SUMMARY
Accumulating evidence suggests that Notch signaling is active at multiple points during hematopoiesis. Until recently, the majority of such studies focused on Notch signaling in lymphocyte differentiation and knowledge of individual Notch receptor roles has been limited due to a paucity of genetic tools available. In this manuscript we generate and describe animal models to identify and fate-map stem and progenitor cells expressing each Notch receptor, delineate Notch pathway activation, and perform in vivo gain and loss of function studies dissecting Notch signaling in early hematopoiesis. These models provide comprehensive genetic maps of lineage-specific Notch receptor expression and activation in hematopoietic stem and progenitor cells. Moreover, they establish a previously unknown role for Notch signaling in the commitment of blood progenitors towards the erythrocytic lineage and link Notch signaling to optimal organismal response to stress erythropoiesis.
INTRODUCTION
Notch signaling defines a conserved, fundamental pathway, responsible for determination in metazoan development and is widely recognized as an essential component of lineage specific differentiation and stem cell self-renewal in many tissues (Artavanis-Tsakonas et al., 1995; Kopan and Ilagan, 2009), including the hematopoietic system. Hematopoiesis is a complex process that requires coordination between proliferation, self-renewal, and differentiation of stem and progenitor cells to generate mature blood cells (Orkin and Zon, 2008). All Notch receptor paralogs (Notch1-4) and their ligands have been implicated in the regulation of diverse functions in the hematopoietic system. The best-described functions of Notch are in the emergence of fetal hematopoietic stem cells (HSC) (Clements et al., 2011; Dzierzak and Speck, 2008; Kumano et al., 2003) as well as T cell commitment and early development. Indeed, the significance of Notch1 for T lymphocyte commitment, differentiation and oncogenic transformation has been well established (Ciofani and Zúñiga-Pflücker, 2005; Grabher et al., 2006; Tanigaki et al., 2002). Recent studies have also suggested a function for Notch in hematopoietic regeneration (Butler et al., 2010; Varnum-Finney et al., 2011), however its relevance for the self-renewal and maintenance of adult HSC has been questioned (Maillard et al., 2008).
On the other hand, data regarding its involvement in non-lymphoid adult blood lineages is scarce and often controversial. Recent studies suggested a role for Notch4 in megakaryocyte differentiation (Mercher et al., 2008), however further studies in human hematopoietic progenitors challenged this conclusion (Poirault-Chassac et al., 2010). Furthermore, there is little evidence connecting specific Notch receptors with non-lymphoid hematopoietic lineages. We recently reported that the conditional silencing of Notch signaling in the bone marrow results in the expansion of granulocyte-monocyte progenitors (GMP) and that eventually these animals develop a chronic myelo-monocytic leukemia (CMML)-like disease (Klinakis et al., 2011) suggesting that Notch signaling might be involved in early stem/progenitor cell fate decisions.
To fate-map Notch receptor expression and pathway activity in the hematopoietic system we used tamoxifen-inducible CreER knock-in mice for individual Notch receptors in combination to a Notch reporter strain (Hes1GFP). Our lineage-tracing studies have revealed an intriguing division of labor between Notch1 and Notch2, with the former marking mainly lymphocyte progenitors and the latter reaching peak levels during early erythropoiesis. Interestingly, Hes1 or Notch2 expressing progenitors were enriched for erythroid potential and upregulated the expression of an erythroid gene program. Accordingly, conditional Notch gain-of-function in hematopoietic progenitors promoted erythroid commitment and Notch loss-of-function decreased the number of erythroid progenitors and increased peripheral blood platelet counts. Using a combination of genetic fate mapping, transgenic reporters, and conditional Notch gain/loss-of-function we define lineages regulated by individual Notch receptors and reveal a role for Notch signaling in physiological and stress erythropoiesis.
RESULTS
Notch receptor in vivo lineage tracing reveals a division of labor during early hematopoiesis
To lineage-trace Notch receptor expression in hematopoiesis we have used mice with the Cre-ERT2 cassette knocked into the endogenous loci of each of the Notch receptors. We crossed them to the ROSA26-RFP reporter strain (Luche et al., 2007) (Figure 1A). After tamoxifen administration, Notch(1-4)CreER mice were analyzed following various periods of chase and only Notch1 and Notch2 were detectable in bone marrow progenitors. After both 3 and 7 day chase, Notch1CreER predominantly labeled bone marrow progenitors with lymphoid potential including lymphoid-primed multipotent progenitors (L-MPP) and common lymphoid progenitors (CLP). (Figure 1B-D, Figure S1A, B). In contrast, Notch2CreER-labeled cells were found mostly in non-lymphoid progenitors indicating that there is lineage-specific expression of Notch receptors in stem/progenitor cells. The peaks of Notch2 labeling were within the HSC and pre-erythrocytic stages of differentiation. Interestingly, short chase labeling experiments indicated an almost complete absence of Notch1 labeling within all non- lymphoid progenitor subsets, likely reflecting the lack of receptor expression of Notch1 in all such subsets. Intriguingly, even after 20wks of chase there were sustained higher levels of Notch2 labeling within the myelo-erythroid fraction (Figure S1C, D).
