
Li
R
Hartley
L
Kontgen
F
Harvey
RP
Metcalf
D
Begley
CG
Absence of yolk sac hematopoiesis from mice with a targeted disruption of the scl gene.
Proc Natl Acad Sci USA
92
1995
7075
7
Yamada
Y
Warren
AJ
Dobson
C
Forster
A
Pannell
R
Rabbitts
TH
The T cell leukemia LIM protein Lmo2 is necessary for adult mouse hematopoiesis.
Proc Natl Acad Sci USA
95
1998
3890
8
Porcher
C
Swat
W
Rockwell
K
Fujiwara
Y
Alt
FW
Orkin
SH
The T cell leukemia oncoprotein SCL/tal-1 is essential for development of all hematopoietic lineages.
Cell
86
1996
47
9
Warren
AJ
Colledge
WH
Carlton
MB
Evans
MJ
Smith
AJ
Rabbitts
TH
The oncogenic cysteine-rich LIM domain protein rbtn2 is essential for erythroid development.
Cell
78
1994
45
10
Angel
P
Karin
M
The role of Jun, Fos and the AP-1 complex in cell-proliferation and transformation.
Biochem Biophys Acta
1072
1991
129
11
Piechaczyk
M
Blanchard
JM
c-fos proto-oncogene regulation and function.
Crit Rev Oncol Hematol
17
1994
93
12
Karin
M
Liu
Zg
Zandi
E
AP-1 function and regulation.
Curr Opin Cell Biol
9
1997
240
13
Kovary
K
Bravo
R
The jun and fos protein families are both required for cell cycle progression in fibroblasts.
Mol Cell Biol
11
1991
4466
14
Kovary
K
Bravo
R
Expression of different Jun and Fos proteins during the G0-to-G1 transition in mouse fibroblasts: In vitro and in vivo associations.
Mol Cell Biol
11
1991
2451
15
Wang
ZQ
Grigoriadis
AE
Mohle-Steinlein
U
Wagner
EF
A novel target cell for c-fos-induced oncogenesis: Development of chondrogenic tumours in embryonic stem cell chimeras.
EMBO J
10
1991
2437
16
Wang
ZQ
Liang
J
Schellander
K
Wagner
EF
Grigoriadis
AE
c-fos-induced osteosarcoma formation in transgenic mice: Cooperativity with c-jun and the role of endogenous c-fos.
Cancer Res
55
1995
6244
17
Miao
GG
Curran
T
Cell transformation by c-fos requires an extended period of expression and is independent of the cell cycle.
Mol Cell Biol
14
1994
4295
18
Lord
KA
Abdollahi
A
Hoffman-Liebermann
B
Liebermann
DA
Proto-oncogenes of the fos/jun family of transcription factors are positive regulators of myeloid differentiation.
Mol Cell Biol
13
1993
841
19
Balsalobre
A
Jolicoeur
P
Fos proteins can act as negative regulators of cell growth independently of the fos transforming pathway.
Oncogene
11
1995
455
20
Wang
ZQ
Ovitt
C
Grigoriadis
AE
Mohle-Steinlein
U
Rüther
U
Wagner
EF
Bone and haematopoietic defects in mice lacking c-fos.
Nature
360
1992
741
21
Johnson
RS
Spiegelman
BM
Papaioannou
V
Pleiotropic effects of a null mutation in the c-fos proto-oncogene.
Cell
71
1992
577
22
Okada
S
Wang
ZQ
Grigoriadis
AE
Wagner
EF
von Ruden
T
Mice lacking c-fos have normal hematopoietic stem cells but exhibit altered B-cell differentiation due to an impaired bone marrow environment.
Mol Cell Biol
14
1994
382
23
Jain
J
Nalefski
EA
McCaffrey
PG
Johnson
RS
Spiegelman
BM
Papaioannou
V
Rao
A
Normal peripheral T-cell function in c-Fos-deficient mice.
Mol Cell Biol
14
1994
1566
24
Gingrich
JR
Roder
J
Inducible gene expression in the nervous system of transgenic mice.
Annu Rev Neurosci
21
1998
377
25
Bachiller
D
Rüther
U
Inducible expression of the proto-oncogene c-fos in transgenic mice.
