We used a murine model containing a disruption of the murine homologue (Fac) of Fanconi Anemia group C (FAC) to evaluate the role of Fac in the pathogenesis of bone marrow (BM) failure. Methylcellulose cultures of BM cells fromFac−/− and Fac+/+ mice were established to examine the growth of multipotent and lineage-restricted progenitors containing inhibitory cytokines, including interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and macrophage inflammatory protein-1α (MIP-1α). Clonogenic growth of Fac−/− progenitors was reduced by 50% at 50- to 100-fold lower concentrations of all inhibitory cytokines evaluated. We hypothesized that the aberrant responsiveness to inhibitory cytokines in clonogenic cells may be a result of deregulated apoptosis. To test this hypothesis, we performed the TUNEL assay on purified populations of primary BM cells enriched for hematopoietic progenitors or differentiated myeloid cells. After stimulation with TNF-α, accentuated apoptosis was observed in both populations of Fac−/− cells. In addition, deregulated apoptosis was also noted in the most immature phenotypic population of hematopoietic cells after stimulation with MIP-1α.Together these data suggest a role of Fac in affecting the signaling of multiple cytokine pathways and support cytokine-mediated apoptosis as a major mechanism responsible for BM failure observed in FA patients.

FANCONI ANEMIA (FA) is a complex genetic disorder characterized by progressive acquisition of bone marrow (BM) aplasia, chromosomal instability, predisposition to malignancies, and hypersensitivity to bifunctional alkylating agents.1-4 At least eight complementation groups have been identified on the basis of somatic cell fusion studies that result in a generally similar phenotype.5,6 Three of the eight complementation groups (A, C, and D) have been mapped to different chromosomal loci,5,7,8 and the cDNAs of the A and C genes have been identified.9-11 Unfortunately, the cloning of the A and C genes have not provided insight into the function of FA genes in cellular homeostasis.

Maintenance of normal hematopoiesis involves complex interactions between the stem/progenitor cells and multiple stimulatory and inhibitory molecules.12,13 The observation that Fanconi anemia complementation type C (FAC) patients acquire a progressive BM failure suggests that FAC plays a role in proliferation and/or survival of hematopoietic stem and progenitor cells. BM aplasia may occur by multiple mechanisms including alterations in the production or intracellular signaling of stimulatory and inhibitory cytokines. Two murine models containing a disruption of the murine homologue of FAC (Fac) have recently been developed to facilitate functional studies in primary cells in vitro and in vivo.14,15 Chen et al14 created a disruption in exon 8 of Fac, while Whitney et al15 used homologous recombination to create a disruption in exon 9. In both models, spontaneous chromosomal aberrations were observed, as well as an increase in chromosome breaks in splenic lymphocytes in response to bifunctional alkylating agents analogous to lymphocytes in FA patients.14,15 

Whitney showed that hematopoietic progenitors from mice containing a disruption of Fac (Fac−/−) were hypersensitive to IFN-γ and, recently, Rathbun et al, using the model developed by Whitney,15 determined that low doses of IFN-γ affected programmed cell death inFac−/− hematopoietic progenitors by inducing Fas expression. Similar results were obtained in studies using primary cells from a patient with Fanconi anemia type C.16 

Other inhibitory cytokines, such as tumor necrosis factor-α (TNF-α), have also been implicated in the pathogenesis of aplastic anemia.17 In addition, TNF-α is frequently elevated in patients with Fanconi anemia.18,19 TNF-α is known to induce Fas-mediated apoptosis,17 as well as an alternative apoptosis pathway involving TNF-α receptor-associated death domain (TRADD).20 On the basis of these observations, we hypothesized that Fac-deficient cells may be hypersensitive to TNF-α and potentially other inhibitory cytokines.

In this report, using a genetically distinct model14 from that previously used by Whitney,15 we confirm that disruption of both alleles of Fac(Fac−/−) results in deregulated colony formation in response to IFN-γ. We also show that a disruption in exon 8 of Fac results in a hypersensitive reduction in progenitor growth in cultures containing TNF-α as well as another inhibitory cytokine macrophage inflammatory protein-1α (MIP-1α). Further, we show that Fac is crucial for prevention of TNF-α and MIP-1α–mediated apoptosis in primary immature myeloid hematopoietic cells. Finally, we observed altered proliferation kinetics in pluripotent and lineage restrictedFac−/− hematopoietic progenitors in vivo. These results implicate deregulated apoptosis in response to inhibitory cytokines in the pathogenesis of BM aplasia in Fanconi anemia type C.

Mice.

Heterozygote Fac mice14 were housed in our animal facility. DNA was extracted from mouse tails using a standard phenol-chloroform method. Genotyping was conducted by amplifying sequences unique to exon 8 and neomycin resistance gene by polymerase chain reaction (PCR). The following primers were used: 5′CCTGCCATCTTCAGAATTGT3′ (exon 8 primer), 5′GAGCAACACAAATGGTAAGG3′ (intron 8 primer), and 5′TTGAATGGAAGGATTGGAGC3′ (neomycin resistance gene primer).

Cells.

BM cells were flushed from femurs and tibias ofFac−/− and Fac+/+littermate controls with Iscove's modified Dulbecco's medium (IMDM) (GIBCO-BRL, Gaithersburg, MD) supplemented with 5% fetal calf serum (FCS) (Hyclone, Logan, UT). Spleen cell suspensions were prepared by flushing cells from spleens and passing cells through a 23-gauge needle. Total nucleated cell number of BM and spleens was determined before manipulation of cells for further experimentation. Low-density mononuclear cells (LDMNC) were prepared by centrifugation on Ficoll-Hypaque (density 1.119; Sigma Chemical Co, St Louis, MO). Unfractionated BM and spleen cells were used in all experiments except for mitomycin C (MMC) dose-response experiments, which used LDMNC.

Clonogenic assays.