Figure 1. Notch(1-4)CreER in vivo lineage tracing suggests a division of labor between Notch1&2 during early adult hematopoiesis.
(A) Schematic depiction of the Notch(1-4)CreER × ROSA26-tdRFP mouse strain. (B) Analysis of RFP reporter expression 72hrs after a single injection of tamoxifen focusing at defined stem and progenitors populations. Frequency of reporter labeling is represented as mean ± standard deviation (SD) (n=5 for each cohort). (C-D) Analysis of RFP reporter expression in LT-HSC and lymphoid progenitors and granulocyte/monocyte (G/M) and erythrocyte-megakaryocyte (Ery/Mk) progenitor subsets one week after a single tamoxifen injection (n=5). Bars denote mean ± SD. (E) Antibody staining with APC conjugated anti-Notch1 and PE conjugated anti-Notch2. Notch1 expression (blue histograms) and Notch2 (red histograms) are over-layed on isotype controls (grey filled histograms) for the same population. *p<0.05, **p<0.005
HSC: Linneg/cKit+/Sca1+/Flt3−/CD48−/CD150+, 48DP: Linneg/cKit+/Sca1+/Flt3−/CD48+/CD150+, 48SP: Linneg/cKit+/Sca1+/Flt3−/CD48+/CD150−, LMPP: Linneg/cKit+/Sca1+/Flt3+/IL7rα−, CLP: Linneg/cKitlo/Sca1lo/Flt3+/IL7rα+, GMP: Linneg/cKit+/Sca1−/CD41−/CD150−/FcgRII/III+, pre-MegE: Linneg/cKit+/Sca1−/CD41−/FcγRII/III−/CD150+/CD105−, CFU-E: Linneg/cKit+/Sca1−/CD41−/FcγRII/III−/CD150−/CD105+, MkP: Linneg/cKit+/Sca1−/CD41+/CD150+, DN3: CD4−/CD8−/CD44−/cKit−/CD25+, pro/preB: B220+/IgM−. See also Figure S1.
Closer analysis of progenitor subsets in Notch2CreER mice revealed that the percentage of RFP+ cells was significantly higher in erythroid and megakaryocytic progenitors (pre-MegE, CFU-E, and MkP) than in granulocyte/monocyte progenitors (GMP) at both 3 and 7 days after tamoxifen injection. After a 3-day chase, approximately 15% and 30% of pre-MegE and CFU-E respectively were RFP+ compared to 2% in the GMP subset (Figure 1B). This implied that expression of Notch2 could be important in the megakaryocyte-erythrocyte (Meg-E) versus granulocyte-monocyte (GM) cell fate decision at an early progenitor stage. Additionally, Notch2CreER-labeled cells were found with increasing frequency within the erythroid progenitor fraction (up to 35%) in comparison to megakaryocyte progenitors (MkP<10%) (Figure 1B,D). From the bipotent pre-megakarocyte-erythrocyte progenitor (pre-MegE) to the CFU-E erythroid progenitor, there was a sustained increase in RFP+ cells whereas the frequency actually decreased between the pre-MegE and the MkP at both time points. Notch3 and Notch4 labeling was not detected in the bone marrow (Figure 1B-D). Notch3 labeling appeared in early thymic T cell progenitors and was detectable in mature T cells with longer periods of chase (Figure 1C and data not shown). Notch4 labeling was extremely infrequent throughout the hematopoietic system but was detectable in splenic CD8+CD11c+ dendritic cells (DC), in agreement with previous qPCR data (Sekine et al., 2009) (Figure S1F). To confirm lineage tracing results at the level of transcription we carried quantitative RT-PCR analysis for the four Notch receptors (Figure S1E). Similar to our lineage tracing results, mainly Notch2 mRNA was detected in hematopoietic stem cells and only low levels of Notch1 mRNA. In the myelo-erythroid progenitor compartment, Notch1 was detected at low level in GMP and pre-MegE whereas Notch2 was detected at significantly higher levels. Only Notch2 mRNA was detected in CFU-E and MkP progenitors. Consistently, no expression of Notch3 and Notch4 mRNA was detected in these populations.
