Abstract
MDM2, an E3 ubiquitin ligase, is an important negative regulator of tumor suppressor p53. In turn the Mdm2 gene is a transcriptional target of p53, forming a negative feedback loop that is important in cell cycle control. It has recently become apparent that the ubiquitination of p53 by MDM2 can be inhibited when certain ribosomal proteins, including RPL5 and RPL11, bind to MDM2. This inhibition, and the resulting increase in p53 levels has been proposed to be responsible for the red cell aplasia seen in Diamond-Blackfan anemia (DBA) and in 5q- myelodysplastic syndrome (MDS). DBA and 5q- MDS are associated with inherited (DBA) or acquired (5q- MDS) haploinsufficiency of ribosomal proteins. A mutation in Mdm2 causing a C305F amino acid substitution blocks the binding of ribosomal proteins. Mice harboring this mutation (Mdm2C305F), retain a normal p53 response to DNA damage, but lack the p53 response to perturbations in ribosome biogenesis. While studying the interaction between RP haploinsufficiency and the Mdm2C305F mutation we noticed that Mdm2C305F homozygous mice had altered hematopoiesis. These mice developed a mild macrocytic anemia with reticulocytosis. In the bone marrow (BM), these mice showed a significant decrease in Ter119hi cells compared to wild type (WT) littermates, while no decrease in the number of mature erythroid cells (Ter119hiCD71low) was found in the spleen, which showed compensated bone marrow hematopoiesis. In methylcellulose cultures, BFU-E colonies from the mutant mice were slightly reduced in number and there was a significant reduction in CFU-E colony numbers in mutant mice compared with WT controls (p < 0.01). This erythropoietic defect was abrogated by concomitant p53 deficiency (Trp53ko/ko). Further investigation revealed that in Mdm2C305F animals, there was a decrease in Lin-Sca-1+c-Kit+ (LSK) cells, accompanied by significant decreases in multipotent progenitor (MPP) cells (p < 0.01). Competitive BM repopulation experiments showed that donor BM harboring the Mdm2C305F mutation possessed decreased repopulation capacity compared to WT BM, suggesting a functional stem cell deficit. These results suggest that there is a fine tuned balance in the interaction of ribosomal proteins with the MDM2/p53 axis which is important in normal hematopoiesis.
Introduction
Diamond-Blackfan Anemia (DBA) is a bone marrow failure syndrome characterized by macrocytic anemia and red cell aplasia, sometimes associated with developmental anomalies, chiefly cleft palate, thumb abnormalities and cardiovascular problems [1, 2]. Patients are usually diagnosed in their first year and can be treated with steroids or blood transfusion. In at least 70% of cases the disease is due to an inherited mutation in one of several ribosomal proteins, though the mechanism whereby haploinsufficiency for ribosomal proteins, which are required in all dividing cells, leads to specific problems with erythropoeisis, has not been elucidated [3]. In acquired 5q- MDS, patients also have a defect in erythroid cell differentiation with macrocytic anemia and red cell aplasia [4]. The gene for a ribosomal protein RPS14 is present in the deleted region and appears to be responsible for the development of the red cell defect [5]. In animal and cellular models, defects in red cell production caused by ribosomal protein deficiencies are rescued by the absence of p53, which responds to cellular signals and controls progression through cell cycle checkpoints [6–9]. p53 is regulated by the E3 ubiquitin ligase MDM2, which ubiquitinates p53 stimulating its turnover within the nucleus [10–15]. The Mdm2 gene is a transcriptional target of p53 thus forming a feedback loop that controls p53 and MDM2 levels [16]. Certain ribosomal proteins, notably RPL5 and RPL11, bind to MDM2 and block its ability to inhibit p53. This mechanism was demonstrated by the inability of ribosome biogenesis to affect p53 in cells homozygous for a mutant MDM2 with a C305F mutation, which fails to bind these ribosomal proteins (RPs) [17]. Cells homozygous for Mdm2C305F have a normal p53 response to DNA damage but not to ribosomal stress.
A crucial stage in mammalian erythropoiesis takes place in erythroblasts, rapidly proliferating cells with a maximum rate of ribosome production, needed for globin synthesis. The co-ordination between ribosome synthesis and cell division must be closely regulated in these cells, given the abortive effects on erythropoiesis when ribosome synthesis is perturbed. We therefore hypothesized that the RP/MDM2/p53 axis may play a crucial role in normal erythropoiesis. To test our hypothesis we studied erythropoiesis in homozygous Mdm2C305F mice compared to wild type (WT) mice. We found that Mdm2C305F mice had distinct abnormalities in erythropoiesis that point to the importance of RP/MDM2 binding in regulating red cell production.