Clonogenic methylcellulose assays were plated in triplicate at 1 × 104 to 5 × 104 BM cells/mL and 5 × 105 spleen cells/mL. Cultures were established in 1% IMDM methylcellulose (Stem Cell Technology, Vancouver, BC, Canada), with 30% FCS, 50 ng/mL recombinant murine Steel factor (a generous gift from Immunex, Seattle, WA) or SCF (Peprotech, Rocky Hill, NJ), 4 U/mL recombinant human erythropoietin (Amgen), 5% vol/vol pokeweed mitogen spleen conditioned media (PWMSCM) and 0.1 mmol hemin (Sigma). Cells were incubated at 37°C, 5% CO2, and lowered (5%) O2. BM cells were cultured in methylcellulose progenitor assays with increasing concentrations of MMC (Sigma), recombinant murine IFN-γ (R&D Systems, Minneapolis, MN), recombinant murine TNF-α (R&D Systems), recombinant murine MIP-1α (R&D Systems), and thrombopoietin (Genzyme, Cambridge, MA) to generate dose-response curves for Fac−/− andFac+/+ hematopoietic progenitor cells. Cultures to establish the responsiveness of Fac−/−cells to MMC were conducted exactly as above, except recombinant murine GM-CSF (Peprotech) was substituted for PWMSCM, and cultures were incubated at 21% O2. CFU-GM, BFU-E, and CFU-GEMM colonies were scored on day 7 of culture. Levels of significance were determined using Student's t-distribution.

Progenitor suicide assays.

The proportion of hematopoietic progenitor cells in S phase was estimated by means of suicide assays that used either3H-thymidine or hydroxyurea as previously described.21-23 BM and spleen cells were pulse-treated for 30 minutes at 37°C with either high specific activity3H-thymidine (50 mCi/mL, specific activity = 20 Ci/mmol; New England Nuclear, Boston, MA) or control medium. Cells were washed twice with control medium before plating in methylcellulose cultures as described above. The percentage suicide was determined by the following calculation: (progenitors in control − progenitors in3H-thymidine) divided by progenitors in control. The level of significance was determined using Student's t-test. These results were compared with the percentage suicide determined by treatment of cells with hydroxyurea (HU) as described elsewhere.23 

TUNEL assay.

Pooled samples of BM LDMNC from fourFac−/− and fourFac+/+ animals were stained with Sca1-PE, B220-FITC, and CD3-FITC (PharMingen, San Diego, CA) for 15 minutes at 4°C, washed twice, and sorted by a Becton Dickinson fluorescence activated cell sorter for Sca1+B220CD3 and Sca1B220CD3. Sca1+B220CD3 cells were incubated at a density of 2 × 105 cells/mL in a 96-well tissue culture plate in IMDM 20% FCS and were cultured in the following combinations of cytokines: (1) GM-CSF (200 ng/mL) and SCF (100 ng/mL) only, (2) GM-CSF and SCF together with 1 ng/mL TNF-α or 100 ng/mL MIP-1α, and (3) TNF-α (1 ng/mL) or MIP-1α (100 ng/mL). Cultures containing Sca1B220CD3cells were established at a density of 2 × 106cells/mL using the same concentrations of GM-CSF, TNF-α, and MIP-1α as above described for Sca1+B220CD3. After 24 hours, cytospins were made for each condition.

Apoptosis was evaluated using the terminal deoxynucleotidyl transferase (tDt)-mediated dUTP nick end-labeling (TUNEL) assay24 as specified by the manufacturer (Boehringer Mannheim, Indianapolis, IN). Briefly, cells were fixed with 4% formaldehyde (Sigma) for 30 minutes at room temperature and permeabilized with 0.1% sodium citrate (Fisher Scientific, Fair Lawn, NJ) 0.1% Triton-X100 (Boehringer Mannheim) for 2 minutes at 4°C. Cells were incubated with tDt and dUTP-FITC in a humidified environment at 37°C, 5% CO2 for 1 hour. After incubation, cytospins were evaluated by fluorescence microscopy. Photographs were obtained of each condition and scoring of apoptotic cells was conducted. As an independent control to verify the function of the assay, BM cells were irradiated (700 rads) and then maintained in liquid culture for 24 hours. The TUNEL assay was performed on these cells with and without the addition of tDt as positive and negative controls respectively. Three or four independent experiments were conducted with each inhibitory cytokine. Statistical significance was determined using the Student's t-test.

Fac−/− hematopoietic progenitor cells are hypersensitive to MMC.

A characteristic feature of Fanconi anemia is the hypersensitivity of cells to clastogenic agents such as mitomycin C. Because FA patients have defects in hematopoietic progenitor cell function, it was critical to determine whether hematopoietic progenitors fromFac−/− mice were hypersensitive to MMC as observed in FA patients. As shown in Fig1, a 50% reduction in maximal colony formation was observed at a 10-fold lower concentration of MMC inFac−/− progenitors as compared withFac+/+ littermates. We conclude that the MMC hypersensitivity observed in Fac−/−progenitors is remarkably similar to that seen in FA patients.

Fig. 1.

MMC hypersensitivity of Fac−/−hematopoietic progenitor cells. BM LDMNC fromFac−/− (▪) and Fac+/+(•) animals were cultured in clonogenic methylcellulose progenitor assays at 1 × 104 cells/mL with increasing concentrations of MMC. Each condition was plated in triplicate. The total number of colony-forming units (CFU) per 1 × 104 LDMNC was determined on day 7 of culture. Error bars represent standard error of the means (SEM). Fac−/− hematopoietic progenitor cells were significantly more sensitive to MMC at 5-, 10-, 50-, and 100-nmol concentrations. n = 6 for each group. *P< .05.

Fig. 1.