Furthermore, we were able to confirm these fate-mapping studies using receptor-specific antibodies (Fiorini et al., 2009). Indeed, Notch1 expression increased during commitment to the lymphocytic lineage and did not appear to be detectable outside the lymphoid lineage. Conversely, Notch2 was not expressed in early lymphoid progenitors (Figure 1E). Expression of Notch2 was low in CMP and GMP, but significantly higher in erythroid progenitors (Pre-MegE, CFU-E, ProE). Notch2 surface expression reached a peak in CFU-E progenitors and pro-erythroblasts but began to decrease at the basophilic erythroblast stage (Figure 1E and not shown). In comparison to erythroid progenitors, MkP expressed lower levels of surface Notch2. Whereas only a fraction of cells from populations like HSC were stained in the lineage tracing experiments, most of these cells appeared to be stained using antibodies. This discrepancy is most likely reflecting a partial recombination efficiency of Cre-ER, particularly after limited number of tamoxifen injection. Consistently with the lineage tracing results, Notch4 surface expression was detectable in a subset of splenic DC (Figure S1G) and undetectable in hematopoietic stem and progenitor cells in the bone marrow (Figure S1H), once more suggesting that Notch4 signaling plays no role during early hematopoiesis. Our combined lineage tracing and antibody labeling studies suggested a very tightly regulated division of labor between the Notch1 and Notch2 receptors during early stages of hematopoiesis.
In vivo mapping of Notch pathway activation during early hematopoiesis
To combine lineage tracing and receptor expression to pathway activation we used a novel reporter knock-in allele (Fre et al., 2011), in which the Hes1 locus, a well-known direct transcriptional target of Notch, drives expression of GFP (Hirata et al., 2002; Ntziachristos et al., 2012) (Figure 2A). To validate the model, we initially characterized the thymus, where the role of Notch1 is well established. We found that Hes1 mRNA expression was more than 50-fold higher in cells expressing Hes1GFP and that expression of other well-characterized Notch target genes was also increased in Hes1GFP+ cells, confirming that Hes1-expressing cells reflected the activation of the Notch pathway (Figure S2A). In the thymus, Hes1GFP expression was first detected in a fraction of early T cell progenitors (ETP) and increased during T cell commitment up to the DN3 (CD4−CD8−CD25+cKit−CD44−) stage where Notch activation reached its peak (Figure S2B,C). After this stage of differentiation, Notch signaling activity is down-regulated (Kleinmann et al., 2008). Accordingly, we found that in CD4+CD8+ double positive T cells (DP), the expression of Hes1GFP was also down-regulated (Figure S2B,C). Consistent with this differentiation stage-specific Hes1 expression, GFP+ cells were located primarily in the thymic cortex (Figure S2D). These studies validated the use of the HES1GFP mice as a faithful Notch reporter in the immune system. In agreement with this idea, high levels of Hes1 expression were also found in marginal zone B cells (MZB), which require Notch signaling (Moriyama et al., 2008; Tanigaki et al., 2002) (not shown).
Figure 2. The Hes1GFP reporter identifies novel points of Notch pathway activity.
(A) Targeting strategy used to express GFP from the endogenous Hes1 promoter. (B) Analysis of LSK stem and progenitor cells in bone marrow of Hes1-GFP reporter mice. (C) High resolution separation of Linneg/cKit+/Sca1− progenitors on the basis of CD105, CD150, CD16/32 (FcγRII/III) and CD41 expression and expression of Hes1GFP in erythroid progenitors in bone marrow and (D) spleen (A representative of more than five experiments is shown). (E) Immunofluorescence staining of spleen from Hes1GFP reporter mouse showing co-localization of GFP and CD71 staining in the red pulp. (F) Representative histograms showing GFP levels in erythroid progenitor in bone marrow (upper panel) and spleen (lower panel) of Ncstnwt/wt Hes1wt/GFP and Ncstn−/− Hes1wt/GFP. (G) Bar graphs representing average percentage of GFP positive cells in displayed populations from bone marrow (upper panel) and spleen (lower panel). See also Figure S2.