Materials and Methods
Mdm2 mutant mice and animal experiments
We obtained mice carrying Mdm2C305F mutation from Dr. Zhang [18], and Trp53ko (129S2/SvPas) from the Jackson Laboratory. Animals were interbred to maintain Mdm2C305F, which was bred to C57BL/6 mice for studies. All mice were handled in strict accordance with our protocol (IACUC 911) as approved by the institutional animal care and use committee at the Children’s Hospital of Philadelphia Animal Care Facility.
Peripheral Blood cell counts were performed using a Hemavet HV950FS analyzer (Drew Scientific). Absolute reticulocyte counts and erythrocyte adenosine deaminase (eADA) were performed as previously described [19, 20]. For erythropoietin, lactate dehydrogenase (LDH), and bilirubin serum levels, serum from the mice was collected by centrifugation of whole peripheral blood at 1,000 g for 20 minutes. Erythropoietin levels in serum were determined by ELISA assay kit (R&D Systems) [21]. LDH and bilirubin in serum were analyzed used a Vistros 350 chemistry system (Ortho Clinical).
Histology and immunohistochemistry
Mouse peripheral blood smears were stained with May-Grunwald (Sigma-Aldrich) and Gimsa (Sigma-Aldrich) reagents. Mouse bone marrow and spleen tissue were fixed in 10% formalin. Bones were decalcified before dehydration, paraffin-embedded, and cut into 5 μm sections. For immunohistochemistry, Ter119 antibody (BD Biosciences) was used to stain formalin fixed paraffin embedded TMA slides. The sections were then incubated with TER-119 antibody at a 1:50 dilution for 1 hour at room temperature and then were incubated with biotinylated anti-rat IgG (Vector Laboratories) for 30 min at room temperature. After rinsing, sections were then incubated with the avidin biotin complex (Vector Laboratories) for 30 min at room temp. The sections were then rinsed and incubated with DAB (DAKO Cytomation) for 10 min at room temperature. The sections were also counterstained with hematoxylin and eosin (Sigma-Aldrich), rinsed, dehydrated in ethanol followed by xylene, then coverslipped. The preparations were examined by light microscopy (BX43; Olympus).
Flow cytometric measurements
For assessment of bone marrow and spleen progenitor cells by flow cytometry, single-cell suspensions of freshly prepared bone marrow and spleen were incubated with the following anti-mouse monoclonal antibodies: Ter119 (Ly-76, BD Biosciences), CD71 (C2, BD Biosciences), CD34 (RAM34, BD Biosciences), CD135 (A2F10, eBioscience), CD127 (A7R34, eBioscience), c-Kit (ACK2, eBioscience), SCA-1 (LY-6A/E, BD Biosciences), eFluor520 (eBioscience), and 7-AAD (eBioscience). Lineage (lin) staining included anti-mouse Ter119 (BD Biosciences), GR-1 (RB6-8C5, BD Biosciences), B220 (RA3-6B2, BD Biosciences), CD4 (RM4-5, BD Biosciences), CD8 (53–6.7, BD Biosciences), and CD11b (M1/70, BD Biosciences) antibodies [22–24]. Multicolor data acquisition for bone marrow progenitor subsets was performed on a FACSCanto II flow cytometer, and data were analyzed with FlowJo 7.6.1.
Colony-forming cell assays
Bone marrow (BM) cells were plated in triplicate in Methocult M3334 (StemCell Technologies) into 35 mm dishes at the density of 2 x 105 cells per dish and cultured for 2 days (CFU-E) or 4 days (mature BFU-E). BM cells were plated in triplicate in Methocult M3434 (StemCell Technologies) into 35 mm dishes at the density of 2 x 105 or 2 x 104 cells per dish and cultured for 7 days (BFU-E and CFU-GEMM) or 12 days (CFU-GM) [25]. Cells were stained with Benzidine dihydrochloride (Sigma-Aldrich) solution and visible colonies were counted.