MMC hypersensitivity of Fac−/−hematopoietic progenitor cells. BM LDMNC fromFac−/− (▪) and Fac+/+(•) animals were cultured in clonogenic methylcellulose progenitor assays at 1 × 104 cells/mL with increasing concentrations of MMC. Each condition was plated in triplicate. The total number of colony-forming units (CFU) per 1 × 104 LDMNC was determined on day 7 of culture. Error bars represent standard error of the means (SEM). Fac−/− hematopoietic progenitor cells were significantly more sensitive to MMC at 5-, 10-, 50-, and 100-nmol concentrations. n = 6 for each group. *P< .05.

Close modal
Fac−/− hematopoietic progenitor cells are hypersensitive to multiple inhibitory cytokines.

IFN-γ, TNF-α, and MIP-1α are cytokines known to inhibit hematopoietic cell growth.25-29 We examined the effect of these inhibitory cytokines on growth of multipotential and lineage restricted progenitor cell growth of Fac mice in vitro (Fig 2). Multipotent (CFU-GEMM), erythroid (BFU-E), and granulocyte-macrophage (CFU-GM) progenitors cultured fromFac−/− BM cells showed a 50% reduction in maximal colony formation at a 50- to 100-fold lower concentration of each of the respective cytokines as compared with progenitors cultured from WT mice. No differences in colony formation were observed betweenFac−/− and Fac+/+hematopoietic progenitors when increasing concentrations of a noninhibitory cytokine (thrombopoietin) were added to the cultures (data not shown).

Fig. 2.

Hypersensitivity of Fac−/−hematopoietic progenitors to inhibitory cytokines. Unfractionated BM cells from −/− (▪) and +/+ (•) animals were cultured in triplicate in clonogenic methylcellulose progenitor assays at 5 × 104 cells/mL with increasing concentrations of IFN-γ (0.1, 0.5, 1, 5, and 10 ng/mL), TNF-α (0.1, 0.5, 1, 5, 10 ng/mL), or MIP-1α (1, 5, 10, 50 ng/mL). After 7 days in culture, CFU-GM, BFU-E, and CFU-GEMM were scored. Percentage maximal colony formation was determined by dividing the number of progenitors scored at a given concentration of cytokine by the number of progenitors scored without the addition of inhibitory cytokine. The range of control numbers upon which the percentage change are based:+/+ CFU-GM 77-141, BFU-E 41-43, CFU-GEMM,11-13 and −/− CFU-GM 80-154, BFU-E 11-43, and CFU-GEMM.5-13 The error bars represent SEM. CFU-GM, BFU-E, and CFU-GEMM from Fac−/−unfractionated BM cells were significantly more sensitive to IFN-γ, TNF-α, and MIP-1α at multiple cytokine concentrations. n = 4 for each group. *P < .05.

Fig. 2.

Hypersensitivity of Fac−/−hematopoietic progenitors to inhibitory cytokines. Unfractionated BM cells from −/− (▪) and +/+ (•) animals were cultured in triplicate in clonogenic methylcellulose progenitor assays at 5 × 104 cells/mL with increasing concentrations of IFN-γ (0.1, 0.5, 1, 5, and 10 ng/mL), TNF-α (0.1, 0.5, 1, 5, 10 ng/mL), or MIP-1α (1, 5, 10, 50 ng/mL). After 7 days in culture, CFU-GM, BFU-E, and CFU-GEMM were scored. Percentage maximal colony formation was determined by dividing the number of progenitors scored at a given concentration of cytokine by the number of progenitors scored without the addition of inhibitory cytokine. The range of control numbers upon which the percentage change are based:+/+ CFU-GM 77-141, BFU-E 41-43, CFU-GEMM,11-13 and −/− CFU-GM 80-154, BFU-E 11-43, and CFU-GEMM.5-13 The error bars represent SEM. CFU-GM, BFU-E, and CFU-GEMM from Fac−/−unfractionated BM cells were significantly more sensitive to IFN-γ, TNF-α, and MIP-1α at multiple cytokine concentrations. n = 4 for each group. *P < .05.

Close modal
Increased apoptosis in immature and differentiated myeloid hematopoietic cells from Fac−/− mice after stimulation with TNF-α and MIP-1α.

To test whether Fac−/− cells are predisposed to TNF-α– or MIP-1α–mediated apoptosis, two populations of hematopoietic cells were cultured in the presence of TNF-α or MIP-1α, then analyzed by the TUNEL assay. One population was highly enriched for myeloid hematopoietic progenitors (Sca1+B220CD3), and a second population was highly enriched for myeloid differentiated cells (Sca1B220CD3). More than 90% of Sca1B220CD3cells had macrophage (Mac1) or granulocyte (GR-1) surface markers (data not shown). Figure 3 shows representative examples of Sca1+B220CD3 cells after liquid culture with TNF-α and TUNEL assay. The summary of experiments evaluating apoptosis ofFac−/− cells in response to TNF-α are shown in Table 1. Low equivalent levels of apoptosis were observed in both Fac−/−and Fac+/+ cells when cultured with GM-CSF and SCF; cytokines that protect hematopoietic cells from apoptosis.30,31 The addition of TNF-α to cultures containing GM-CSF and SCF resulted in increased apoptosis inFac−/− primitive myeloid cells, but not in the Fac+/+ cells. When hematopoietic cells were cultured with TNF-α alone, a dramatic increase in apoptosis was observed in immature (Sca1+B220CD3) and differentiated (Sca1B220CD3) myeloid cells from Fac−/− mice.

Fig. 3.

Increased apoptosis of Sca1+B220CD3 and Sca1B220CD3 cells fromFac−/− mice in response to TNF-α. BM LDMNC from Fac−/− and Fac+/+animals were purified by fluorescence cytometry for Sca1+B220CD3 and Sca1B220CD3 cells. These two populations of cells were incubated in liquid culture for 24 hours with growth factors alone, growth factors plus TNF-α (1 ng/mL), or TNF-α alone. TUNEL assays were conducted on cytospins of each condition. A total of 100 to 200 cells were evaluated to determine the percentage of apoptotic cells in each condition. Sca1+B220CD3 cells in conditions containing TNF-α are depicted. InFac−/− mice, the percentage of Sca1+B220CD3 cells that are apoptotic increases significantly when cultured with TNF-α.