Notch pathway activation marks initiation of commitment to the erythroid lineage
In the bone marrow, Hes1GFP expression was detected within the Linnegc-Kit+ hematopoietic fraction containing stem and progenitor cells. Interestingly, Hes1 expression was detected within the LT-HSC fraction and was virtually undetectable at the downstream multipotential progenitor (MPP) subsets (Figure 2B). Whole-transcriptome studies of Hes1GFP-positive and negative Linneg/cKit+/Sca1+ (LSK) cells supported our phenotypic analysis as they correlated the Hes1GFP fraction to the expression of genes, characteristic of the HSC stage (Figure S3A). Interestingly, Hes1GFP LSK cells also expressed gene signatures correlating to erythrocytic differentiation, suggesting an early molecular bias towards the erythrocytic and against the GM/Mk lineages (Figure S3B-C). Further fractionation of LT-HSC based on their GFP expression showed that the observed erythroid bias seen at the level of LSK population was also evident in this compartment. Indeed, LT-HSC expressed higher levels of key erythroid genes such as Klf1 or Epor and lower levels of myelo/lymphoid genes such as Sfpi1 (Figure S3D). Furthermore, when LSK GFP+ and GFP− were tested for their erythroid potential in methylcellulose CFU-E colony assay, LSK cells expressing Hes1 showed higher erythroid colony forming potential (Figure S3E). In agreement with this expression profiling and our aforementioned lineage tracing experiments, which demonstrated an erythroid bias in Notch2CreER labeling, Hes1 expression was virtually undetectable at the CMP and GMP progenitor subsets (Figure S2E) and significantly de-repressed at the bipotent pre-MegE stage and subsequently increased as cells committed to the erythroid lineage, at the pre CFU-E and CFU-E stages (Figure 2C). The extent of Notch activation was more pronounced in the spleen, a major site of erythrocytic differentiation in the mouse, as high levels of Hes1GFP were detected in erythrocytic progenitors (Figure 2D, E). This higher pathway activity in the spleen could not be explained by differential surface expression of Notch2 and most likely reflects differential ligand availability in this tissue (data not shown). Interestingly, the activation of the Notch pathway was transient in both tissues as it was down-regulated at later stages of erythrocytic differentiation (Figure S2F). Hes1 expression remained low in MkP suggesting that Notch is not active (or is attenuated) in early megakaryocyte development (Figure S2G).
To directly connect Hes1GFP expression to Notch signaling we have crossed the Hes1GFP mice to animals that conditionally lack gamma-secretase activity and thus Notch signaling (Vav1-cre Ncstnf/f). We had previously shown that Ncstn deletion phenocopies Notch receptor loss and that Ncstn−/− phenotypes could be rescued totally by the expression of an activated form of Notch (Klinakis et al., 2011). Using Vav1-cre Ncstnf/f Hes1GFP mice we were able to show that Notch inactivation led to an almost complete reduction of Hes1GFP levels in erythrocyte progenitors, suggesting direct regulation of Hes1 expression by Notch signaling in erythrocytic progenitors (Figure 2F,G).
Differential patterns of Notch activity between fetal and adult hematopoiesis
As initiation of erythropoiesis is a crucial event during fetal hematopoiesis we decided to study the activation of Notch signaling and its relationship to red blood cell differentiation in the embryo. We first looked at primitive hematopoiesis in the yolk sack. We carried quantitative RT-PCR for each of the Notch receptors as well as for Hes1. Yolk sack HSPC expressed both Notch1 and Notch2 but showed no Hes1 expression suggesting that Notch signaling is not activated at this stage. Interestingly, megakaryocytes and red blood cells showed only expression of Notch2 and red blood cells also expressed moderate but significant level of Hes1, suggesting activation of the Notch pathway (Figure S3F-G). We then focused on fetal liver embryonic hematopoiesis. We probed Notch receptor expression by RT-PCR and found mainly Notch2 expression (Figure 3A). Notch1 had low expression level on HSC, CMP and GMP but was not expressed in MEP, consistent with our observation in adult stem and progenitor cells. We then used the Hes1-GFP reporter strain to probe activation of the Notch pathway. Interestingly, we found that, fetal liver hematopoiesis follows a distinct pattern of activation than what we reported in adults. At embryonic day 13.5, the majority of the phenotypic LT-HSC cells expressed significant amounts of Hes1 however this expression was absent from more committed progenitors (Figure 3B). Hes1GFP+ HSC cells were bona fide LT-HSC capable of long-term full lineage reconstitution of lethally irradiated recipients (data not shown). Similar to sorted Hes1GFP cells from adult mice, fetal liver HSC expressing Hes1 were characterized by a Notch transcriptional signature (Figure 3C, D), as judged by transcriptome profiling and qPCR studies. Interestingly, Hes1GFP expressing fetal HSC also revealed a lymphocytic gene expression bias in contrast to the erythroid bias found in adult Hes1GFP expressing HSC (Figure 3D, E). Furthermore, Hes1 expression and Notch pathway activation was undetectable in fetal erythroid progenitors, underlining the differences between fetal and adult hematopoiesis in regard to Notch activation.