Arch Geschwulstforsh
60
1990
357
26
Kobayashi
K
Phuchareon
J
Inada
K
Tomita
Y
Koizumi
T
Hatano
M
Miyatake
S
Tokuhisa
T
Overexpression of c-fos inhibits down-regulation of a cyclin-dependent kinase-2 inhibitor p27Kip1 in splenic B cells activated by surface Ig cross-linking.
J Immunol
158
1997
2050
27
Hu
L
Hatano
M
Rüther
U
Tokuhisa
T
Overexpression of c-Fos induces apoptosis of CD43+ pro-B cells.
J Immunol
157
1996
3804
28
Inada
K
Okada
S
Phuchareon
J
Hatano
M
Sugimoto
T
Moriya
H
Tokuhisa
T
c-Fos induces apoptosis in germinal center B cells.
J Immunol
161
1998
3853
29
Morrison
SJ
Uchida
N
Weissman
IL
The biology of hematopoietic stem cells.
Ann Rev Cell Dev Biol
11
1995
35
30
Visser
JW
Bauman
JG
Mulder
AH
Eliason
JF
de Leeuw
AM
Isolation of murine pluripotent hemopoietic stem cells.
J Exp Med
159
1984
1576
31
Muller-Sieburg
CE
Whitlock
CA
Weissman
IL
Isolation of two early B lymphocyte progenitors from mouse marrow: A committed pre-pre-B cell and a clonogenic Thy-1-lo hematopoietic stem cell.
Cell
44
1986
653
32
Okada
S
Nakauchi
H
Nagayoshi
K
Nishikawa
S
Nishikawa
S
Miura
Y
Suda
T
Enrichment and characterization of murine hematopoietic stem cells that express c-kit molecule.
Blood
78
1991
1706
33
Spangrude
GJ
Heimfeld
S
Weissman
IL
Purification and characterization of mouse hematopoietic stem cells.
Science
241
1988
58
34
Osawa
M
Hanada
K
Hamada
H
Nakauchi
H
Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell.
Science
273
1996
242
35
Li
CL
Johnson
GR
Rhodamine123 reveals heterogeneity within murine Lin-, Sca-1+ hemopoietic stem cells.
J Exp Med
175
1992
1443
36
Okada
S
Nakauchi
H
Nagayoshi
K
Nishikawa
S
Miura
Y
Suda
T
In vivo and in vitro stem cell function of c-kit- and Sca-1-positive murine hematopoietic cells.
Blood
80
1992
3044
37
Colotta
F
Polentarutti
N
Sironi
M
Mantovani
A
Expression and involvement of c-fos and c-jun protooncogenes in programmed cell death induced by growth factor deprivation in lymphoid cell lines.
J Biol Chem
267
1992
18278
38
Yamamoto
H
Hatano
M
Iitsuka
Y
Mahyar
NS
Yamamoto
M
Tokuhisa
T
Two forms of Hox11 a T cell leukemia oncogene, are expressed in fetal spleen but not in primary lymphocytes.
Mol Immunol
32
1995
1177
39
Morrison
SJ
Hemmati
HD
Wandycz
AM
Weissman
IL
The purification and characterization of fetal liver hematopoietic stem cells.
Proc Natl Acad Sci USA
92
1995
10302
40
Spangrude
GJ
Scollay
R
A simplified method for enrichment of mouse hematopoietic stem cells.
Exp Hematol
18
1990
920
41
Iscove
NN
Sieber
F
Winterhalter
KH
Erythroid colony formation in cultures of mouse and human bone marrow: Analysis of the requirement for erythropoietin by gel filtration and affinity chromatography on agarose-concanavalin A.
J Cell Physiol
83
1974
309
42
Till
JE
McCulloch
EA
A direct measurement of radiation sensitivity of normal mouse bone marrow cells.
Radiat Res
14
1961
213
43
Kodama
HA
Amagai
Y
Koyama
H
Kasai
S
A new preadipose cell line derived from newborn mouse calvaria can promote the proliferation of pluripotent hemopoietic stem cells in vitro.