Fig. 3.

Increased apoptosis of Sca1+B220CD3 and Sca1B220CD3 cells fromFac−/− mice in response to TNF-α. BM LDMNC from Fac−/− and Fac+/+animals were purified by fluorescence cytometry for Sca1+B220CD3 and Sca1B220CD3 cells. These two populations of cells were incubated in liquid culture for 24 hours with growth factors alone, growth factors plus TNF-α (1 ng/mL), or TNF-α alone. TUNEL assays were conducted on cytospins of each condition. A total of 100 to 200 cells were evaluated to determine the percentage of apoptotic cells in each condition. Sca1+B220CD3 cells in conditions containing TNF-α are depicted. InFac−/− mice, the percentage of Sca1+B220CD3 cells that are apoptotic increases significantly when cultured with TNF-α.

Close modal
Table 1.

Percentage Apoptotic Scal+B220CD3 and ScalB220CD3 Cells After 24-Hour Liquid Culture

CytokinesScal+B220CD3ScalB220CD3
−/− +/+ −/− +/+
GM-CSF/SCF-150 9 ± 3 6 ± 2  11 ± 2  13 ± 5  
GM-CSF/SCF/-150 TNF-α 15 ± 7-151 2 ± 1  26 ± 9  14 ± 5 
TNF-α  31 ± 10-152 10 ± 2  43 ± 5-152 24 ± 3 
CytokinesScal+B220CD3ScalB220CD3
−/− +/+ −/− +/+
GM-CSF/SCF-150 9 ± 3 6 ± 2  11 ± 2  13 ± 5  
GM-CSF/SCF/-150 TNF-α 15 ± 7-151 2 ± 1  26 ± 9  14 ± 5 
TNF-α  31 ± 10-152 10 ± 2  43 ± 5-152 24 ± 3 

Results are expressed as mean ± SEM.

F0-150

SCF not added to ScalB220CD3 cells.

F0-151

P ≤ .03 comparing percentage change from GM-CSF/SCF to GM-CSF/SCF/TNF-α in −/− and +/+.

F0-152

P ≤ .05 comparing −/− with +/+.

Experiments evaluating apoptosis inFac−/− cells after culture with MIP-1α are summarized in Table 2. Again, low levels of apoptosis were detected inFac−/− and Fac+/+primitive and differentiated cells when cultured with GM-CSF and SCF. Similar levels of apoptosis were observed when MIP-1α was added to the cultures. However, culture of Sca1+B220CD3 cells from Fac−/− mice with MIP-1α alone resulted in a dramatically increased level of apoptosis as compared with Fac+/+ mice whose level of apoptosis was unchanged from conditions with protective cytokines. A trend (though not statistically significant) toward increased apoptosis was observed in Sca1B220CD3cells from Fac−/− mice. Together these data are consistent with the enhanced growth inhibition ofFac−/− progenitors in response to TNF-α and MIP-1α observed in the clonogenic assays (Fig 2).

Table 2.

Percentage Apoptotic Scal+B220CD3 and ScalB220CD3 Cells After 24-Hour Liquid Culture

CytokinesScal+B220CD3ScalB220CD3
−/− +/+ −/− +/+
GM-CSF/SCF* 9 ± 3 12 ± 2  15 ± 8  15 ± 8  
GM/CSF/SCF/* MIP-1α 9 ± 2  11 ± 3  11 ± 3  13 ± 6  
MIP-1α 25 ± 3 11 ± 4  21 ± 6  12 ± 9 
CytokinesScal+B220CD3ScalB220CD3
−/− +/+ −/− +/+
GM-CSF/SCF* 9 ± 3 12 ± 2  15 ± 8  15 ± 8  
GM/CSF/SCF/* MIP-1α 9 ± 2  11 ± 3  11 ± 3  13 ± 6  
MIP-1α 25 ± 3 11 ± 4  21 ± 6  12 ± 9 

Results are expressed as mean ± SEM.

*

SCF not added to ScalB220CD3 cells.

P ≤ .03 comparing −/− with +/+.

Increased suicide in hematopoietic progenitor cells cultured fromFac−/− mice.

The maintenance of normal hematopoietic cell populations results from a complex homeostasis of cell production and apoptosis. To determine whether there were differences in the absolute number of progenitors per organ in mice homozygous for disruption of Fac,methylcellulose cultures of splenic and BM cells were established. The absolute number of progenitors in the BM and spleen as well as total nucleated cells per organ of Fac−/−mice were similar to those of their Fac+/+littermates (Table 3). BecauseFac−/− hematopoietic progenitors are hypersensitive to multiple inhibitory cytokines and both immature and differentiated myeloid cells undergo increased apoptosis in response to TNF-α and MIP-1α, it is curious thatFac−/− mice have equivalent numbers of myeloid progenitors and differentiated cells asFac+/+ mice (Table 3). We hypothesized that if deregulated apoptosis were occurring in vivo,Fac−/− progenitors may exhibit abnormal proliferation kinetics. To investigate this hypothesis, BM and spleen cells from Fac−/− andFac+/+ mice were pulse treated with tritiated thymidine or hydroxyurea and cultured in methylcellulose for growth of progenitors. The percentage suicide of multipotential (CFU-GEMM), erythroid (BFU-E), and granulocyte-macrophage (CFU-GM) progenitors in both the spleen and BM was dramatically higher inFac−/− mice as compared withFac+/+ controls (Table 3). The percentage suicide was comparable using either tritiated thymidine or hydroxyurea. These data infer that an increased proportion of clonogenic hematopoietic progenitors from Fac−/− mice are in S phase and that altered proliferation kinetics exist in vivo.

Table 3.

Comparative Analysis of Absolute Numbers and Cycling Status of Myeloid Progenitor Cells From Bone Marrow and Spleen ofFac−/− and Wild-Type Littermate Controls

Progenitor Suicide Rates
MiceNucleated Cells × 106Progenitors × 103/OrganCFU-GMBFU-ECFU-GEMM
CFU-GMBFU-ECFU-GEMM3HTdRHU3HTdrHU3HTdrHU
Bone marrow  
+/+ 21.8 ± 2.1 33.2 ± 4.1 5.1 ± 1.3  1.7 ± 0.3  22.0 ± 3.9 21.5 ± 2.2  16.2 ± 5.1  14.7 ± 6.2 32.4 ± 5.0  29.1 ± 4.7  
−/−  19.3 ± 2.3 33.9 ± 6.6  7.0 ± 1.9  1.5 ± 0.3 58.9 ± 3.6  68.5 ± 1.4  59.0 ± 2.4 58.2 ± 2.8  69.8 ± 5.0  79.9 ± 3.4 
 (NS) (NS) (NS) (NS) (<.001) (<.001) (<.001) (<.001) (<.001) (<.001) 
Spleen  
+/+  119.6 ± 13.3  6.7 ± 1.4 6.4 ± 1.7  0.6 ± 0.1  0.2 ± 0.2  0 ± 0 1.9 ± 1.6  0.5 ± 0.3  1.6 ± 1.6  3.1 ± 3.2 
−/−  94.0 ± 10.6  6.5 ± 0.6  8.4 ± 2.2 0.7 ± 0.2  53.5 ± 3.6  50.9 ± 8.1 54.1 ± 3.0  52.4 ± 3.4  66.4 ± 4.5 70.3 ± 4.4 
 (NS) (NS) (NS) (NS) (<.001) (<.001) (<.001) (<.001) (<.001) (<.001) 
Progenitor Suicide Rates
MiceNucleated Cells × 106Progenitors × 103/OrganCFU-GMBFU-ECFU-GEMM
CFU-GMBFU-ECFU-GEMM3HTdRHU3HTdrHU3HTdrHU
Bone marrow  
+/+ 21.8 ± 2.1 33.2 ± 4.1 5.1 ± 1.3  1.7 ± 0.3  22.0 ± 3.9 21.5 ± 2.2  16.2 ± 5.1  14.7 ± 6.2 32.4 ± 5.0  29.1 ± 4.7  
−/−  19.3 ± 2.3 33.9 ± 6.6  7.0 ± 1.9  1.5 ± 0.3 58.9 ± 3.6  68.5 ± 1.4  59.0 ± 2.4 58.2 ± 2.8  69.8 ± 5.0  79.9 ± 3.4 
 (NS) (NS) (NS) (NS) (<.001) (<.001) (<.001) (<.001) (<.001) (<.001) 
Spleen  
+/+  119.6 ± 13.3  6.7 ± 1.4 6.4 ± 1.7  0.6 ± 0.1  0.2 ± 0.2  0 ± 0 1.9 ± 1.6  0.5 ± 0.3  1.6 ± 1.6  3.1 ± 3.2 
−/−  94.0 ± 10.6  6.5 ± 0.6  8.4 ± 2.2 0.7 ± 0.2  53.5 ± 3.6  50.9 ± 8.1 54.1 ± 3.0  52.4 ± 3.4  66.4 ± 4.5 70.3 ± 4.4 
 (NS) (NS) (NS) (NS) (<.001) (<.001) (<.001) (<.001) (<.001) (<.001) 

Results are expressed as mean ±1 SEM for 8 mice per group (from a total of 2 experiments), with each mouse assayed individually, for all values shown except for the progenitor suicide rates determined by hydroxyurea (HU) treatment in which 4 mice per group (from 1 experiment) were each assayed individually. The progenitor suicide rates determined by HU treatment can be compared with those values determined by the high specific activity tritiated thymidine (3THdr) kill assay. P values indicated in parentheses.

Abbreviation: NS, not significant (P > .05).

The development of murine models using homologous recombination to delete the murine homologue of a gene known to cause human disease is extremely useful for understanding the normal function of the gene, the pathogenesis of the human disease, and potential treatment strategies. Some murine models replicate the human disease completely, whereas the phenotype of other models is somewhat variant from the human disease.32 A characteristic feature of Fanconi anemia is the hypersensitive reduction in growth of hematopoietic progenitors cultured in methylcellulose in the presence of MMC. Therefore, it was critical to determine whether hematopoietic progenitors cultured fromFac−/− mice used in these studies were hypersensitive to MMC. The observation that hematopoietic progenitors derived from the murine BM are hypersensitive to MMC is particularly significant because it supports using this model to gain insight into the molecular mechanisms involved in the development of the progressive BM failure in FA patients.

BM failure or acquisition of myeloid leukemia, or both, are the most common causes of mortality in patients with FA.33,34 The precise role of FA genes in maintaining normal hematopoietic cellular homeostasis is the current challenge of many laboratories.35 The role of FA proteins in DNA repair,36 redox status of the cell,37-39 and apoptosis38,40,41 are three active areas of investigation. Cumming et al40 provided evidence that FAC may have a role in apoptosis when they demonstrated that overexpression of FACin a megakaryocytic IL-3–dependent cell line (MO7e cells), resulted in the prevention of growth factor dependent apoptosis. Other studies reported increased spontaneous apoptosis in FA lymphoblasts but decreased apoptosis when stressed with γ-irradiation as compared with lymphoblasts derived from normal donors.41 Although these studies in immortalized cell lines implicate FAC in the modulation of apoptosis, it is difficult to determine how accurately they reproduce the biochemical defect and cellular physiology of primary hematopoietic progenitor cells.