Bone marrow transplantation and long-term repopulating assay
Competitive repopulation assays were performed by injecting 2.5×106 bone marrow cells from either experimental (Mdm2C305F/C305F) or control (Mdm2C305F/+, C57BL/6) donor animals on a CD45.2 background, along with 2.5×106 competitor cells from CD45.1 C57BL/6 mice into irradiated (900 cGy) C57BL/6 recipient mice (CD45.1) [26, 27]. Peripheral blood samples were collected at 4, 8, and 12 weeks following transplant to assess for the percentage of reconstitution of peripheral blood cell lineages with CD45.2 cells, thus defining relative repopulation capacity of bone marrow harvested from our experimental versus control mouse strains in each lineage analyzed. Leukocyte subsets were assessed after RBC lysis with ammonium chloride, using the following antibodies: FITC antibodies against, B220 (RA3-6B2, BD Biosciences), TCR β (H57-597, BD Biosciences), CD45.2 (104, BD Biosciences), and CD4 (GK1.5, BD Biosciences); PE antibodies against, CD11b (M1⁄70, BD Biosciences), Gr-1 (RB6-8C5, BD Biosciences), CD8 (53–6.7, eBioscience), and CD45.1 (A20, BD Biosciences).
Statistical analysis
Data were analyzed for statistical significance with the 2-tailed paired Student’s t-test (*p < 0.05;**P < 0.01, Excel 2013, Microsoft Corporation) for 2-group comparisons. Error bars were generated using the standard error of the mean.
Results
Hematologic characterization in Mdm2C305F/C305F mutant mice
To examine our hypothesis that co-ordination of ribosome biogenesis and the cell cycle is important in hematopoiesis we first studied peripheral blood counts in mice homozygous for Mdm2C305F/C305F. These mice displayed a mild macrocytic anemia with reticulocytosis (Table 1). Red cell morphology, other than a mild macrocytosis, was normal and there were no signs of hemolysis. Erythropoietin levels were higher in mutant mice compared to wild type, but this did not reach statistical significance (p = 0.118). eADA levels, known to be increased in red blood cells from patients with DBA due to ribosomal protein gene mutations, were normal in red blood cells from Mdm2C305F/C305F mutant mice. White blood cells and platelets were not significantly different between mutant and control mice. The anemia correlated with Mdm2C305F gene dosage with the homozygous mice exhibiting lower levels of hemoglobin and number of red blood cells than the heterozygous mice.
Table 1. Effects of Mdm2C305F mutant on blood counts, lactate dehydrogenase, bilirubin, erythropoietein level, and eADA activity.
| C57BL/6 | Mdm2C305F/+ | Mdm2C305F/C305F | Mdm2C305F/C305F;Trp53ko/ko | |
|---|---|---|---|---|
| No. of animals | 13 | 12 | 12 | 14 |
| WBC(x103/μL) | 7.19 ± 2.30 | 6.73 ± 1.54 | 6.27 ± 1.91 | 7.46 ± 1.65 |
| Neutrophil(x103/μL) | 0.69 ±0.28 | 0.89 ± 0.42 | 0.78 ± 0.24 | 1.51 ± 0.34 a§ |
| Lymphocyte(x103/μL) | 6.94 ± 1.96 | 5.60 ± 1.33 | 5.26 ± 1.65 | 5.70 ± 1.43 |
| Monocyte(x103/μL) | 0.27 ± 0.12 | 0.23 ± 0.07 | 0.20 ± 0.06 | 0.23 ± 0.09 |
| Eosinophil(x103/μL) | 0.02 ± 0.02 | 0.02 ± 0.01 | 0.03 ± 0.02 | 0.01 ± 0.01 c§ |
| Basophil(x103/μL) | 0.00 ± 0.01 | 0.01 ± 0.01 | 0.00 ± 0.01 | 0.00 ± 0.01 |
| RBC(x106/μL) | 11.45 ± 0.59 | 10.68 ± 0.93 | 9.87 ± 1.03 a† | 10.05 ± 0.50 |
| Hb(g/dL) | 14.74 ± 0.75 | 13.58 ± 1.00 | 13.53 ± 1.19 b† | 13.10 ± 0.79 |
| MCV(fL) | 51.89 ± 4.05 | 54.83 ± 4.14 | 58.88 ± 3.79 a† | 52.66 ± 0.94 a§ |
| Reticulocyte(‰) | 3.09 ± 0.81 | 4.05 ± 0.93 | 4.64 ± 1.29 b† | 4.89 ± 0.94 |
| Platelets(x103/μL) | 729.54 ± 113.81 | 722.08 ± 134.73 | 695.50 ± 86.27 | 812.00 ± 80.29 c§ |
| No. of animals | 7 | 7 | 6 | 4 |
| LDH(IU/L) | 522.71 ± 60.2 | 505.00 ± 53.42 | 565.67 ± 95.28 | 617.8 ± 159.01 |
| T-Bil(mg/dL) | 0.39 ± 0.12 | 0.39 ± 0.14 | 0.43 ± 0.05 | 0.40 ± 0.12 |
| No. of animals | 17 | 12 | 13 | 15 |
| Epo(pg/ml) | 168.73 ± 46.60 | 169.07 ± 44.40 | 204.93 ± 71.78 | 168.71 ± 66.43 |
| No. of animals | 9 | 8 | 12 | 5 |
| eADA(IU/g Hb) | 1.15 ± 0.18 | 1.16 ± 0.16 | 1.16 ± 0.22 | 1.11 ± 0.16 |
| No. of animals | 15 | 8 | 8 | 9 |
| Body weight(g) | 27.60 ± 4.48 | 29.27 ± 3.10 | 26.27 ± 3.48 | 29.17 ± 4.21 |
| No. of animals | 15 | 8 | 8 | 13 |
| Bone marrow cells(x107 cells) | 4.534 ± 0.561 | 4.337 ± 0.870 | 3.609 ± 0.770 b† | 3.925 ± 0.591 |
| No. of animals | 21 | 9 | 9 | 12 |
| Spleen(mg) | 79.5 ± 13.2 | 84.1 ± 14.2 | 100.3 ± 15.2 b† | 107.0 ± 29.3 |
Blood counts analyses of mice at 6 weeks of age. Bone marrow cellularity determined by flushing 2 femurs and 2 tibias. WBC indicates white blood cell count; RBC, red blood cell count; Hb, hemoglobin; MCV, mean corpuscular volume; LDH, lactate dehydrogenase; T-bil, total bilirubin; Epo, erythropoietin; and eADA, erythrocyte adenosine deaminase activity. All values are given as mean ± standard error of mean, two-tailed Student’s t-test,
aP < 0.001,
bP < 0.01,
cP < 0.05.
† C57BL/6 vs Mdm2C305F/C305F
§ Mdm2C305F/C305F vs Mdm2C305F/C305F;Trp53
We also found that homozygous mutant Mdm2C305F animals had a decreased number of nucleated cells in bone marrow and splenomegaly compared to control animals (Table 1). Mdm2C305F homozygous and heterozygous mice were indistinguishable from wild type mice with no noticeable differences in appearance and body weight. Animals were healthy and during the observation period of 12 months no tumors were observed in mutant or wild type mice.
The cellular makeup of Mdm2C305F homozygous spleens was determined by comparative histology and immunophenotyping using flow cytometry. Germinal centers were histologically normal, but the red pulp was increased (Fig 1), with increased staining for glycophorin-associated Ter119, a marker of erythroid cells.
Fig 1. Effects of Mdm2C305F mutant on peripheral blood, bone marrow, and spleen histology.
(A) Hematoxylin and eosin stained splenic sections (Magnification: 40x). (B) Ter119-stained (brown) splenic sections. Blue is counterstained lymphoid tissue.
In the bone marrow Mdm2C305F homozygous mice showed a significant decrease in Ter119high cells compared to wild type littermates as measured by flow cytometry. In contrast, while the spleen contained a decreased percentage of Ter119high cells, the total number of mature erythroid cells (Ter119hiCD71low) in the spleen was not decreased. This discrepancy between erythroid cell percentage and absolute number in the spleen was due to the splenomegaly and expanded red pulp seen in Mdm2C305F homozygous mice, which is consistent with compensatory increased extramedullar (splenic) erythropoiesis in these mice (Fig 2).
Fig 2. Effects of Mdm2C305F mutant on erythroid maturation in bone marrow and spleen.
Flow cytometric assessment of cell-surface Ter119 and CD71 expression in bone marrow and spleen, used to define population of proerythroblasts (I; Ter119medCD71hi), basophilic erythroblasts (II; Ter119hiCD71hi), chromatophilic erythroblasts (III; Ter119hiCD71med), and orthochromatic erythroblasts (IV; Ter119hiCD71low). Specific populations in control (n = 25), Mdm2C305F heterozygous (n = 9), Mdm2C305F homozygous (n = 8), and Mdm2C305F/C305F;Trp53ko/ko mice (n = 8). (A) Representative flow cytometry analysis of bone marrow. (B) Mdm2C305F homozygous mice showed a significant decrease in Ter119high cells compared to wild type littermates in the bone marrow. (C) Representative flow cytometry analysis of spleen. (D) Mdm2C305F homozygous mice showed no decrease in the number of mature erythroid cells (Ter119hiCD71low) compared to wild type littermates in the spleen.
In methyl cellulose cultures BFU-E colonies from the mutant mice were slightly reduced in number and there was an even greater reduction in CFU-E colony numbers in mutant mice compared with WT controls (p < 0.001) (Fig 3). CFU-GEMM and CFU-GM were not significantly different between mutant and control mice (Fig 3).
Fig 3. Hematopoietic colony assays in Mdm2C305F mutant mice.
Hematopoietic colony assays in C57BL/6 (n = 14), Mdm2C305F heterozygous (n = 7), Mdm2C305F homozygous (n = 7), and Mdm2C305F/C305F;Trp53ko/ko (n = 7) mice. Mean ± standard error of mean, two-tailed Student’s t-test (*P < 0.05, **P < 0.01). BFU-E colonies from the mutant mice were slightly reduced in number and there was a significant reduction in CFU-E colony numbers in mutant mice compared with wild type (WT) controls.
Mdm2C305F/C305F mutant mice are defective in stem cell renewal
To explore the ontogeny of the erythroid defects caused by the Mdm2C305F mutation in the mouse, we analyzed specific precursor populations in the bone marrow cells. Mdm2C305F mutant animals exhibited a significant decrease in Lin-Sca-1+c-Kit+ (LSK) cells, accompanied by significant decreases in multipotent progenitor (MPP) cells (p < 0.01) (Fig 4).
Fig 4. Effects of Mdm2C305F mutant on selected hematopoietic lineage subpopulation in bone marrow.
(A) Representative flow cytometry analysis of the specific precursor populations in control (n = 7), Mdm2C305F homozygous (n = 6), and Mdm2C305F/C305F;Trp53ko/ko mice (n = 6). (B) Mdm2C305F mutant animals exhibited a significant paucity in the HSPC population of LSKs (P < 0.01), accompanied by significant decreases of MPP populations (P < 0.01). HSPC indicates hematopoietic stem and progenitor cell; LSK, Lin-Sca-1+c-Kit+; LT-HSC, long-term hematopoietic stem cells; ST-HSC, short-term hematopoietic stem cells; MPP, multipotent progenitor; CLP, common lymphoid progenitor; and CMP, common myeloid progenitor.
Next, we were interested in whether the abnormality in Mdm2C305F mutant occurs during hematopoietic differentiation or also at the stem cell level. For this, competitive BM transplant experiments were performed using a 50:50 mixture of BM cells from either Mdm2C305F homozygous or heterozygous mutant mice and WT mice all at 8 weeks of age. After primary transplantation BM cells from Mdm2C305F homozygous mice contributed only little to circulating B and T lymphocytes compared to WT derived BM cells, whereas the contribution to the myeloid lineage in peripheral blood at day 28 after transplant was approximately equal to WT competitor cells but decreased with time after transplant (S1 Fig). In contrast, for Mdm2C305F heterozygous and WT BM cells the contribution was higher and remained stable during the observation period suggesting, a defect at an earlier precursor, possibly at a stem cell level.
The results from competitive BM transplant experiments indicate that Mdm2C305F homozygous BM cells have an intrinsic proliferative defect at an early progenitor level as well and possibly more pronounced during erythroid differentiation compared to Mdm2C305F heterozygous or WT mice.
Effects of Mdm2C305F/C305F are partially corrected by Trp53ko/ko
We next asked whether p53 was required for the observed hematologic changes in the Mdm2C305F mutant mouse. For this Mdm2C305F animals were crossbred with mice carrying a p53 knockout allele, to generate mice homozygous for Mdm2C305F and Trp53ko. In Mdm2C305F animals, deficiency for p53 (Trp53ko/ko) mitigated the increase in mean corpuscular volume (MCV) at 6 weeks of age, although the RBC count and Hemoglobin did not significantly increase and reticulocytosis persisted (Table 1). Double homozygous Mdm2C305F/C305F;Trp53ko/ko mice had a persistent splenomegaly with a significant increase in basophilic and chromatophilic erythroblasts (Fig 2). Furthermore, BFU-E's, CFU-E's, CFU-GEMM's, and MPP's from the Mdm2C305F/C305F;Trp53ko/ko mice were significantly increased in number compared to Mdm2C305F mutant mice (Figs 3 and 4). This suggests that the loss of p53 rescues impaired mutant Mdm2C305F erythropoiesis at least in part. Mdm2C305F/C305F;Trp53ko/ko mice showed also a significant increase in the number of neutrophil and platelets, however this was not different to the neutrophil and plated counts in p53 mutant mice thus likely caused by the p53 knockout background [28].
Discussion
DBA and 5q- MDS are associated with inherited and acquired haploinsufficiency of ribosomal proteins. Recent studies have implicated the role of p53 in mediating a cellular response to abnormal ribosome biogenesis and a ribosome deficient animal model showed p53-dependent abnormalities in development and hematopoiesis [29].
Oncogene-induced p53 activation is a major tumor-suppressor mechanism [10–12]. MDM2 is an important negative regulator of p53 that is important in cell cycle control. Recent study has demonstrated that Mdm2C305F mutation disrupted ribosomal protein binding and exhibited a normal p53 response to DNA damage but failed to activate p53 after perturbations in ribosome biogenesis [17].
While studying the interaction between RP haploinsufficiency, we noticed that Mdm2C305F mutant mice themselves had perturbed hematopoiesis. These mice showed a dramatic decrease in progenitor cells and a delayed erythroid differentiation without obvious myeloid and lymphoid lineages defects (Fig 5), a phenotype reminiscent to the defect described in CD34+ cells from DBA patients with inherited RPL11 mutations, underlining the importance of the co-ordination of ribosome biogenesis with cell cycle control in erythropoiesis[30].
Fig 5. Diagram of the hematopoietic lineage and differences observed in Mdm2C305F mutant mice.
(A) Mdm2C305F phenotype corresponded to blocks at MPP and orthochromatic erythroblast. LSK indicates Lin-Sca-1+c-Kit+; LT-HSC, long-term hematopoietic stem cells; ST-HSC, short-term hematopoietic stem cells; MPP, multipotent progenitor; CLP, common lymphoid progenitor; CMP, common myeloid progenitor; GMP, granulocyte/macrophage progenitor; PreMegE, megakaryocyte–erythroid progenitor; and MKP, megakaryocyte progenitor. (B) RP/MDM2 interaction may be important for the mature hematopoiesis rather than for the HSC proliferation.
Similar to the experiments using RPL11 deficient human CD34+ cells from patients with DBA, p53 knockout in our Mdm2 mutant animals partially mitigated the decrease in progenitor cells and the delayed hematopoiesis in our mice. However, although the progenitor numbers from Mdm2C305F mutant mice were mitigated by p53 knockout the animals were still anemic although they lost the Mdm2C305F associated macrocytosis, suggesting that the half-life of the mature Mdm2C305F p53 mutant red cells is decreased. Indeed double mutant Mdm2C305F p53 knockout mice had slightly elevated LDH levels and the accumulation of iron in their spleen (Table 1, S2 Fig).
Previous studies showed that MDM2 deficiency in mice results in embryonic lethality due to massive apoptosis and this phenotype was rescued by the p53515C allele, which encodes an apoptosis-deficient protein [16]. These mice died early, due to a severe impairment of progenitor cell expansion during postnatal hematopoiesis, leading to p53-dependent cell cycle arrest. These results show the regulation of p53 by MDM2 is important in hematopoiesis. In contrast to these studies, our Mdm2C305F mutant mice show similar survival to wild type [17].
Very recently the RPS19-deficient DBA with Mdm2C305F knock-in mice were published[31]. Mdm2C305F improved expansion of hematopoietic progenitors, but the extent of the erythroid rescue by Mdm2C305F was only partial, and the selective erythroid defect caused by Mdm2C305F.
Taken together, our findings with the Mdm2C305F knock in mice provide in vivo evidence that there is a delicate balance between ribosomal proteins and MDM2 binding and the stabilization of p53 as both increased RPL11 binding as well as the inability to bind MDM2 leads to p53 stabilization, a decreased number of progenitor cells and defective erythroid maturation.
Supporting Information
The percentage of total cell counts derived from donor mice is plotted for granulocytes and monocytes (Gr-1 and CD11b), B lymphocytes (B220), T lymphocytes (TCR β), CD4+ lymphocytes (CD4), CD8+ lymphocytes (CD8), and from peripheral blood of irradiated recipients 4, 8, or 12 weeks after BM transplantation with 5 x 106 Mdm2C305F mutant Bone marrow cells (CD45.2) mixed with CD45.1 allotype-marked wild type (WT) cells in 1:1 ratio. Donor ages are 6 weeks. n = 3–4 animals per genotype.
(TIF)
Prussian blue stained splenic sections (Magnification: 400x).
(TIF)
Acknowledgments
We thank Patricia Marko and Peter Nicholas for care of mice; Daniel Martines for histology and immunohistochemistry at the pathology core laboratory.
Data Availability
All relevant data are within the paper and its Supporting Information files.
Funding Statement
This work was supported by the Research Grant of Pediatric Oncology Research Fellowship from Children’s Cancer Association of Japan to TK, NIH/NHLBI K08 HL 122306 to TSO, and NIH R01-CA105312 to MB.
References
- 1.Vlachos A, Ball S, Dahl N, Alter BP, Sheth S, Ramenghi U, et al. Diagnosing and treating Diamond Blackfan anaemia: results of an international clinical consensus conference. Br J Haematol. 2008;142(6):859–76. 10.1111/j.1365-2141.2008.07269.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Lipton JM, Ellis SR. Diamond-Blackfan anemia: diagnosis, treatment, and molecular pathogenesis. Hematol Oncol Clin North Am. 2009;23(2):261–82. 10.1016/j.hoc.2009.01.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Farrar JE, Vlachos A, Atsidaftos E, Carlson-Donohoe H, Markello TC, Arceci RJ, et al. Ribosomal protein gene deletions in Diamond-Blackfan anemia. Blood. 2011;118(26):6943–51. 10.1182/blood-2011-08-375170 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Olney HJ, Le Beau MM. The cytogenetics of myelodysplastic syndromes. Best practice & research Clinical haematology. 2001;14(3):479–95. 10.1053/beha.2001.0151 . [DOI] [PubMed] [Google Scholar]
- 5.Ebert BL, Pretz J, Bosco J, Chang CY, Tamayo P, Galili N, et al. Identification of RPS14 as a 5q- syndrome gene by RNA interference screen. Nature. 2008;451(7176):335–9. 10.1038/nature06494 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.McGowan KA, Pang WW, Bhardwaj R, Perez MG, Pluvinage JV, Glader BE, et al. Reduced ribosomal protein gene dosage and p53 activation in low-risk myelodysplastic syndrome. Blood. 2011;118(13):3622–33. 10.1182/blood-2010-11-318584 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Lohrum MA, Ludwig RL, Kubbutat MH, Hanlon M, Vousden KH. Regulation of HDM2 activity by the ribosomal protein L11. Cancer cell. 2003;3(6):577–87. . [DOI] [PubMed] [Google Scholar]
- 8.Zhang Y, Wolf GW, Bhat K, Jin A, Allio T, Burkhart WA, et al. Ribosomal protein L11 negatively regulates oncoprotein MDM2 and mediates a p53-dependent ribosomal-stress checkpoint pathway. Molecular and cellular biology. 2003;23(23):8902–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Bhat KP, Itahana K, Jin A, Zhang Y. Essential role of ribosomal protein L11 in mediating growth inhibition-induced p53 activation. The EMBO journal. 2004;23(12):2402–12. 10.1038/sj.emboj.7600247 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Levine AJ. p53, the cellular gatekeeper for growth and division. Cell. 1997;88(3):323–31. . [DOI] [PubMed] [Google Scholar]
- 11.Vogelstein B, Lane D, Levine AJ. Surfing the p53 network. Nature. 2000;408(6810):307–10. 10.1038/35042675 . [DOI] [PubMed] [Google Scholar]
- 12.Jin S, Levine AJ. The p53 functional circuit. J Cell Sci. 2001;114(Pt 23):4139–40. . [DOI] [PubMed] [Google Scholar]
- 13.Dey A, Lane DP, Verma CS. Modulating the p53 pathway. Seminars in cancer biology. 2010;20(1):3–9. 10.1016/j.semcancer.2010.02.004 . [DOI] [PubMed] [Google Scholar]
- 14.Lane D, Levine A. p53 Research: the past thirty years and the next thirty years. Cold Spring Harbor perspectives in biology. 2010;2(12):a000893 10.1101/cshperspect.a000893 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bond GL, Hu W, Levine AJ. MDM2 is a central node in the p53 pathway: 12 years and counting. Curr Cancer Drug Targets. 2005;5(1):3–8. . [DOI] [PubMed] [Google Scholar]
- 16.Liu G, Terzian T, Xiong S, Van Pelt CS, Audiffred A, Box NF, et al. The p53-Mdm2 network in progenitor cell expansion during mouse postnatal development. The Journal of pathology. 2007;213(4):360–8. 10.1002/path.2238 . [DOI] [PubMed] [Google Scholar]
- 17.Macias E, Jin A, Deisenroth C, Bhat K, Mao H, Lindstrom MS, et al. An ARF-independent c-MYC-activated tumor suppression pathway mediated by ribosomal protein-Mdm2 Interaction. Cancer cell. 2010;18(3):231–43. 10.1016/j.ccr.2010.08.007 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Lindstrom MS, Jin A, Deisenroth C, White Wolf G, Zhang Y. Cancer-associated mutations in the MDM2 zinc finger domain disrupt ribosomal protein interaction and attenuate MDM2-induced p53 degradation. Molecular and cellular biology. 2007;27(3):1056–68. 10.1128/MCB.01307-06 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Chin-Yee I, Keeney M, Lohmann RC. Flow cytometric reticulocyte analysis using thiazole orange; clinical experience and technical limitations. Clin Lab Haematol. 1991;13(2):177–88. . [DOI] [PubMed] [Google Scholar]