Figure 3. Differential patterns of Notch activity between fetal and adult hematopoiesis.
(A) Quantitative RT-PCR analysis of Notch receptor genes in Linneg/cKit+/Sca1+/CD48−/CD150+ HSC, Linneg/cKit+/Sca1−/CD34+/FcgRII/IIIlo CMP, Linneg/cKit+/Sca1−/CD34−/FcgRII/III− MEP, Linneg/cKit+/Sca1−/CD34+/FcgRII/IIIhi GMP and CD71+/Ter119+ RBC from E13.5 Fetal Liver. Data represent mean ± SD of 3 biological replicates (B) Expression of Hes1-GFP in Fetal Liver Linneg/cKit+/Sca1+ stem and multipotential progenitor populations separated into LT-HSC (CD48−/CD150+), CD48+/CD150+ double positive cells (CD48DP) and CD48+/CD150− single positive cells (CD48SP) (upper panel); in Linneg/ cKit+/Sca1−/CD41−/FcgRII/III− Pre-MegE (CD150+/CD105−), PreCFU-E (CD150+/CD105+) and CFU-E (CD150−/CD105+) (middle panel) and late erythrocyte progenitors (lower panel). (C) Quantitative RT-PCR analysis of Notch signaling pathway target genes in Linneg/cKit+/Sca1− progenitors (MP) and Linneg/cKit+/Sca1+ (LSK) GFP+ and GFP− from E13.5 Fetal Liver. Data represent mean ± SD of 3 biological replicates (D) Gene set enrichment plots of fractionated Linneg/cKit+/Sca1+ GFP+ versus GFP− from E13.5 Fetal liver of Hes1-GFP mice for the indicated genesets. (E) Heat map of lymphoid differentiation genes and Notch signaling pathway target genes expressed in Linneg/cKit+/Sca1+ GFP+ versus GFP− from E13.5 Fetal Liver. See also Figure S3.
Hes1+ progenitors show increased erythroid potential and an erythroid transcriptional profile
Next, we returned to adult hematopoiesis and further focused on the connection of Notch activity to erythropoiesis. Linneg cKit+ Sca1− progenitors (cKit+ progenitors) were sorted based on Hes1GFP expression and plated into methylcellulose or collagen-based semi-solid medium to assay for lineage potential. Hes1 expressing cells were enriched in erythroid lineage potential whereas Hes1neg mainly differentiated into granulocytes/monocytes or megakaryocytes (Figure 4A). FACS analysis demonstrated Gr1+CD11b+ cells were virtually absent in colonies generated by Hes1+ progenitors and that most cells expressed the erythroid marker Ter119. This enhancement in erythroid potential was supported by CFU-E assays, which confirmed this striking difference in the presence of Hes1 expression (Figure 4B). In addition, there were less CD41+ cells in colonies from Hes1+ progenitors (Figure 4A). We further addressed the megakaryocyte potential of c-Kit+ progenitors in acetylcholinesterase (AchE) stained collagen based semi-solid cultures, which is the definitive way to measure CFU-Mk potential (Jackson, 1973). Hes1+ cells formed significantly fewer AchE+ megakaryocyte colonies than Hes1neg cells (Figure 4C). Furthermore, the infrequent CFU-Mk colonies generated by Hes1+ progenitors were usually much smaller than CFU-Mk colonies from cells that were Hes1neg (data not shown). The absence of a correlation between Notch activation and promotion of the megakaryocytic (Mk) lineage further questions previous experiments suggesting a role for Notch4 signaling in this lineage.
Figure 4. Hes1 expression and Notch activity are predictive of commitment to the erythrocytic lineage.
(A) One week after methylcellulose culture in a complete cocktail of cytokines, colonies from Hes1 expressing (upper panels) and Hes1 negative Linneg/cKit+/Sca1− progenitors (lower panels) were analyzed by FACS for expression of Gr1 vs CD11b and Ter119 vs CD41. Representative images of colonies are shown. (B) CFU-E in methylcellulose supplemented with EPO and (C) CFU-Mk assays in collagen gel with TPO and IL-3 of sorted Linneg/cKit+/Sca1− progenitors from Hes1GFP bone marrow. (D) