Abstract The Ser-Thr kinase CK2 plays important roles in sustaining cell survival and resistance to stress and these functions are exploited by different types of blood tumors. Yet, the physiological involvement of CK2 in normal blood cell development is poorly known. Here, we discovered that the β regulatory subunit of CK2 is critical for normal hematopoiesis in the mouse. Fetal livers of conditional CK2β knockout embryos showed increased numbers of hematopoietic stem cells associated to a higher proliferation rate compared to control animals. Both hematopoietic stem and progenitor cells (HSPCs) displayed alterations in the expression of transcription factors involved in cell quiescence, self-renewal, and lineage commitment. HSPCs lacking CK2β were functionally impaired in supporting both in vitro and in vivo hematopoiesis as demonstrated by transplantation assays. Furthermore, KO mice developed anemia due to a reduced number of mature erythroid cells. This compartment was characterized by dysplasia, proliferative defects at early precursor stage, and apoptosis at late-stage erythroblasts. Erythroid cells exhibited a marked compromise of signaling cascades downstream of the cKit and erythropoietin receptor, with a defective activation of ERK/JNK, JAK/STAT5, and PI3K/AKT pathways and perturbations of several transcriptional programs as demonstrated by RNA-Seq analysis. Moreover, we unraveled an unforeseen molecular mechanism whereby CK2 sustains GATA1 stability and transcriptional proficiency. Thus, our work demonstrates new and crucial functions of CK2 in HSPC biology and in erythropoiesis. __________________________________________________________________ [68]graphic file with name hs9-7-e978-g001.jpg [69]Open in a new tab INTRODUCTION Hematopoiesis is a finely tuned developmental process regulated by cell-intrinsic (epigenetic, gene transcription) and extrinsic (cytokines, chemokines, and growth factors) mechanisms/molecules.^[70]1,[71]2 Signals generated by Stem cell factor (Scf)-cKit axis^[72]3 and Erythropoietin-Receptor (EPO-R)^[73]4 are fundamental for hematopoietic stem/progenitor cell (HSPC) growth and for red blood cell (RBC) production, respectively, through the activation of shared pathways, such as ERK/MAP kinases, JAK/STAT and PI3K/AKT signaling cascades. The EPO-R signaling culminates in the activation of the transcription factor GATA1^[74]4–[75]6 which sustains erythroid cell viability,^[76]6,[77]7 the switching from embryonic/fetal to adult globin chains,^[78]8 heme biosynthesis^[79]9 and supports also its own and Epo-R^[80]10 transcription. Protein kinase CK2 is a Ser/Thr kinase constituted by a tetramer of 2 catalytic (α or α’)^[81]11 and 2 regulatory (β) subunits, with CK2β being essential for the assembly of the kinase in a tetramer and for the selection of phospho-targets.^[82]12 However, the α and β subunits exert also functions independently from the holoenzyme^[83]12 and, importantly, free β dimers can bind and stabilize other proteins.^[84]13 CK2 is involved in cell proliferation, resistance to stress, as well as many other processes.^[85]14 One main property of CK2 is to sustain cell viability, hampering apoptosis by inhibiting procaspase-2 and caspase 8 and regulating BCL-2 family members.^[86]15 In particular, CK2 prevents the activation of pro-apoptotic BID and enhances the expression of anti-apoptotic BCL-XL.^[87]16 In addition, this kinase positively regulates survival pathways, such as NF-κB (in which it activates p65/RelA through phosphorylation in S529^[88]17 and enhances IκBα^[89]18 degradation and PI3K/AKT (through the inhibition of the phosphatase PTEN^[90]19 and the activation of AKT^[91]20,[92]21). We and others have demonstrated that CK2 is essential for acute myeloid leukemia (AML) and AML-derived leukemic stem cells,^[93]22,[94]23 multiple myeloma^[95]24–[96]26 and non-Hodgkin B cell lymphoma^[97]27–[98]30 cell survival, proliferation, and resistance to several intrinsic and extrinsic stresses.^[99]31 Since literature has also widely reported the key role of CK2 in embryogenesis,^[100]32–[101]34 we hypothesized that this kinase might also be essential for the ontogenesis of blood cells. In a recent article published by our group, we have demonstrated the importance of CK2 in B cell commitment towards follicular fate and in the germinal center reaction using mice bearing loxP-flanked CK2β alleles and expressing the Cre recombinase under the control of the CD19 promoter.^[102]35 In the present work, we crossed the same model of loxP mice with animals expressing the Cre recombinase under the control of the Vav1 promoter, thereby achieving CK2β KO in all the hematopoietic cells. We focused the analysis on the HSPC and erythroid compartments. CK2β KO during hematopoiesis dramatically affected mouse HSPC numbers and function ex vivo and in vivo. Mechanistically, we provided evidence that CK2β influences the expression of multiple transcription factors implicated in HSPC biology. Also, by exploiting an in vitro model of erythroid differentiation, we showed that CK2β is an essential stage-specific regulator of RBC differentiation, proliferation, and viability and we unraveled a molecular crosstalk between this kinase and the transcription factor GATA1. METHODS Mice C57BL/6 VAV1-CRE mice were purchased from Jackson Laboratory, C57BL/6 Csnk2b 2loxP mice were described in,^[103]34 C57BL/6 LY5.1 (CD45.1) mice were purchased from Charles River. Mice were kept in a pathogen-free colony at the animal facility of the Veneto Institute of Molecular Medicine, Padova, Italy. The University of Padova organization in charge for animal wellness approved the experimentation and declared that it fulfilled the National and European rules for animal studies. All the experimental procedures were authorized by the Italian Ministry of Health (Protocol N° 126-2015 and 205/2022-PR). For details on mice breeding, genotyping, and sample processing, see [104]Suppl. Materials and Methods. Repopulation assay In competitive transplantation, CD45.1 mice were used both as recipient and competitor cells. Females, in synchronized gestations, were sacrificed at 14.5dpc; mixtures of equal amounts (1x10^6 cells) of donor (either Vav1^+/Cre control or Csnk2b^fl/fl/Vav1^+/Cre KO from CD45.2^+ genetic background) and wild-type competitor (CD45.1^+) total fetal liver cells were intravenously injected in lethally γ-irradiated (900rad) CD45.1^+ recipient mice. In non-competitive transplantation assay, CD45.1 recipient mice were injected only with (1 × 10^6 cells) CD45.2^+ cells from either Vav1^+/Cre control or Csnk2b^fl/fl/Vav1^+/Cre KO. Engraftment was assessed at 13/14 days for both experimental assays and after 1 month for competitive experiments in PB, BM, Spleen, Thymus, and Lymph Nodes. Repopulation efficiency of donor and competitor populations was investigated through fluorescence activated cell sorting (FACS) analysis after staining of PB or smashed hematopoietic tissues with Fc Block, anti-CD45.1, and anti-CD45.2 antibodies followed by erythrocyte cell lysis. Cell culture and chemicals G1E-ER cells, a kind gift of Prof. M. Weiss (St. Jude Childrens’ Hospital, Memphis, TN) were maintained as described in.^[105]36 Erythropoietin and 1-Thioglycerol were purchased from Sigma (Mo), SCF from Peprotec (UK). For cell treatment, β-estradiol (Merck) was used at 0.2 µM, CX-4945 (Activate Scientific GmbH) at 2.5 µM, Z-VAD-FMK (SelleckChem) caspase inhibitor at 5 µM, proteasome inhibitor bortezomib (SelleckChem) at 5 nM and cycloheximide (Merck) at 0.4 µg/ml. RNA-Seq The sequencing was performed on TER119^+ cells purified from 14.5 dpc fetal livers by FACS sorting. The complete RNA-Seq data are available in the Gene Expression Omnibus (GEO) ([106]http://www.ncbi.nlm.nih.gov/geo) under accession number [107]GSE158665. For the complete list of differently expressed genes see [108]Suppl. Tables S1 and [109]S2. For further details on the analysis methods, see [110]SDC file. Statistical analysis Data were analyzed with the 2-tailed unpaired Student’s t-test. Values were considered statistically significant at P values below *0.05, **0.01, ***0.001. For further Materials and Methods, see the [111]Supplemental Digital Content file. Data availability All data required to produce the results of this study are provided within the article, via supplemental files or with link to the dataset GEO with the accession number. Also included in supplemental file [112]Suppl. Tables S3 - S7 reporting in order: list of primers for genotyping, qRT-PCR primers, antibodies for WB and flow cytometry, hemochrome of adult mice. RESULTS CK2β loss in the hematopoietic compartment is embryonic lethal and alters hematopoietic cell number, viability, and proliferation We generated conditional CK2β-deficient mice in Vav1-expressing cells. In utero lethality between 19.5 and 20.5 days post conception (dpc) was observed in 100% of CK2β KO embryos (Csnk2b^fl/fl;Vav1^+/Cre). Control mice (Csnk2b^+/+;Vav1^+/Cre) and CK2β heterozygous mice (Csnk2b^+/fl;Vav1^+/Cre) were viable and fertile. We therefore analyzed fetuses at 