J Cell Physiol
112
1982
89
44
Nicoletti
I
Migliorati
G
Pagliacci
MC
Grignani
F
Riccardi
C
A rapid and simple method for measuring thymocyte apoptosis by propidium iodide staining and flow cytometry.
J Immunol Methods
139
1991
271
45
Fukuda
T
Yoshida
T
Okada
S
Hatano
M
Miki
T
Ishibashi
K
Okabe
S
Koseki
H
Hirosawa
S
Taniguchi
M
Miyasaka
N
Tokuhisa
T
Disruption of the Bcl6 gene results in an impaired germinal center formation.
J Exp Med
186
1997
439
46
Novick
D
Cohen
B
Rubinstein
M
The human interferon α/β receptor: Characterization and molecular cloning.
Cell
77
1994
391
47
Ikebuchi
K
Clark
SC
Ihle
JN
Hirai
Y
Ogawa
M
Interleukin-6 enhancement of interleukin 3-dependent proliferation of multipotential hemopoietic progenitors.
Proc Natl Acad Sci USA
84
1988
3445
48
Miura
N
Okada
S
Zsebo
KM
Miura
S
Suda
T
Rat stem cell factor and IL-6 preferentially support the proliferation of c-kit-positive murine hemopoietic cells rather than their differentiation.
Exp Hematol
21
1993
143
49
Hodgson
GS
Bradley
TR
Properties of haematopoietic stem cells surviving 5-fluorouracil treatment: Evidence for a pre-CFU-S cell?
Nature
281
1979
381
50
Lajtha
LG
Stem cell concepts.
Differentiation
14
1979
23
51
Jordan
CT
Lemischka
IR
Clonal and systemic analysis of long-term hematopoiesis in the mouse.
Genes Dev
4
1990
220
52
Kovary
K
Bravo
R
Existence of different fos/jun complexes during the G0-to-G1 transition and during exponential growth in mouse fibroblasts: Differential role of Fos protein.
Mol Cell Biol
12
1992
5015
53
Phuchareon
J
Tokuhisa
T
Deregulated c-Fos/AP-1 accelerates cell cycle progression of B lymphocytes stimulated with lipopolysaccharide.
Immunobiology
193
1995
391
54
Brusselbach
S
Mohle-Steinlein
U
Wang
ZQ
Schreiber
M
Lucibello
FC
Muller
R
Wagner
EF
Cell proliferation and cell cycle progression are not impaired in fibroblasts and ES cells lacking c-Fos.
Oncogene
10
1995
79
55
Hu
E
Mueller
E
Oliviero
S
Papaioannou
VE
Johnson
R
Spiegelman
BM
Targeted disruption of the c-fos gene demonstrates c-fos-dependent and-independent pathways for gene expression stimulated by growth factors or oncogenes.
EMBO J
13
1994
3094
56
Grigoriadis
AE
Wang
ZQ
Cecchini
MG
Hofstetter
W
Felix
R
Fleisch
HA
Wagner
EF
c-Fos: A key regulator of osteoclast-macrophage lineage determination and bone remodeling.
Science
266
1994
443
57
Smeyne
RJ
Vendrell
M
Hayward
M
Baker
SJ
Miao
GG
Schilling
K
Robertson
LM
Curran
T
Morgan
JI
Continuous c-fos expression precedes programmed cell death in vivo.
Nature
363
1993
166
58
Schreiber
M
Baumann
B
Cotten
M
Angel
P
Wagner
EF
Fos is an essential component of the mammalian UV response.
EMBO J
14
1995
5338
59
Leary
AG
Zeng
HQ
Clark
SC
Ogawa
M
Growth factor requirements for survival in G0 and entry into the cell cycle of primitive human hemopoietic progenitors.
Proc Natl Acad Sci USA
89
1992
4013
60
Katayama
N
Clark
SC
Ogawa
M
Growth factor requirement for survival in cell-cycle dormancy of primitive murine lymphohematopoietic progenitors.
Blood
81
1993
610

Author notes

Address reprint requests to Takeshi Tokuhisa, MD, Department of Developmental Genetics, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; e-mail: tokuhisa@med.m.chiba-u.ac.jp.

Sign in via your Institution