Rathbun et al16 recently made the first observation that deregulated apoptosis was evident in primaryFac−/− hematopoietic progenitor cells These investigators reported data demonstrating decreased clonogenic growth in human and murine Fac−/−progenitors in response to IFN-γ and/or anti-Fas activating antibody, which infers an increased sensitivity to Fas-mediated apoptosis.16 We chose to extend those observations by evaluating multiple inhibitory cytokines that modulate hematopoietic cell growth by alternative intracellular signaling pathways.17,20,42-44 IFN-γ and TNF-α have been implicated in the pathogenesis of acquired aplastic anemia by upregulating Fas expression on hematopoietic cells.19,42,43TNF-α has additional signaling mechanisms to regulate apoptosis through TNF receptor associated proteins, such as TRADD and receptor-interacting protein (RIP), and activation of NF-κB.20 MIP-1α is a chemokine that appears to induce growth inhibition through the Raf-1 kinase pathway.44 A further distinction between MIP-1α and both TNF-α and IFN-γ is that TNF-α and IFN-γ induce apoptosis in normal hematopoietic cells at high cytokine concentrations,45 whereas MIP-1α had not yet been evaluated for induction of apoptosis.

Our data, using a genetically distinct murine model, confirm previous findings that homozygous disruption of Fac results in hypersensitivity to IFN-γ.15,16 This hypersensitivity is particularly interesting as few data are available regarding the role of inhibitory cytokines in other genomic instability syndromes.46-49 In addition to hypersensitivity ofFac−/− progenitors to IFN-γ, we have shown decreased clonogenic growth in response to TNF-α and MIP-1α which was not observed previously.15 Furthermore, our data also support the concept of Fac directly or indirectly influencing programmed cell death by showing TNF-α and MIP-1α induced apoptosis using a methodology that permitted direct detection of apoptosis in primary progenitor and differentiated cells. We showed that purified populations of primary cells enriched for either hematopoietic progenitor or myeloid differentiated cells fromFac−/− mice undergo increased cytokine-mediated apoptosis after culture with TNF-α alone or in combination with cytokines that protect hematopoietic cells from apoptosis. In addition, we showed that primitive myeloid cells (Sca1+B220CD3) undergo enhanced apoptosis when cultured with MIP-1α as a single cytokine. This is the first description of a chemokine inducing apoptosis in hematopoietic cells. Together our findings and previous data16 are consistent with the hypothesis that there is a progressive loss of hematopoietic progenitor and stem cells inFac−/− mice and FA patients resulting directly or indirectly from inhibitory cytokine-mediated apoptosis.

We hypothesized that if apoptosis is involved in the pathogenesis of FA BM failure in vivo in Fac−/− mice, an increase in the number of proliferating hematopoietic progenitors would be required to sustain normal numbers of differentiated cells. To test this hypothesis we evaluated the suicide of hematopoietic progenitors using two independent agents. The markedly increased suicide observed in multipotent and lineage-restricted clonogenic cells from BM and spleen of Fac−/− mice supports the hypothesis that a higher proportion ofFac−/− hematopoietic progenitors in vivo are cycling due to loss of immature and differentiated cells from increased apoptosis. Alternatively, the increased suicide rate observed in Fac−/− progenitors may reflect an increased sensitivity to tritium and hydroxyurea. The rationale for using hydroxyurea as an agent to assess the cycling status of clonogenic progenitors was because of recent data showing that FAC-deficient lymphoblast cell lines exhibit normal cell-cycle kinetics after 24-hour exposure to the drug.50 The lack of hypersensitivity to hydroxyurea and the similar suicide rates between the two agents support the hypothesis that the increased suicide rate in Fac−/− progenitors is due to an accelerated proportion of clonogenic cells in S phase, rather than to hypersensitivity to the agents used to conduct these studies. However, to further delineate and confirm that an increased proportion ofFac−/− progenitor cells are in S phase, future studies evaluating phenotypically defined populations of cells are indicated.

In summary, we have shown that loss of Fac results in deregulated apoptosis in myeloid hematopoietic cells in response to inhibitory cytokines with distinctive intracellular signaling pathways. It will be interesting in future experiments to evaluate how the loss of Fac disturbs the tightly regulated intracellular signaling of IFN-γ, TNF-α, and MIP-1α in mediating hematopoietic cell growth. Fac−/− mice provide a valuable model system to evaluate the role of Fac in myeloid growth control.

We thank our colleagues at Indiana University and Dr Kevin Shannon (University of California, San Francisco) for reading the manuscript. We also thank Patricia Fox for secretarial support.

Supported by US Public Health Services Grants No. PO1 GK53586, P50 DK49218, RO1 HL56416, RO1 HL54037, R29 CA74177-01, and IF32 HL09851-01, and by the National Cancer Institute of Canada, Bayer Red Cross Program, and International Fanconi Anemia Research Foundation.

Address reprint requests to D. Wade Clapp, MD, Departments of Pediatrics and Microbiology/Immunology, Herman B Wells Research Center, 702 Barnhill Dr, Cancer Center 421, Indianapolis, IN 46202.

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

1
Auerbach
A
Fanconi anemia and leukemia: Tracking the genes.
Leukemia
6
1992
1
2
Auerbach
A
Fanconi anemia diagnosis and the diepoxybutane (DEB) test.
Exp Hematol
21
1993
731
3
Buchwald
M
Ng
J
Clarke
C
Duckworth-Rysiecki
G
Studies of gene transfer and reversion to mitomycin C resistance in Fanconi anemia cells.
Mutat Res
184
1987
153
4
Schroeder
T
Tilgen
D
Kruger
D
Vogel
F
Formal genetics of Fanconi's anemia.
Hum Genet
32
1976
257
5
Strathdee
CA
Duncan
AMV
Buchwald
M
Evidence for at least four Fanconi Anemia genes including FACC on chromosome 9.
Nature Genet
1
1992
196
6
Joenje
H
Oostra
AB
Wijker
M
di Summa
FM
van Berkel
CGM
Rooimans
MA
Ebell
W
van Weel
M
Pronk
JC
Buchwald
M
Arwert
F
Evidence for at least eight Fanconi anemia genes.
Am J Hum Genet
61
1997
940
7
Pronk
JC
Gibson
RA
Savoia
A
Wijker
M
Morgan
NS
Melchionda
S
Ford
D
Temtamy
S
Ortega
JJ
Jansen
S
Harenga
C
Cohn
RJ
de Ravel
TJ
Roberts
I
Westerveld
A
Easton
DJ
Joenje
H
Mathew
CG
Arwert
F
Localization of the Fanconi anemia complementation group A gene to chromosome 16q24.3.
Nature Genet
11
1995
338
8
Whitney
M
Thayer
M
Reifsteck
C
Olson
S
Smith
L
Jakobs
PM
Leach
RK
Naylor
S
Joenje
H
Grompe
M
Microcell mediated chromosome transfer maps the Fanconi anemia groups D gene to chromosome 3p.
Nature Genet
11
1995
341
9
Strathdee
C
Gavish
H
Shannon
W
Buchwald
M
Cloning of cDNAs for Fanconi's anaemia by functional complementation.
Nature
356
1992
763
10
Lo Ten Foe
JR
Rooimans
MA
Bosnoyan-Collins
L
Alon
N
Wijker
M
Parker
L
Lightfoot
J
Carreau
M
Callen
DF
Savoia
A
Cheng NC van Berkel
CG
Strunk
MH
Gille
JJ
Pals
G
Kruyt
FA
Pronk
JC
Arwert
F
Buchwald
M
Joenje
H
Expression cloning of a cDNA for the major Fanconi anaemia gene, FAA.
Nature Genet
14
1996
320
11
The Fanconi anaemia/breast cancer consortium
Positional cloning of the Fanconi anaemia group A gene.
Nature Genet
14
1996
324
12
Broxmeyer
HE
Suppressor cytokines and regulation of myelopoiesis: Biology and possible clinical uses.
Am J Pediatr
14
1992
22
13
Broxmeyer
HE
Myelosuppressive cytokines and peptides
Whetton
T
Gordon
T
Hemopoietic Growth Factors. Blood Cell Biochemistry, vol 7.
1996
121
Plenum
London, UK
14
Chen
M
Tomkins
D
Auerbach
W
McKerlie
C
Youssoufian
H
Liu
L
Gan
O
Carreau
M
Auerbach
A
Groves
T
Guidos
C
Freedman
M
Cross
J
Percy
D
Dick
J
Joyner
A
Buchwald
M
Inactivation of Fac in mice produces inducible chromosomal instability and reduced fertility reminiscent of Fanconi anaemia.
Nature Genet
12
1996
448
15
Whitney
M
Royle
G
Low
M
Kelly
M
Axthelm
M
Reifsteck
C
Olson
S
Braun
R
Heinrich
M
Rathbun
R
Bagby
G
Grompe
M
Germ cell defects and hematopoietic hypersensitivity to γ-interferon in mice with a targeted disruption of the Fanconi anemia C gene.
Blood
88
1996
49
16
Rathbun
RK
Faulkner
GR
Ostroski
MH
Christianson
TA
Hughes
G
Jones
G
Cahn
R
Maziarz
R
Royle
G
Keeble
W
Heinrich
MC
Grompe
M
Tower
PA
Bagby
GC
Inactivation of the Fanconi Anemia Group C gene augments interferon-γ–induced apoptotic responses in hematopoietic cells.
Blood
90
1997
974
17
Maciejewsk
IJ
Selleri
C
Anderson
S
Young
N
Fas antigen expression on CD34+ human marrow cells is induced by interferon γ and tumor necrosis factor α and potentiates cytokine-mediated hematopoietic suppression in vitro.
Blood
85
1995
3183
18
Schultz
J
Shahidi
N
Tumor necrosis factor-alpha overproduction in Fanconi's anemia.
Am J Hematol
42
1993
196
19
Rosselli
F
Sanceau
J
Gluckman
E
Wietzerbin
J
Moustacchi
E
Abnormal lymphokine production: A novel feature of the genetic disease Fanconi anemia. II. In vitro and in vivo spontaneous overproduction of tumor necrosis factor alpha.
Blood
83
1994
1216
20
Liu
Z
Hsu
H
Goeddel
DV
Karin
M
Dissection of TNF receptor 1 effector functions: JNK activation is not linked to apoptosis while NF-κB activation prevents cell death.
Cell
87
1996
565
21
Broxmeyer
H
Cooper
S
Williams
D
Hangoc
G
Gutterman
J
Vadhan-Raj
S
Growth characteristics of marrow hematopoietic progenitor/precursor cells from patients on a phase I clinical trial with purified recombinant human granulocyte-macrophage colony-stimulating factor.
Exp Hematol
16
1988
594
22
Broxmeyer
H
Benninger
L
Cooper
S
Hague
N
Benjamin
R
Vadhan-Raj
S
Effects of in vivo treatment with PIXY321 (GM-CSF/IL3 fusion protein) on proliferation kinetics of bone marrow and blood myeloid progenitor cells in patients with sarcoma.
Exp Hematol
23
1995
335
23
Broxmeyer
HE
Baker
F
Galbraith
Relationship of colony stimulating activity to apparent kill of human colony forming cells by irradiation and hydroxyurea.
Blood
47
1976
403
24
Negoescu
A
Lorimier
P
Labat-Molieur
F
Azoti
L
Robert
C
Guillermet
C
Brambilla
C
Brambilla
E
TUNEL: Improvement and evaluation of the method for in situ apoptotic cell identification.
Biochemica
2
1997
12
25
Broxmeyer
HE
Lu
L
Platzer
E
Feit
C
Juliaro
L
Rubin
BY
Comparative analysis of the influences of human gamma, alpha and beta interferons on human multipotential (CFU-GEMM), erythroid (BFU-E), and granulocyte-macrophage (CFU-GM) progenitor cells.
J Immunol
131
1993
1300
26
Broxmeyer
HE
Williams
DE
Lu
L
Cooper
S
Anderson
SL
Beyer
GS
Hoffman
R
Rubin
BY
The suppressive influences of human tumor necrosis factor on bone marrow hematopoietic cells from donors and patients with leukemia: Synergism of tumor necrosis factor and gamma interferon.
J Immunol
136
1986
4487
27
Graham
GJ
Wright
EG
Hewick
R
Wolpe
SD
Wilkie
NM
Donaldson
D
Lorimore
S
Pragnell
IB
Identification and characterization of an inhibitor of haemopoietic stem cell proliferation.
Nature
344
1990
442
28
Broxmeyer
HE
Sherry
B
Lu
L
Cooper
S
Oh
KO
Tekamp-Olson
P
Kwon
BS
Cerami
A
Enhancing and suppressing effects of recombinant murine macrophage inflammatory proteins on colony formation in vitro by bone marrow myeloid progenitor cells.
Blood
76
1990
1110
29
Cooper
S
Mantel
C
Broxmeyer
HE
Myelosuppressive effects in vivo with very low dosages of monomeric recombinant murine macrophage inflammatory protein-1α.
Exp Hematol
22
1994
186
30
Sakai
I
Kraft
AS
The kinase domain of Jak2 mediates induction of bcl-2 and delays cell death in hematopoietic cells.
J Biol Chem
272
1997
12350
31
Lotem
J
Sachs
L
Hematopoietic cytokines inhibit apoptosis induced by transforming growth factor beta 1 and cancer chemotherapy compounds in myeloid leukemic cells.
Blood
80
1992
1750
32
Wynshaw-Boris
A
Model mice and human disease.
Nature Genet
13
1996
259
33
Young
NS
The bone marrow failure syndromes
Nathan
DG
Oski
FA
Hematology of Infancy and Childhood, vol 1
4
1993
216
Saunders
Philadelphia, PA
34
Butturini
A
Gale
RP
Verlander
PC
Adler-Brecher
B
Gillio
A
Auerbach
AD
Hematologic abnormalities in Fanconi anemia. An International Fanconi Anemia Registry study.
Blood
84
1994
1650
35
D'Andrea
AD
Grompe
M
Molecular biology of Fanconi anemia: Implications for diagnosis and therapy.
Blood
90
1997
1725
36
Lambert
MW
Tsongalis
GJ
Lambert
WC
Parrish
DD
Correction of the FNA repair defect in Fanconi anemia complementation groups A and D cells.
Biochem Biophys Res Commun
230
1997
587
37
Joenje
H
Oostra
AB
Effect of oxygen tension on chromosomal aberrations in Fanconi anaemia.
Hum Genet
65
1983
99
38
Clarke
AA
Philpott
NJ
Gordon-Smith
EC
Rutherford
TR
The sensitivity of Fanconi anaemia group C cells to apoptosis induced by mitomycin C is due to oxygen radical generation, not DNA crosslinking.
Br J Haematol
96
1997
240
39
Ruppitsch
W
Meiblitzer
C
Weirich-Schwaiger
M
Klocker
H
Scheidereit
C
Schweiger
M
Hirsch-Kauffmann
M
The role of oxygen metabolism for the pathological phenotype of Fanconi anemia.
Hum Genet
99
1997
710
40
Cumming
RC
Liu
JM
Youssoufian
H
Buchwald
M
Suppression of apoptosis in hematopoietic factor-dependent progenitor cell lines by expression of the FAC gene.
Blood
88
1996
4558
41
Ridet
A
Guillouf
C
Duchaud
E
Cundari
E
Fiore
M
Moustacchi
E
Rosselli
F
Deregulated apoptosis is a hallmark of the Fanconi anemia syndrome.
Cancer Res
57
1997
1722
42
Selleri
C
Maciejewski
JP
Sato
T
Young
NS
Interferon-gamma constituitively expressed in the stromal microenvironment of human marrow cultures mediates potent hematopoietic inhibition.
Blood
87
1996
4149
43
Young
NS
Maciejewski
J
The pathophysiology of acquired aplastic anemia.
N Engl J Med
336
1997
1365
44
Aronica
SM
Mantel
C
Gonin
R
Marshall
MS
Sarris
A
Cooper
S
Hague
N
Zhang
XF
Broxmeyer
HE
Interferon-inducible protein 10 and macrophage inflammatory protein 1α inhibit growth factor stimulation of Raf-1 kinase activity and protein synthesis in a human growth factor-dependent hematopoietic cell line.
J Biol Chem
270
1995
21998
45
Selleri
C
Sato
T
Anderson
S
Young
NS
Maciejewski
JP
Interferon-gamma and tumor necrosis factor-alpha suppress both early and late stages of hematopoiesis and induce programmed cell death.
J Cell Physiol
165
1995
538
46
Suzuki
N
Suzuki
H
Kojima
T
Sugita
K
Takakubo
Y
Okamoto
S
Effects of human interferon on cellular response to UV in UV-sensitive human cell strains.
Mutat Res
198
1988
207
47
Gaspari
AA
Fleisher
TA
Kraemer
KH
Impaired interferon production and natural killer cell activation in patients with the skin cancer-prone disorder, xeroderma pigmentosum.
J Clin Invest
92
1993
1135
48
Siddoo-Atwal
C
Haas
AL
Rosin
MP
Elevation of interferon β-inducible proteins in ataxia telangiectasia cells.
Cancer Res
56
1996
443
49
Auerbach
AD
Verlander
PC
Disorders of DNA replication and repair.
Curr Opin Pediatr
9
1997
599
50
Heinrich
MC
Hoatlin
ME
Zigler
AJ
Silvey
KV
Bakke
AC
Keeble
WW
Zhi
Y
Reifsteck
CA
Grompe
M
Brown
MG
Magenis
RE
Olson
SB
Bagby
GC
DNA cross-linker-induced G2/M arrest in group C Fanconi anemia lymophoblasts reflects normal checkpoint function.
Blood
91
1998
275
Sign in via your Institution