12.5, 14.5, and 17.5 dpc. CK2β KO fetuses were pale and hemorrhagic, in some cases with the appearance of hydrops fetalis, more evident at 17.5 dpc (Figure [113]1A). Analysis of CK2β mRNA (Figure [114]1B) and protein levels (Figure [115]1C) in the fetal liver, primary site of hematopoiesis at this stage,^[116]37 and in the brain from 14.5 dpc fetuses showed complete Csnk2b deletion restricted to the hematopoietic tissue. No significant changes were observed in CK2α protein levels (Figure [117]1C). CK2β-null embryos displayed smaller fetal livers with reduced cellularity as compared to controls (Figure [118]1D). FACS analysis of the CD45^+ (leukocyte) and Ter119^+ (erythroid) markers showed a reduction in the percentage of erythroid cells in the CK2β KO mice, while the percentage of leukocytes appeared slightly increased as compared to control littermates (Figure [119]1E). A deepened analysis of the white cell compartment revealed an accumulation in percentages of granulocytes and monocytes/macrophages in CK2β KO embryos (Figure [120]1F and [121]1G, left panels) that could account for the increased frequency of CD45^+ cells; however, the absolute counts of Mac1^+Gr1^+ and Mac^+F4/80^+ populations in total fetal livers resulted to be reduced in the KO samples compared to the control ones (Figure [122]1F and [123]1G, right panels). Moreover, also the B cell compartment was altered in CK2β KO mice with accumulation in percentages of immature B cells (B220^+ CD19^-) (Figure [124]1H, left panel) and expansion of pre/pro B cells (Figure [125]1I, left panel). Also in this case the counts in the total fetal livers show discrepancies compared to the frequencies due to the reduced global cellularity of livers in KO embryos; indeed, there were no significant changes in B220^+ CD19^- absolute numbers (Figure [126]1H, right panel) and there was a decrease of pre/pro cells in KO embryos (Figure [127]1I, right panel). Annexin V (AnnV) and 7-Aminoactinomycin D (7-AAD) staining revealed a higher percentage of apoptotic cells in CK2β KO fetal livers that became more evident at 17.5 dpc (Figure [128]1J). However, analysis of Ki-67 expression showed a mild increase in cell proliferation in CK2β KO compared to control mice (Figure [129]1K). Figure 1. [130]Figure 1. [131]Open in a new tab KO of CK2β in the fetal hematopoietic system results in embryonic lethality with reduction in fetal liver cellularity and alteration in cell viability and proliferation. (A) Pictures of Csnk2b^fl/fl/Vav1^+/Cre (KO) and Csnk2b^+/+/Vav1^+/Cre (control) littermate fetuses at 14.5 and 17.5 days post conception (dpc). (B) Csnk2b mRNA levels evaluated by qRT-PCR at 14.5 dpc in control and KO fetal livers. Data are representative of 5 samples for each genotype. Gapdh was used as reference gene. (C) WB analysis of CK2β and CK2α protein expression at 14.5 dpc in fetal liver cells compared to non-hematopoietic tissue (brain); upper panel: WB image representative of 4 samples for each genotype; lower panel: densitometric analysis; GAPDH was used as loading control. (D) Upper level: representative image of control and KO fetal livers at 17.5 dpc; lower level: fetal liver cell count at different development time points (12.5 dpc: controls n = 9, KO n = 6; 14.5 dpc controls n = 17, KO n = 18; 17.5 dpc: controls n = 18, KO n = 21). (E) FACS analysis of the white population (CD45^+) and erythroid cells (Ter119^+) in fetal livers at 17.5 dpc: on the left representative contour plot, on the right histograms displaying the percentages of the 2 hematopoietic compartments (controls n = 8; KO n = 3). (F) Scatter plots representing percentages of cells (on the left; controls n = 11, KO n = 3) or absolute counts (on the right; controls n = 10, KO n = 6) after Gr1 and Mac1 staining and FACS analysis in 17.5dpc fetal livers. (G) FACS analysis of macrophages (Mac1^+F4/80^+) in 17.5 dpc fetal livers represented as percentage on the left (controls n = 3, KO n = 3) and as absolute counts on the right (controls n = 9, KO n = 6). (H) Graphs indicating the percentages (on the left) (controls n = 15, KO n = 6) and absolute counts (on the right) (controls n = 10, KO n = 6) of B cells in 17.5dpc fetal livers. (I) FACS analysis of precursors B cells in 17.5 dpc fetal livers: graphs indicating the percentages (on the left) and absolute counts (on the right) (controls n = 8, KO n = 3). (J) Evaluation of apoptosis at 14.5 and 17.5 dpc by FACS analysis, after staining with Annexin V (AnnV) and 7-AAD, performed on total liver population (at 14.5 dpc controls n = 4, KO n = 5; at 17.5 dpc controls n = 20, KO n = 6). (K) Analysis of Ki-67 expression in 14.5dpc total liver cells (controls n = 5, KO n = 3). In (C–K), graphs show means ± SD; Student’s t-test: *P < 0.05; **P < 0.01; ***P < 0.001. FACS = fluorescence activated cell sorting; KO = knockout; WB = western blot. Deletion of CK2β affects hematopoietic stem and progenitor cell proliferation kinetics and transcriptional programs Next, we analyzed the immature hematopoietic compartment represented by stem (HSC) and progenitor cells (HPCs) evaluating the effects of Csnk2b ablation. FACS analysis of HSCs^[132]38 and HPCs^[133]39 revealed a significant increase in the percentages of HSCs (Lin^- cKit^+ Sca-1^+ CD150^+ CD48^-) (Figure [134]2A and [135]2D), MEPS (Lin^- cKit^+ Sca-1^- CD34^- FcyRIII/II^-), CMPs (Lin^- cKit^+ Sca-1^- CD34^+ FcyRIII/II^low) and GMPs (Lin^- cKit^+ Sca-1^- CD34^+ FcyRIII/II^high) (Figure [136]2B and [137]2D) in KO fetal livers compared to controls with no significant changes in CLPs (Lin^- IL-7R^+ cKit^+ Sca-1^-/low) (Figure [138]2C and [139]2D). However, analysis of absolute counts in the total fetal livers confirmed in the KO samples a significant expansion limited to the LSK compartment and a trend to higher counts for GMPs (Figure [140]2E). Notably, the intensity of cKit expression, a receptor essential for HSC and precursor survival and self-renewal, was found substantially reduced (mean fluorescence intensity, MFI) in CK2β KO HSPCs (Figure [141]2F). Figure 2. [142]Figure 2. [143]Open in a new tab Absence of CK2β increases the amount of hematopoietic stem cells and precursors and perturbs their maturation program. Representative FACS dot plot of hematopoietic stem cells (HSCs) (A), common myeloid progenitors (CMPs), megakaryocyte/erythrocyte progenitors (MEPs), granulocyte/monocyte progenitors (GMPs) in 14.5 dpc fetal livers (B), common lymphoid progenitors (CLPs) (C); the representative dot plots show the percentage of cells; 1x10^6 and at least 4 × 10^5 events were acquired for LT-HSCs and the other progenitors respectively. (D) Histogram summarizing the percentages of each population (for HSC controls n = 5, KO n = 4; for the other precursors: controls n = 9, KO n = 6). (E) Graphs representing absolute cell counts of LSK, MEPs, CMPs, and GMPs populations referred to the total fetal liver (controls n = 3, KO n = 3). (F) Histograms exhibiting mean fluorescence intensity (MFI) in HSCs and each precursor sub-population (controls n = 9, KO n = 6). (G) BrdU proliferation assay in 14.5 dpc fetal livers cells: cell cycle was analyzed inside the LSK (Lin^-Sca-1^+cKit^+) fraction after staining with anti-BrdU and 7-AAD. Left panel: representative contour plots and gating strategy; right panel: histograms summarizing the percentages of the cells in each stage (controls n = 4, KO n = 4). (H) qRT-PCR showing the relative mRNA expression of Scl/Tal1, Bmi-1, Runx1, Jun, Pu.1, Cebpa, Gata1, Gata2 genes in HSCs (controls n = 3, KO n = 3), CMPs (controls n = 4, KO n = 5), GMPs (controls n = 4, KO n = 5) and MEPs (controls n = 4, KO n = 5) purified through sorting from 14.5dpc fetal livers. Gapdh was used as reference gene. In all panels (D–H) the graphs report mean ± SD values; Student’s t-test: *P < 0.05; **P < 0.01; ***P < 0.001. CLPs = common lymphoid progenitors; CMPs = common myeloid progenitors; FACS = fluorescence activated cell sorting; GMPs = granulocyte/monocyte progenitors; HSCs = hematopoietic stem cells; KO = knockout; LSK = Lin-Kit+Sca1+; MEPs = megakaryocyte/erythrocyte progenitors. Since during blood cell development proliferation and differentiation must be tightly coordinated, we analyzed the cell cycle of LSK cells, using BrdU and 7-AAD staining and FACS analysis. As shown in Figure [144]2G, a higher percentage of cells was in G2/M, S, and sub-G[0] phases and displayed a greater DNA content (gate >2n) in KO mice compared to controls. We next inquired if CK2β loss could affect the expression of transcription factors important for HSC and HPC early lineage commitment. By qRT-PCR we evaluated the expression of specific mRNAs coding for transcription factors involved in cell maturation (Gata1,^[145]5,[146]40 Jun^[147]41) or in the maintenance of stem cell quiescence and self-renewal (Scl/Tal1,^[148]42 Bmi-1,^[149]43 Runx1,^[150]44,[151]45 Pu.1,^[152]46 Cebpa,^[153]47 Gata2^[154]48), in FACS-sorted LSKs, CMPs, GMPs and MEPs (Figure [155]2H). In CK2β KO embryos, the LSK compartment exhibited a reduced expression of Scl/Tal1 and Pu.1 with no significant changes in Bmi-1 and Runx1 mRNA levels compared to controls. Loss of CK2β caused a decrease of Runx.1, Pu.1 and Cepba expression in CMPs and a decrease of Cebpa and Gata1 in GMPs. MEPs from KO mice showed alterations in almost all the genes analyzed with a significant reduction of Scl/Tal1, Bmi-1, Runx1, and Gata1 and an increase of Jun expression. In summary, lack of CK2β causes an accumulation and increased proliferation of HSCs and an altered expression of transcription factors involved in the regulation of HSPC quiescence and commitment. CK2β is essential for hematopoietic reconstitution capability in vitro and in vivo To assess the differentiative potential of progenitors, we performed methylcellulose-based colony-forming assays of fetal liver HSPCs. Strikingly, KO cells displayed an impairment to generate erythroid and myeloid colonies at 3 and 6 days ([156]Suppl. Figures S1A and S1B). In vivo competitive repopulation assays of 14.5 dpc fetal liver cell transplanted in C57BL/6 congenic mice expressing the differential leukocyte marker CD45.1 were then performed (Figure [157]3A).^[158]49,[159]50 As summarized in Table [160]1, an almost complete restoration of hematopoiesis was obtained as early as 2 weeks after transplantation with comparable blood cell percentages between mice injected with both kinds of cell mix. To note, FACS analysis of peripheral blood revealed an equal contribution to hematopoiesis when control CD45.2^+ and competitor CD45.1^+ donor cells were injected, while the injection of CK2β KO CD45.2^+ did not contribute to mature blood cells (Figure [161]3B). The same phenotype was observed after 4 weeks in all the hemo-lymphopoietic organs (Figure [162]3B–[163]3D). We also evaluated the contribution to the generation of LSK and HSCs from CD45.1^+ and CD45.2^+ cell populations. Remarkably, in recipient mice injected with the competitive KO transplant there was a complete absence of CD45.2 expression both in LSK (Figure [164]3E) and in HSCs (CD48^-CD150^+ in LSK gated cells)^[165]51 (Figure [166]3F). Since the lack of engraftment in the BM could be linked to a problem of homing of HSCs, we evaluated by FACS the expression of CXCR4 (a known key mediator of HSC homing to the bone marrow^[167]52) in LSK compartment of control and CK2β KO fetal livers (Figure [168]3G). Unexpectedly, CK2β KO embryos showed higher percentages of CXCR4^+ LSK cells and higher MFI of this marker as compared to control counterpart, suggesting that the observed phenotype is likely not dependent on this molecule. To exclude the possibility that the lack of KO cell engraftment was due to the inability of CD45.2^+ CK2β null LSK to compete with CD45.1 cells for the BM niches, we also performed non-competitive transplantation assays. As described in [169]Suppl. Figure S1C, CD45.1 adult mice were irradiated and injected with only CD45.2 cells from control or KO fetal livers; 2 out of 5 mice injected with CK2β null CD45.2^+ cells died at day 12, thus we anticipated the experimental end point at 13 days. In all the compartments analyzed (SP, BM, and spleen) we evidenced almost the complete absence of CD45.2 cells from KO samples ([170]Suppl. Figure S1D). Moreover, analysis of CD45.2^+ LSK frequencies and events in the BM ([171]Suppl. Figure S1E) underlined that CK2β KO progenitors and stem cells were unable to engraft and sustain hematopoiesis. Figure 3. [172]Figure 3. [173]Open in a new tab Fetal liver cell transplantation from KO fetuses in recipient adult mice results in the lack of hematopoietic reconstitution. (A) Transplant workflow: a mixed population with equal proportions of CD45.2 (donor from control or KO) and CD45.1 (competitor) fetal liver cells were injected in recipient CD45.1 adult mice after lethal total body irradiation (900 rads). The transplantation with CD45.2 cells from control embryos plus CD45.1 competitor cells is indicated as Mix Control; when CD45.2 cells were obtained from CK2β-null embryos plus CD45.1 competitor cells, the injected population is called Mix KO. Hematopoietic restoration was assessed at 2 and 4 weeks from irradiation. Injection of PBS was used as control of irradiation efficacy. Sample size: 7 mice injected with mix control, 7 with mix KO, and 2 with PBS. (B–F) Flow cytometry analysis of CD45.2 and CD45.1 relative amounts in (B) peripheral blood (PB), (C) bone marrow (BM), spleen (SP), and (D) lymph nodes and thymus, (E) inside the gate of LSK and (F) inside long term HSCs. On the left of each figure are depicted the representative dot plots and on the right are the histograms with CD45.1 and CD45.2; (G) Flow cytometry analysis of CXCR4 expression in 14.5 dpc fetal livers; upper panel: representative dot plots; lower panel: histograms representing the percentages and mean fluorescence intensities (controls n = 5, KO n = 3). Data in the graphs (B–G) are shown as mean ± SD, Student’s t-test: *P < 0.05, **P < 0.01, ***P < 0.001 comparing donor CD45.2 vs competitor CD45.1; °°P < 0.01, °°°P < 0.001 comparing CD45.1 population from KO mix vs that one of control mix; #P < 0.05, ##P < 0.01, ###P < 0.001 comparing CD45.2 population from KO mix vs that one of control mix. BM = bone marrow; HSCs = hematopoietic stem cells; KO = knockout; LSK = Lin-Kit+Sca1+; PB = peripheral blood; PBS = phosphate buffered saline; SP = spleen. Table 1. Hemochrome of Transplanted Mice Parameters 2 wks 4 wks CTRL KO CTRL KO WBC (10^3/µL) 2.87 ± 0.70 2.60 ± 0.35 9.83 ± 1.53 7.17 ± 1.24[174]^** RBC (10^6/µL) 10.12 ± 0.32 8.11 ± 1.73 12.00 ± 1.87 10.80 ± 0.67 HGB (g/dL) 15.00 ± 0.53 11.80 ± 3.12 15.87 ± 2.13 14.73 ± 0.86 HCT (%) 42.33 ± 1.62 34.67 ± 9.27 49.30 ± 7.75 44.77 ± 2.78 MCV (fL) 41.90 ± 0.92 42.40 ± 3.00 41.05 ± 0.28 41.43 ± 0.29[175]^* PLT (10^3/µL) 1062.00 ± 419.24 650.00 ± 68.79 1842.67 ± 152.39 1764.00 ± 151.61 MPV (fL) 7.00 ± 0.66 8.90 ± 1.10 5.07 ± 0.38 5.07 ± 0.38 LYM% 51.73 ± 8.35 61.03 ± 3.65 75.80 ± 5.36 70.85 ± 5.59 MID% 10.07 ± 1.79 6.73 ± 1.94 9.55 ± 0.94 12.27 ± 2.37[176]^* GRA% 38.20 ± 6.56 32.23 ± 2.40 14.65 ± 5.99 16.88 ± 3.46 [177]Open in a new tab ^* P < 0.05; ^** P < 0.01 compared to mice injected with cells from control embryos. CTRL = controls; GRA = granulocytes; HCT = hematocrit; HGB = hemoglobin; KO = knockout; LYM = lymphocytes; MCV = mean corpuscolar volume; MID = monocytes; MPV = mean platelet volume; PLT = platelets; RBC = red blood cell; WBC = white blood cell. Erythropoiesis is defective in CK2β KO mice Deletion of CK2β in purified Ter119^+ mature erythroid cells was complete (Figure [178]4A). The levels of the catalytic α subunit were found decreased and a CK2-specific kinase assay showed a 60% residual enzymatic activity (Figure [179]4A), likely attributable to the remaining α dimers. FACS analysis of CD71 and Ter119 markers in fetal livers at 14.5 and 17.5 dpc^[180]53,[181]54 revealed that CK2β KO embryos had unbalanced frequencies of erythroid subfractions compared to control mice (Figure [182]4B). KO samples displayed accumulation of both CD71^low/- Ter119^- and CD71^high Ter119^- immature populations and, particularly at 17.5 dpc, depletion of the most mature Ter119^+ CD71^- fraction (Figure [183]4B, left and middle panels). Analysis on pellets of peripheral blood obtained from 17.5 dpc embryos confirmed a reduced amount of mature hemoglobinized cells in CK2β KO samples (Figure [184]4B). Furthermore, KO fetal liver cells presented a reduced expression of both Ter119 and CD71 (Figure [185]4B, right panels). Similarly to the HSPCs, the MFI of cKit expression was found reduced in CK2β-deficient CD71^+Ter119^-/low population mainly composed of CFU-E, BFU-E and pro-Erythroblasts (Figure [186]4C). Complete blood cell count analysis performed on peripheral blood of 17.5 dpc embryos pointed out a strong reduction of RBC counts in KO samples (Figure [187]4D). To further support the evidence of terminal erythroid maturation impairment, since the extrusion of the nucleus is one central step of erythrocyte production, we evaluated the amount of nucleated and anucleated erythroid cells within the gate of Ter119^+ FSC^low cells.^[188]55,[189]56 The nuclear staining through Syto-16 tracker evidenced a higher percentage of nucleated cells (Syto-16^+) with parallel reduction of enucleated cells (Syto-16^-) in KO samples compared to controls (Figure [190]4E). Cytospin preparations of Ter119^+ fetal liver cells (Figure [191]4F, upper panel) revealed dysplastic features with membrane irregularities and blebs (blue arrows) of immature erythroblasts in KO mice. Reticulocytes looked irregularly shaped with intracellular inclusions resembling in size and color (but not in number) the Howell-Jolly bodies (red arrows). Also, peripheral blood smears (Figure [192]4F, bottom panel) showed larger erythrocytes, which in some instances appeared swollen and vacuolized. Since these morphological alterations could rely on defects of red cell membrane properties, such as the ability to change shape under the effect of mechanical forces, we evaluated erythrocyte deformability capacity in 17.5 dpc embryos by means of an ektacytometer. As shown in Figure [193]4G, erythrocytes from peripheral blood of KO animals presented a significant reduction in the elongation index, thus less deformability, when they were exposed to a range of applied stress from 0.3 to 30 Pa. Figure 4. [194]Figure 4. [195]Open in a new tab Ablation of Csnk2b in the hematopoietic compartment causes ineffective erythropoiesis with red blood cell morphological alteration, impairment of cell viability, and unbalance in cell proliferation. (A) Top: WB analysis of CK2β and CK2α expression; bottom: CK2 kinase activity displayed as count per minute (CMP) in Ter119^+ cells from 17.5 dpc fetal livers. (B) Study of erythroid maturation stages by FACS at 14.5 and 17.5 dpc, analyzing transferrin receptor (CD71) and glycophorin-associated protein Ter119. In detail: (B) left panel: representative dot plot with the gating strategy and sub-populations 1 (Ter119^-CD71^-/low), 2 (Ter119^-CD71^high), 3 (Ter119^+CD71^high) 4 (Ter119^+CD71^-/low) and representative pellets of peripheral blood samples from 17.5dpc embryos; middle panel: histograms summarizing percentages of each erythroid sub-population; right panel: peaks and graphs showing Ter119 and CD71 expression represented as mean intensity fluorescence (MFI). Sample size: n = 19 controls and 9 KO at 14.5 dpc; n = 20 controls and 6 KO at 17.5 dpc. (C) MFI of cKIT in the gate of Ter119^-/low CD71^+ cells (14.5dpc fetal livers; n = 16 controls and n = 7 KO). (D) Red blood cell counts (RBC) in the peripheral blood of 17.5 dpc embryos (n = 7 controls, n = 3 KO). (E) Evaluation of nucleated and enucleated erythroid cells through Syto-16 staining. Left, contour, and dot plots showing the gating strategy; right, graphs summarizing the percentages of nucleated (Syto-16^+) and enucleated (Syto-16^-) cells in control and KO erythroid cells. Sample size: n = 3 controls and n = 3 KO. (F) Upper panel: cytospin of Ter119^+ cells purified from fetal livers; lower panel peripheral blood smears from 17.5 dpc fetuses. Samples stained with May-Grunwald Giemsa. Red arrows indicate cells with membrane blebs, and blue arrows indicate abnormal reticulocytes. Sample size: data are representative of 4 controls and 4 KO, at least 3 images were acquired for each sample. (G) Histogram representing the deformability of red blood cells in 17.5 embryos; test performed using Lorca ektacytometer on whole peripheral blood. Rigidity and plasticity are evaluated as Elongation Index (EI); deformability was assessed over a range of shear stresses between 0.3 and 30 Pa. Sample size: data are representative of 14 controls and 6 KO. Results were expressed as means ± SEM. Multiple t-tests were corrected with Holm-Sidak method: *P < 0.05, **P < 0.01, ***P < 0.001. (H) Evaluation of apoptosis at 14.5 and 17.5 dpc by flow cytometry analysis, after staining with Annexin V (AnnV) and 7-AAD; analysis was performed on erythroid sub-populations (CD71^+Ter119^-) and (Ter119^+); top panel: representative dot plot, bottom panel: histograms showing cell percentages. Sample size: controls n = 8, KO n = 4 at 14.5 dpc; controls n = 9, KO n = 4 at 17.5 dpc. (I) WB of CASPASE-3, PARP, the anti-apoptotic BCL-xL, and the P53 downstream target P21 levels. Sample size: n = 4 controls and 4 KO at 14.5 dpc; 12 controls and 13 KO at 17.5 dpc). For BAX, MCL-1, and PUMA the sample size was composed of n = 3 controls and 3 KO at 14.5 dpc; n = 6 controls and 5 KO at 17.5 dpc. (J) Analysis of cell cycle stages in erythroid sub-populations after anti-BrdU and 7-AAD staining combined with Ter119/CD71 markers. Sample size: n = 6 controls and 5 KO. In (A–E) and (H–J), graphs report means ± SD. In (A) and (I) densitometric analysis, data were normalized over GAPDH and over the mean value of controls. Student’s t test: *P < 0.05, **P < 0.01, ***P < 0.001. EI = elongation index; FACS = fluorescence activated cell sorting; KO = knockout; RBC = red blood cell counts; WB = western blot. Both fetal and adult CK2β heterozygous (het) mice did not suffer an anemic condition: for detailed description of their erythroid phenotype, see [196]Suppl. Figure S2, S Results and [197]Suppl. Table S7 that reports the hemochrome values. Thus, loss of CK2β results in abnormal erythroid maturation, with reduction of terminally differentiated cells. CK2β regulates erythroid cell survival and proliferation by impinging on multiple intracellular pathways To understand the mechanism underlying deficient erythroid maturation, we evaluated apoptosis by AnnV and 7-AAD staining (Figure [198]4H). The mature Ter119^+ population from KO mice presented, as early as 14.5 dpc and to a higher extent at 17.5 dpc, an increase of cells in late-stage apoptosis (AnnV^+/7-AAD^+) and necrosis (AnnV^-/7-AAD^+) as compared to controls. On the contrary, the immature CD71^+Ter119^- fraction did not present significant alterations in cell viability. WB analysis of Ter119^+ purified cells (Figure [199]4I) revealed in CK2β-deficient cells a significant decrease of PRO-CASPASE-3 with accumulation of its active cleaved form, an increase of PARP cleavage and a reduction of BCL-xL, a BCL-2 family member pivotal for the survival of late stages erythroblasts. Investigation of other BCL-2 family members highlighted an up-regulation of the pro-apoptotic proteins BAX and PUMA in KO samples at both 14.5 and 17.5 dpc and the same trend was also observed for the anti-apoptotic factor MCL-1 that is reported be essential in immature erythroblasts and erythroid progenitors.^[200]57 Moreover, 14.5 and 17.5 dpc fetal liver KO cells expressed markedly higher levels of the TP53 downstream target P21 (Figure [201]4I). To assess the proliferation kinetics, we performed an in vivo BrdU incorporation assay. FACS analysis showed a reduction of CD71^+ Ter119^- immature cells in the S phase and an increase of Ter119^+ mature cells in the subG[0]/G[1] phase without significant changes in S and G[2]/M phases, in KO mice compared to CTRL (Figure [202]4J). Next, we carried out RNA-Sequencing in Ter119^+ fetal liver cells. As many as 1143 differentially expressed genes were identified in CK2β-null mice as compared to controls (See [203]Suppl. Tables S1 and S2 for the complete list of up and down-modulated genes respectively). Pathway enrichment analysis revealed in KO samples (Figure [204]5A) an up-regulation of some P53-targets, such as Cdkn1a (P21), Zmat, and Gadd45, a deregulation of genes associated with the JAK/STAT pathway, in particular a down-modulation of Stat5, essential for erythroid cell viability,^[205]58 and overexpression of Socs2, coding for an inhibitor of this signaling cascade.^[206]59 Moreover, we observed a down-regulation of Kit and of genes associated to the PI3K/AKT pathway such as Pi3kr1 and Gab1. Interestingly, there was the up-regulation of Jun, co-factor of PU.1, known to inhibit erythroid differentiation counteracting the transcriptional capability of GATA1, the master regulator of terminal erythroid maturation.^[207]5 Another dysregulated gene category was the group of solute carrier transporters (Slc) implicated in ion homeostasis.^[208]60 Intriguingly, mRNA expression of Siglec1, Vcam1, and Selplg were decreased in KO samples. These are markers expressed by macrophages of the erythroblastic islands^[209]61 and it was described in a recent work that a contamination of these cells is frequently observed in purified Ter119 fraction because of the tight connection between erythroid and macrophage populations.^[210]62 We therefore validated by qRT-PCR the expression of some of the deregulated genes identified by RNA-Seq (genes highlighted in yellow). Analysis was performed at 17.5 dpc (Figure [211]5B) and 14.5 dpc ([212]Suppl. Figure S3A) on Ter119^+ fetal liver cells. A complementary analysis obtained interrogating RNA-seq data with Gene Ontology terms of our interest (see [213]Supplemental material for details on the analysis) showed in KO samples the significant deregulation of other 114 genes ([214]Suppl. Figure S3B) involved in erythroid cell membrane stability (Ebp4.1-pink highlighted), water homeostasis (Aqp1-green highlighted^[215]63), heme biosynthesis (Tmem14c-violet highlighted), and in particular genes related to the mitotic process (blue highlighted genes). Furthermore, we evaluated the deregulation in expression of genes coding for kinases, confirming the down-regulation of Csnk2b in KO samples ([216]Suppl. Figure S3C, top), and of genes coding for transcription factors, showing a reduction in KO cells of Ikzf1 (Ikaros1) and Klf13, both involved in erythroid maturation ([217]Suppl. Figure S3C, bottom).