- 20.Glader BE, Backer K, Diamond LK. Elevated erythrocyte adenosine deaminase activity in congenital hypoplastic anemia. N Engl J Med. 1983;309(24):1486–90. 10.1056/NEJM198312153092404 . [DOI] [PubMed] [Google Scholar]
- 21.Ikeda K, Mason PJ, Bessler M. 3'UTR-truncated Hmga2 cDNA causes MPN-like hematopoiesis by conferring a clonal growth advantage at the level of HSC in mice. Blood. 2011;117(22):5860–9. 10.1182/blood-2011-02-334425 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.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. 1992;80(12):3044–50. . [PubMed] [Google Scholar]
- 23.Keel SB, Doty RT, Yang Z, Quigley JG, Chen J, Knoblaugh S, et al. A heme export protein is required for red blood cell differentiation and iron homeostasis. Science. 2008;319(5864):825–8. 10.1126/science.1151133 . [DOI] [PubMed] [Google Scholar]
- 24.Olson TS, Caselli A, Otsuru S, Hofmann TJ, Williams R, Paolucci P, et al. Megakaryocytes promote murine osteoblastic HSC niche expansion and stem cell engraftment after radioablative conditioning. Blood. 2013;121(26):5238–49. 10.1182/blood-2012-10-463414 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Dutt S, Narla A, Lin K, Mullally A, Abayasekara N, Megerdichian C, et al. Haploinsufficiency for ribosomal protein genes causes selective activation of p53 in human erythroid progenitor cells. Blood. 2011;117(9):2567–76. 10.1182/blood-2010-07-295238 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Szilvassy SJ, Humphries RK, Lansdorp PM, Eaves AC, Eaves CJ. Quantitative assay for totipotent reconstituting hematopoietic stem cells by a competitive repopulation strategy. Proceedings of the National Academy of Sciences of the United States of America. 1990;87(22):8736–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Jaako P, Flygare J, Olsson K, Quere R, Ehinger M, Henson A, et al. Mice with ribosomal protein S19 deficiency develop bone marrow failure and symptoms like patients with Diamond-Blackfan anemia. Blood. 2011;118(23):6087–96. 10.1182/blood-2011-08-371963 . [DOI] [PubMed] [Google Scholar]
- 28.TeKippe M, Harrison DE, Chen J. Expansion of hematopoietic stem cell phenotype and activity in Trp53-null mice. Experimental hematology. 2003;31(6):521–7. . [DOI] [PubMed] [Google Scholar]
- 29.Danilova N, Sakamoto KM, Lin S. Ribosomal protein S19 deficiency in zebrafish leads to developmental abnormalities and defective erythropoiesis through activation of p53 protein family. Blood. 2008;112(13):5228–37. 10.1182/blood-2008-01-132290 . [DOI] [PubMed] [Google Scholar]
- 30.Moniz H, Gastou M, Leblanc T, Hurtaud C, Cretien A, Lecluse Y, et al. Primary hematopoietic cells from DBA patients with mutations in RPL11 and RPS19 genes exhibit distinct erythroid phenotype in vitro. Cell Death Dis. 2012;3:e356 10.1038/cddis.2012.88 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Jaako P, Debnath S, Olsson K, Zhang Y, Flygare J, Lindstrom MS, et al. Disruption of the 5S RNP-Mdm2 interaction significantly improves the erythroid defect in a mouse model for Diamond-Blackfan anemia. Leukemia. 2015. 10.1038/leu.2015.128 . [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
The percentage of total cell counts derived from donor mice is plotted for granulocytes and monocytes (Gr-1 and CD11b), B lymphocytes (B220), T lymphocytes (TCR β), CD4+ lymphocytes (CD4), CD8+ lymphocytes (CD8), and from peripheral blood of irradiated recipients 4, 8, or 12 weeks after BM transplantation with 5 x 106 Mdm2C305F mutant Bone marrow cells (CD45.2) mixed with CD45.1 allotype-marked wild type (WT) cells in 1:1 ratio. Donor ages are 6 weeks. n = 3–4 animals per genotype.
(TIF)
Prussian blue stained splenic sections (Magnification: 400x).
(TIF)
Data Availability Statement
All relevant data are within the paper and its Supporting Information files.