^[218]64,[219]65 Figure 5. [220]Figure 5. [221]Open in a new tab Lack of CK2β affects erythroid-associated signaling pathways. (A) RNA-Seq analysis on RNA extracted from sorted Ter119^+ control and KO fetal liver cells (14.5 dpc): heat maps of the genes significantly up or down-modulated after pathway enrichment analysis. Values are quantified by the scale bar that visualizes differences in the z score (expression difference/SD) relative to the mean. (B–G) Images were produced using Ter119^+ cells purified with beads from fetal livers. In detail: (B) qRT-PCR performed to validate the different expression of some genes of the heat maps (highlighted in yellow) (cKit, Cdkn1a, Jun) (n = 4 for control and KO samples). (C) Elisa test, evaluation of EPO plasma levels in 17.5 dpc embryos (controls n = 5, KO n = 3). (D) WB displaying the expression of EPO-R at 17.5 dpc; top level: representative dot plot; bottom: densitometric analysis (controls n = 3, KO n = 3); (E) WB showing the expression and activation of molecules downstream EPO-R: left panel: representative images from 14.5 and 17.5 dpc; right panel: densitometric values of 17.5 dpc sample. For total proteins, the graphs report densitometric signal normalized over GAPDH and then over the mean of values in control samples. For the phosphorylated forms, densitometric analysis reports the values as fraction of the phospho over the corresponding total protein, followed by normalization over the mean values of control samples. (F) Assessment of GATA1 transcriptional capability analyzing the expression of its targets by qRT-PCR and on other key genes involved in erythroid differentiation (n = 4 for control and KO samples) and (G) WB. (H) Evaluation by WB of ERK and AKT activation on sorted CD71^+Ter119^low/- populations. On the left, representative dot plot with the gating strategy for sorting; in the middle, representative WB of AKT, ERK, CK2α and CK2β expression after stimulation with SCF for 15’; on the right, graphs representing AKT and ERK activating phosphorylation: densitometric values are reported as phospho/total protein levels normalized on the mean of controls. Samples size: n = 4 pools of control samples, each one composed of 3 fetal livers, and n = 3 pools of KO, each one composed of 2 fetal livers. In (F), Gapdh was chosen as reference gene, (G) and (H) was used as loading control. In the graphs (B–H), data are shown as mean ± SD; statistical significance was analyzed by Student’s t test: *P < 0.05, **P < 0.01, ***P < 0.001. AKT = serine-threonine kinase; EPO = erythropoietin; EPO-R = erythropoietin-receptor; ERK = extracellular signal-regulated kinase; KO = knockout; WB = western blot. CK2β-deficient erythroid cells show deregulated cell signaling and GATA1 activity Since EPO-R is essential in driving erythroid maturation from pro-erythroblast to late stages, we investigated the signaling upstream and downstream of this receptor. ELISA performed on plasma obtained from 17.5 dpc embryos showed a trend for lower EPO levels in CK2β KO samples (Figure [222]5C). EPO-R protein levels on Ter119^+ purified cells resulted unaltered in KO mice compared to controls (Figure [223]5D). Next, we analyzed downstream JAK/STAT, ERK, PTEN, and PI3K/AKT signaling cascades,^[224]4 this latter culminating in the AKT-directed phosphorylation of GATA1 on Ser310 that empowers its transcriptional activity.^[225]66 Immunoblot analysis on Ter119^+ fetal liver cells (Figure [226]5E) revealed markedly decreased levels of total AKT and GATA1 at both 14.5 and 17.5 dpc and of STAT5, ERK1/2 (p44-42) at 17.5 dpc. However, their phosphorylation at CK2-independent or indirectly regulated (ie, AKT S473) sites was not significantly affected when the quantification was normalized against the total protein level, as opposed to the phosphorylation of residue AKT S129^[227]67 (direct CK2 target) which is clearly reduced in KO cells. PTEN showed a significant decrease of the inhibitory phosphorylation on Ser380/Thr382/383, both at 14.5 and 17.5 dpc and a depletion in total protein levels at later time points in CK2β KO mature erythroid cells. Moreover, KO samples displayed a higher phosphorylation, thus activation, of SAPK/JNK kinases, which are involved in stress signaling and in the activation of the transcription factor c-JUN ([228]Suppl. Figure S3D). Consistently, KO mice exhibited reduced mRNA levels of the GATA1 target genes β-globin,^[229]8 Lrf/Zbtb7a^7, and Klf^[230]5 transcription factors and of Alas2,^[231]9 which encodes for 5’-aminolevulinate synthase 2, the enzyme catalyzing the first reaction in heme biosynthesis (Figures [232]5F and [233]S3E). As a further expected consequence of GATA1 down-modulation, we observed the up-regulation of the ε embryonic (β-like) globin chain and of Gata2 transcription factor (Figures [234]5F and [235]S3E). Validation of these findings was also performed at the protein level for ALAS2 and LRF (Figure [236]5G). Moreover, the low levels of GATA1 could also explain the deregulated expression of Slc genes observed in the RNA-seq analysis since they are reported to be controlled by this transcription factor.^[237]60,[238]63 Importantly, in KO samples also FOG1, a key co-factor of GATA1 essential for both erythropoiesis and megakaryopoiesis,^[239]63,[240]68 was down-regulated (Figure [241]5G). Additionally, Prussian blue staining on fetal liver sections clearly showed the presence of reactive ferric iron deposits in CK2β-null mice, likely due to Alas2 deficiency^[242]69 ([243]Suppl. Figure S3F). cKit receptor is known to sustain immature erythroid cells, such as BFU-E, and to cooperate with EPO-R in maintaining CFU-E and pro-erythroblasts. Since we demonstrated that cKit is down-regulated also in these populations (included in Ter119^-/lowCD71^+ gate) (Figure [244]4C), we asked whether erythroid linage impairment could be partly due to a less effective cKit signaling. CD71^+Ter119^-/low fraction was isolated through sorting from 14.5 dpc pools of control or KO fetal livers (Figure [245]5H left panel, gating strategy). Then, cells were treated with SCF for 15 minutes to evaluate the activation of 2 key effectors downstream Kit, that is, ERK and AKT. WB analysis revealed a severe reduction of both ERK and AKT phosphorylation in KO samples compared to controls (Figure [246]5H, middle and right panels). Altogether, these results clearly suggest that CK2 regulates erythropoiesis by controlling signaling pathways downstream EPO-R, and in particular regulating GATA1 levels and transcriptional proficiency, as well as the modulation of cKit expression and its downstream targets. CK2 regulates GATA1 expression, localization, and turnover during erythroid differentiation To understand the CK2-mediated mechanism of signaling and GATA1 regulation, we used the G1E-ER cell line, a model of β-estradiol-inducible GATA1-dependent erythroid differentiation.^[247]36 G1E-ER cells were cultured with or without β-estradiol in the presence of CX-4945 (silmitasertib), a selective inhibitor of CK2 kinase activity.^[248]70 Exposure to β-estradiol led to erythroid differentiation as indicated by the appearance of Ter119^+ CD71^+ cells (Figure [249]6A). G1E-ER cells can differentiate up to the orthochromatic erythroblast stage at most and after 72 hours of culture with β-estradiol they undergo apoptosis.^[250]6,[251]71–[252]73 To note, the addition of silmitasertib caused a significant reduction of Ter119^+ cells and a decrease of CD71 expression. Importantly, cell viability remained comparable in treated and untreated samples up to 24 hours ([253]Suppl. Figure S4A), while at 48 hours, only in the condition where silmitasertib was used as single treatment, cell viability was slightly reduced. Therefore, apoptosis was unlikely the cause of the reduction of differentiated cells in the combined treatment. Tetramethylbenzidine test ([254]Suppl. Figure S4B) performed at 48 hours indicated a decreased content of hemoglobin in the combined treatment confirming the impairment of erythroid maturation. The same analysis was performed upon Csnk2b or Csnk2a knockdown by siRNA (Figure [255]6B). The silencing of both subunits, when combined with β-estradiol, produced a similar impairment of erythroid differentiation, while the decrease of CD71 MFI was observed also in the absence of β-estradiol (Figure [256]6B). AnnV analysis did not highlight significant changes in cell viability except for the condition of Csnk2a siRNA without β-estradiol, in which a mild increase in apoptosis was seen ([257]Suppl. Figure S4C). Figure 6. [258]Figure 6. [259]Open in a new tab CK2 inhibition and Csnk2b knockdown impair erythroid differentiation of G1E-ER cell line through down-modulation of STAT5, GATA1, and FOG1 molecules. G1E-ER cells were induced to differentiate with β-estradiol (0.2 µM) in presence or absence of sub-apoptotic concentration of silmitasertib (CX-4945 2.5 µM). Cells were collected at 24 and 48 hours from transfection. For Csnk2b and Csnk2a down-modulation, cells were electroporated with 300pmol of siRNA or scrambled oligos, 24 hours after cells were treated with β-estradiol (0.2 µM) and collected at 48 and 72 hours. (A) Evaluation of G1E-ER differentiation upon exposure to β-estradiol and CX-4945 or (B) after Csnk2b or Csnk2a gene silencing. In both (A) and (B), on the left, representative dot plots after staining with AnnV and anti-CD71/Ter119 antibodies. The represented percentages of differentiated cells (Ter119^+ CD71^+) refer to the gate of viable cells; on the right histograms show the percentages of differentiated cells and CD71 MFI. (C) and (D) Representative WB of expression and activation of molecules downstream EPO-R upon exposure to β-estradiol and CX-4945 for 24h or (D) Csnk2b silencing for 48 hours (n = at least 6 independent experiments). (E) Gata1 mRNA levels and expression of its target genes after CK2 inhibition or (F) Csnk2b down-modulation. GAPDH was used as loading control. (G) Immunofluorescence using anti-GATA1 antibody after treatment with CX-4945 (top panel) or Csnk2b siRNA (bottom panel). In red GATA1, in blue DAPI. Images were collected by confocal microscope, acquiring 10 sections along the z-stack for a total thickness of 10 µm; images are shown as maximum projection. Scale bar: 10 µm. Green squares represent higher magnification of a single cell. (H) Graphs representing GATA1 mean fluorescence intensity values of IF experiments performed in G1E-ER cells after CK2 inhibition (top panel) or Csnk2b siRNA (bottom panel); quantification was performed with ImageJ subtracting background signal, at least 3 images for each treatment were analyzed; each dot represents a cell. In (A), (B), and (E–H), graphs represent the mean values ± SD. Statistical significance was analyzed by Student’s t-test: *P < 0.05, **P < 0.01, ***P < 0.001; *significant data compared to untreated samples; # significant data compared to β-estradiol alone; ° significant data compared to CX-4945 alone. EPO-R = erythropoietin-receptor; WB = western blot. WB analysis showed that silmitasertib used alone or with β-estradiol did not cause any significant change in EPO-R levels, whilst the combined treatments produced a significant decrease of STAT5 and GATA1 total protein levels and a trend towards reduction in FOG1, thus mirroring the changes detected in fetal liver KO erythroid cells, except for AKT and ERK levels that remained unaffected (Figures [260]6C and [261]S4E). Silmitasertib also caused a decrease of PTEN inhibitory phosphorylation (S380/T382/383) and of CK2α-dependent phosphorylation of AKT on S129 but not of phospho-GATA1 S301, phospho-AKT S473 and phospho-ERK T202/Y204 when the phosphorylated forms were weighted on the total protein levels (Figures [262]6C and [263]S4E). In the experiments of Csnk2b and Csnk2a silencing we confirmed the effective down-modulation of both subunits through WB analysis (Figures [264]6D and [265]S4F); to note, it was already reported that the down-modulation of the catalytic subunit led also to a decrease of the regulatory protein^[266]22 ([267]Suppl. Figure S4D). Csnk2b silencing, even when combined with β-estradiol, did not impact on EPO-R expression, however, it caused a decline in GATA1, FOG1, STAT5, and PTEN total protein levels without significant changes in their phosphorylation sites (Figures [268]6D and [269]S4F). Unlike Csnk2b down-modulation, Csnk2a silencing, when combined with β-estradiol, led only to reduction of GATA1 expression leaving unchanged FOG1 levels ([270]Suppl. Figure S4D). Analyzing some GATA1 transcriptional targets, we discovered that silmitasertib in combination with β-estradiol, caused a drop in the mRNA expression of β-Globin, Alas2, and Lrf/Zbtb7a as well as of Gata1 itself (Figure [271]6E), while Csnk2b siRNA produced effects mostly on Alas2 gene (Figure [272]6F). IF showed that GATA1-ER protein accumulated exclusively in a nuclear-speckled pattern upon differentiation. Strikingly, CK2 inhibition or CK2β down-modulation, especially when combined with β-estradiol, produced a remarkable reduction in nuclear GATA1 and a redistribution in more diffuse puncta (Figure [273]6G). We quantified GATA1 immunofluorescence signal confirming WB results for both CK2 inhibition and Csnk2b silencing (Figure [274]6H). To test if GATA1 protein was degraded through the proteasome, we performed Csnk2b silencing in the presence of β-estradiol and the proteasome inhibitor bortezomib (Bz). Remarkably, WB analyses showed that Bz was effective in recovering GATA1 levels, especially upon Csnk2b siRNA in combination with β-estradiol ([275]Suppl. Figure S4G). Since GATA1 degradation by proteasome is preceded by its inactivation through caspase cleavage,^[276]74 we evaluated whether the blockade of caspases could produce similar results in GATA1 protein level restoration. As expected, the effects produced by the pan-caspase inhibitor Z-Vad-FMK^[277]75 and silmitasertib (data not shown) instead of Csnk2b siRNA ([278]Suppl. Figure S4H and S4I) were comparable to the previous ones obtained with Bz. The blockade of protein synthesis with cycloheximide in combination with β-estradiol and silmitasertib produced a higher decrease of GATA1 levels compared to cycloheximide and β-estradiol combo, indicating a faster turnover of this protein when CK2 is inhibited ([279]Suppl. Figure S4J). Summarizing, these data demonstrate that CK2 regulates GATA1 turnover, intracellular distribution, and transcriptional activity during erythroid differentiation. CK2 regulates HSP70-GATA1 association The chaperone HSP70 binds and protects GATA1 in the nucleus from the premature cleavage by caspase-3 in erythroid precursors.^[280]76 Therefore, we evaluated HSP70 protein levels in Ter119^+ cells purified from fetal livers of KO and control mice. WB showed a trend in decrease of HSP70 expression in 14.5 dpc KO samples that became significant at 17.5 dpc (Figure [281]7A). A similar down-modulation of HSP70 protein levels was observed in G1E-ER cell line upon Csnk2b silencing plus β-estradiol (Figure [282]7B). However, the reduction of HSP70 was only at the protein level; indeed, we detected an opposite trend analyzing Hsp70 mRNA expression in CK2β KO embryos or after its down-modulation in G1E-ER cells ([283]Suppl. Figure S5A upper and lower panels respectively). Unlike CK2β depletion, CK2 inhibition did not produce a decrease of HSP70 protein amount (Figure [284]7C), prompting us to investigate the precise mechanisms of HSP70 regulation. Since HSP70 chaperoning activity is regulated by C-terminal phosphorylation mediated by CK2 and other kinases,^[285]77 we checked HSP70 and phospho-HSP70 expression upon CK2 chemical inhibition with silmitasertib in G1E-ER cells. Unexpectedly, we did not detect any significant change in phosphorylation performing WB analysis in both total cell lysates (Figure [286]7C) and in the nuclear fraction ([287]Suppl. Figure S5B). Moreover, investigation of HSP70 localization by WB in nuclear and cytoplasm extracts ([288]Suppl. Figure S5C) as well as by IF ([289]Suppl. Figure S5D) did not evidence significant changes in its distribution upon 24 hours of treatment with β-estradiol, CX-4945 or the combination of the 2 compounds. We next checked if CK2 inactivation could affect the GATA1/HSP70 binding by performing proximity ligation assay (PLA) (Figure [290]7D). HSP70-GATA1 interaction was confirmed in G1E-ER cell line in untreated condition or after β-estradiol alone. However, proximity was significantly reduced after exposure for 24 hours to silmitasertib with or without β-estradiol. Although at 24 hours the protein levels of GATA1 in the combo treatment were already reduced and could partially account for the decrease of PLA dots, the results upon single treatment with CX-4945, which did not produce changes in GATA1 expression, indicate a real decrease in GATA1/HSP70 interactions. Analysis performed at an earlier time point of 6 hours, when GATA1 protein levels are still not altered by CK2 inhibition, showed a significant reduction of PLA dots in the combo treatment when compared with untreated condition (Figure [291]7D). Figure 7. [292]Figure 7. [293]Open in a new tab CK2 is involved in the control of GATA1 stability. (A) HSP70 expression by WB in Ter119^+ cells purified from fetal liver at 14.5 and 17.5 dpc and (B) in G1E-ER cells upon silencing of Csnk2b and treatment with β-estradiol (0.2 µM). In (A) number of mice is indicated with dots (control) or squares (KO); in (B) n = 9 independent experiments. (C) Evaluation of HSP70 protein levels through WB upon CK2 inhibition with CX-4945 (2.5 µM) and induction of differentiation with β-estradiol (0.2 µM) (n = 16 independent experiments). In (A–C), GAPDH was used as loading control. (D) PLA for HSP70 and GATA1 proteins performed in G1E-ER cells after treatment with CX-4945 and β-estradiol for 6 and 24 hours. Red dots indicate interactions, in blue the nuclei; on the right; graph showing the average number of dots/nuclei ± SD. Objective 63×, zoom 1×, acquiring 10 slices along the z-stack; the represented images are the result of maximum projection. The green squares show higher magnifications. Images are representative of 3 independent experiments. (E) Immunofluorescence of GATA1 (red) and CK2β (green) in liver cells from 14.5 dpc fetuses and (F) in G1E-ER cells after treatment with CX-4945 and β-estradiol for 6 hours. In blue the nuclei. Images are represented as a single plane; scale bar: 10 µm. (G) IF for CK2β (red) and HSP70 (green) localization in untreated G1E-ER cells and (H) proximity, through PLA assay, in the same cell line (top) or in 14.5 dpc total fetal livers of control embryos (bottom). In both (G) and (H) nuclei are stained with DAPI (blue) and images are represented as maximum projection of 10 section z-stack. 63× objective, zoom 2×. Scale bar: 10µm. In (A–D), graphs represent mean ± SD. Statistical significance was analyzed by Student’s t test: *P < 0.05, **P < 0.01, ***P < 0.001; *significant compared to untreated samples; # significant compared to β-estradiol alone. KO = knockout; PLA = proximity ligation assay; WB = western blot. Next, we observed in fetal liver cells of 14.5 dpc control mice, that GATA1 and CK2β displayed partial co-localization in the nuclear compartment (Figure [294]7E). In untreated G1E-ER cells both at 6h (Figure [295]7F) and 24h after plating ([296]Suppl. Figure S5E), CK2β was distributed diffusely and partially co-localized with GATA1 in the nucleus. Co-localization persisted after β-estradiol addition with an enrichment of the signals of both proteins in the nuclei ([297]Suppl. Figure S5E and S5F). Intriguingly, CK2 inhibition caused a redistribution of CK2β in a diffuse pattern much less overlapping with GATA1 (Figure [298]7F). In addition, immunofluorescence (IF) experiments showed that CK2α and β subunits co-localized in G1E-ER cells in untreated conditions and after 24 hours of β-estradiol exposure with accumulation in the nuclear compartment, thus indicating the presence of CK2 holoenzyme during erythroid differentiation ([299]Suppl. Figure S5G). Intriguingly, silmitasertib in combination with β-estradiol produced the reduction of the nuclear amount of both α and β subunits ([300]Suppl. Figure S5G). We also checked CK2β and HSP70 relationship. IF performed on G1E-ER cells at basal condition revealed the absence of co-localization in the nuclear compartment (Figure [301]7G), data confirmed also through PLA assay in this cell line and in fetal liver cells (unsorted) from control embryos (Figure [302]7H). Altogether, these results suggest that CK2 might control GATA1 through a dual mechanism: CK2β is involved in sustaining HSP70 protein levels, while CK2α-dependent kinase activity is important for the maintenance of HSP70-GATA1 interactions, likely independent on HSP70 phosphorylation. A schematic molecular model of our findings is summarized in Figure [303]8. Figure 8. [304]Figure 8. [305]Open in a new tab Visual summary of molecular changes upon CK2β KO/down-modulation or CK2 inhibition. (A) Molecular phenotype of control vs (B) CK2β KO embryos focusing on cKIT and EPO-R signaling pathways and the effects of their alterations on HSPC and erythroid cell fate. KO embryos are pale with a smaller liver, and present a lower expression of cKIT receptor but normal levels of EPO-R with reduced levels of plasma EPO; downstream both receptors in KO samples AKT and ERK effectors are less activated and reduced in total amount; there is also depletion of STAT5 and GATA1 transcription factors. Deregulation of these pathways lead to expansion of LSK population with likely a reduction in self-renewal capability, and a partial impairment of cell viability and differentiation potential. On erythroid compartment, loss of CK2β causes an accumulation of immature Ter119^- erythroblasts with reduction of their proliferation rate and a partial apoptosis; in Ter119^+ fraction CK2β loss leads to the impairment of final differentiation with reduction of mature erythrocytes also due to increased apoptosis. Moreover, more mature erythroid cells show morphologic alterations with increased dimensions, membrane irregular shape, and less deformability. (C) Proposed model of CK2-dependent control of GATA1 stability. The dashed arrow indicates a still unknown mechanism. (1-top) lack of CK2β impairs GATA1 stability and enhances its degradation; CK2β absence causes also HSP70 protein level reduction, likely compromising its synthesis or increasing its turnover, and this event might accelerate the degradation of the transcription factor. (2-down) treatment with CX-4945 during erythroid differentiation causes delocalization of CK2 from the nuclear compartment and in turn, destabilizes HSP70/GATA1 complex. HSP70 levels are not decreased and HSP70 is not de-localized into cytoplasm after treatment with CX-4945; however, there are less interactions between the 2 proteins, making GATA1 more prone for degradation. In both conditions, 1 and 2, GATA1 turnover is increased by means of proteasome and caspase activation; the reduction of GATA1 levels leads to the decrease of its transcription activity. AKT = serine-threonine kinase; EPO-R = erythropoietin-receptor; ERK = extracellular signal-regulated kinase; HSCs = hematopoietic stem cells; KO = knockout. DISCUSSION We have herein described the phenotype of mice lacking CK2β in the hematopoietic compartment and provided substantial evidence of a key role of this protein in 2 processes: (1) HSPCs progression towards more differentiated precursors; (2) regulation of erythropoiesis, exerted by controlling proliferation, apoptosis and GATA1 function. We discovered that CK2β -deficient HSCs are expanded, due to a higher proliferation rate, but at the same time an amount of these cells undergoes apoptosis (Figure [306]2). This is in line with our previous findings in leukemia stem cells^[307]23 and other studies that demonstrate the importance of CK2 in sustaining cell viability and the quite different roles of the 2 CK2 subunits in the control of cell cycle: while CK2α is more associated to cell cycle progression,^[308]78–[309]80 free CK2β is also involved in cell cycle inhibition in particular in the regulation of the checkpoint between G2/M phases.^[310]81,[311]82 Moreover, HSPCs exhibited alterations in the expression of transcription factors important for the maintenance of stemness properties as well as for lineage commitment^[312]40,[313]42,[314]44,[315]46,[316]47 (Figure [317]2H). Thus, a combination of different problems such as the unbalanced expression of these factors, the higher mitotic rate, and reduced cell viability could lead to the loss of quiescence and subsequent exhaustion of HSPC pools coupled with reduction of their differentiation potential. Altogether, these several elements would also explain, at least in part, the impairment of HSPC engraftment in competitive (Figure [318]3) and non-competitive ([319]Suppl. Figure S1C-E) transplantation experiments and the reduced capability to form colonies in a selective environment such as in methylcellulose cultures ([320]Suppl. Figure S1A and S1B). Although we did not observe a reduction in CXCR4 expression in CK2β KO HSPCs (Fig. [321]3G), we do not completely exclude reduced capabilities in homing process to the BM of these cells. Moreover, alteration in transcription factors could favor the commitment towards preferred populations such as monocyte/macrophages and granulocytes observed in KO embryos. Thus, these data suggest that CK2β might tune the HSPCs’ balance between proliferation and differentiation. FACS analysis pointed out that CK2β KO embryos were able to produce some mature hematopoietic cells (Figure [322]1) of the myeloid lineage that likely undergo apoptosis at later time points. Thus, a combination of alterations in cell viability and in differentiation programs could determine the CK2β KO phenotype such as the reduced cellularity of fetal livers. To note, KO embryos also displayed reduction in B mature (B220^+CD19^+) cells with expansion in percentages of immature pre/pro B population, data that support our previous finding obtained in the Csnk2b^fl/fl/CD19^+/Cre KO mouse model.^[323]35 The depletion of mature erythroid compartment (Ter119^+) in CK2β KO embryos is another cause of reduced fetal liver cellularity. We discovered stage-dependent erythroid functions for CK2β. CK2β promotes cell cycle progression in immature erythroblasts (Ter119^-CD71^+) protects from apoptosis and supports differentiation in mature Ter119^+ cells. The significant reduction of cKit expression in KO samples might contribute to the observed alterations in HSPCs and erythroblasts (Figures [324]2F and [325]4C). Indeed, at the BFU-E stage, cells are mainly dependent on cKit receptor for proliferation and differentiation,^[326]83 while in CFU-E and in pro-erythroblasts stages cKit is co-expressed with EPO-R and is able to transactivate EPO-R intracellular domain.^[327]3,[328]58,[329]83 We demonstrated that in Ter119^-/lowCD71^+ population sorted from CK2β KO fetal livers, which contains CFU-E, BFU-E, and pro-E, the key downstream effectors, such as ERK and AKT, are less activated (Fig. [330]5H). Thus, it is conceivable that an impairment of cKit and EPO-R signaling accounts for the proliferative and differentiation defects. Although in CK2β KO more mature erythroid cells (Ter119^+) we did not detect a depletion in EPO-R protein levels (Figure [331]5D), the trend of reduction in EPO concentration could suggest a less effective stimulation upstream of this receptor (Figure [332]5C). Since during fetal life EPO is mainly produced by hepatocytes and macrophages of the blood islands,^[333]84 we cannot exclude that macrophage production of EPO is impaired in CK2β KO embryos. Moreover, we observed in KO embryos the down-modulation of Siglec1, Vacam1, and Selplg (Figure [334]5A) encoding for the adhesion molecules essential for the blood island formation.^[335]61 These data may suggest that there are looser interactions between nurse macrophages and erythroblast cells with potential negative impact on erythroid maturation and viability. Biochemical and RNA-seq experiments confirmed the ineffective activation of cKit and EPO-R downstream cascades (Figure [336]5A and [337]5E). For instance, CK2β was found important to positively regulate STAT5 (Figure [338]5A and [339]5E) that has a key role in EPO-dependent erythropoiesis being mainly involved in the up-regulation of BCL-xL expression.^[340]85,[341]86 The inhibitory effect on BCL-XL seems to be specific to this BCL-2 family member; indeed, focusing on other factors belonging to the same protein family, we observed an increase of the anti-apoptotic factor MCL-1 in Ter119^+ CK2β KO samples, likely due to an (ineffective) attempt to sustain cell viability. While BCL-XL is a key survival factor in more mature erythroid cells, MCL-1 is reported to be more important for erythroid precursors.^[342]57 Ter119^+ cells from CK2β-null samples evidenced also the up-regulation of the pro-apoptotic proteins BAX and PUMA, showing overall a clear unbalance versus cell death signals. We discovered that CK2β sustains 2 other key erythroid transcription factors, GATA1 and FOG1, whose reduction in KO samples was associated to the consequential down-modulation of Lrf, Klf, β-Globin, and Alas2 target genes. To further support our data, in particular the impairment of β-Globin production, Ouyang et al^[343]87 demonstrated that CK2 is involved in the positive regulation of EKLF transcriptional activity through direct phosphorylation of its interaction domain. Altogether, this transcriptional dysfunction might underlie the phenotypic changes of higher apoptosis, accumulation of immature erythroblasts, presence of non-heme iron deposits ([344]Suppl. Figure S3F), and abnormal shape likely due to deregulation of adhesion molecules and ion/water-channels as suggested by alteration of Aqp1 and Slc gene expression.^[345]63 Moreover, we confirmed through ektacytometer analysis less deformability of CK2β KO erythrocytes (Figure [346]4G), underlying a higher risk of hemolysis when these cells are exposed to mechanical stresses in the blood stream. In CK2β KO mice the effect on erythroid lineage is likely due to both lack of β chains and a decrease of CK2 enzymatic activity; depletion of β subunits inhibits holoenzyme formation, thus partially impinging on kinase efficiency, but it might also make the α subunits more unstable and be degraded as suggested by the reduction of CK2α protein levels in CK2β-null Ter119^+ cells (Figure [347]4A). Heterozygous mice are not anemic although adult animals present some alterations in bone marrow and spleen; this mild phenotype could be explained since β subunits are produced in large excess compared to catalytic counterparts,^[348]12 thus the half-dose of CK2β is likely sufficient to keep CK2 kinase activity at normal levels ([349]Suppl. Figure S2B) and to preserve CK2β independent functions without reducing cell viability or influencing dramatically erythroid differentiation. The dysfunctional GATA1/FOG1 activity, by impinging on platelet formation, could also explain the presence of hemorrhages in KO embryos (Figure [350]1A). Indeed, aberrant platelet development and function have been recently demonstrated in the conditional CK2β^fl/flPF4-Cre mouse model in megakaryocytes.^[351]88 We can suppose that the ineffective hemostasis in the CK2β KO embryos might concur with the impaired erythropoiesis to premature lethality. Moreover, given the broad functions of this kinase, it is conceivable that the erythroid phenotype in KO mice could be the result of the simultaneous impairment of other transcription factors (Ikzf1, Klf13, Smad3) as indicated by RNA-seq data (Figures [352]5 and [353]S3B, [354]S3C).^[355]64,[356]65,[357]89 We have provided substantial evidence that CK2 could control GATA1 mainly through post-translational mechanisms. To note, RNA-seq results suggest a widespread control by CK2β of genes that encode for ubiquitin ligases (Trim^[358]90 and Rnf^[359]91 family genes) and deubiquitinases (Usp genes) ([360]Suppl. Tables S1 and [361]S2). Thus, the extensive signaling impairment downstream EPO-R could be linked to a shared CK2β-regulated mechanism, whereby this protein would be essential to avoid an excessively fast protein turnover. To suggest such a possible mechanism, we exploited G1E-ER cell model of erythroid differentiation. We confirmed in these cells, upon CK2 inhibition or Csnk2b silencing and induction of differentiation, the molecular alterations obtained in vivo for the signal cascades downstream EPO-R. However, some discrepancies in the results obtained between the in vivo and in vitro systems could be due to the lack of terminal erythroid differentiation capability of G1E-ER cells^[362]6,[363]71–[364]73 and the residual expression of CK2β in silencing experiments. Moreover, some differences between CK2 inhibitor and Csnk2b silencing effects are reasonable considering that the free amount of β dimers^[365]12 could still work as chaperone when the alpha kinase activity is inhibited. With this cell line model, we partially elucidated the molecular mechanisms of CK2-dependent GATA1 regulation: CK2 maintains the nuclear localization and distribution of GATA1 and upholds its levels, likely by potentiating the activity of HSP70, which is central in regulating GATA1 protein degradation rate preventing its cleavage by caspases.^[366]76 CK2β sustains HSP70 levels in erythroid cells through a mechanism independent from the phosphorylation of the C-terminal region (Figure [367]7C), known to contain target residues of CK2, CK1, and GSK3 kinases.^[368]77 Evidence suggests that while CK2β might keep high the levels of HSP70, CK2α might regulate the stability of the HSP70- GATA1 association (Figure [369]7D). Although CK2 co-localizes with GATA1 in the nuclear compartment at basal condition and during differentiation (Figures [370]7E, [371]7F, and [372]S5E), the exact mechanism exploited by CK2 to control HSP70 abundance, thus influencing GATA1 stability, has not been elucidated yet. Considering the lack of co-localization in IF and signal in PLA assays performed with CK2β and HSP70 probes (Figure [373]7G and [374]7H), a direct interaction between these 2 proteins is unlikely. Also, HSP70 abundance may not be essential in the CK2-mediated stabilization of GATA1 at mid-gestation, since the reduction of GATA1 in fetal liver KO Ter119^+ cells occurred earlier (14.5 dpc) than that of HSP70 (17.5 dpc) (Figures [375]5E and [376]7A). We demonstrated that inhibition of caspases with ZVAD ([377]Suppl. Figures S4H and S4I) or blockade of proteasomes, through bortezomib ([378]Suppl. Figure S4G), can rescue GATA1 nuclear expression upon CK2β down-modulation or CK2 chemical inhibition. These data indicate that in our experimental conditions, both mechanisms are involved in GATA1 degradation. Caspase activity seems to be more linked to CK2-dependent regulation of HSP70, whereas degradation through proteasome might be driven by other mechanisms that have not been elucidated yet: (1) CK2-dependent control of ubiquitin ligases or deubiquitinases, as suggested by RNA-seq data, or (2) CK2 possible influence on other axis of degradation such as the one mediated by phosphorylation and acetylation of GATA1 followed by its interaction with co-chaperone Hsp27.^[379]92 Moreover, the kinase activity was found to be required to keep CK2β and CK2α in the nucleus closer to the GATA1/HSP70 complex (Figures [380]7F and [381]S5D) but it does not seem to influence HSP70 intracellular localization ([382]Suppl. Figures S5C and S5D). To this regard, Filhol O. et al.^[383]93 postulated that nuclear localization may sustain the function of CK2 and it is known that CK2β autophosphorylation on Ser2 and Ser3 relies on to the formation of a CK2 multimeric structure^[384]94 that in turn might reduce the nuclear-cytoplasmic shuttling of the kinase. It has also been reported that autophosphorylation on Ser2, Ser3, and that on Ser209 (this latter indirectly mediated by CK2 through the intermediate effector CDK1 kinase) could be important in the regulation of CK2β degradation.^[385]95,[386]96 Thus, the loss of CK2 might affect GATA1 levels in 2 sequential steps: (1) upon the reduction of CK2β and of CK2α kinase activity an increased rate of GATA1 degradation occurs when differentiation is induced; (2) the subsequent dissociation of the HSP70-GATA1 complex or depletion of HSP70 causes a further degradation of the transcription factor. Since CK2α and CK2β could also work independently from each other, we can hypothesize that they might influence HSP70 in different ways, one playing on HSP70 levels and the other one on HSP70 ability to interact with other proteins. In conclusion, the described novel functions for CK2β in HSPC biology and erythropoiesis and in the EPO-R and GATA1 signaling, indicate a key role for this kinase as a master regulator of blood cell development. ACKNOWLEDGMENTS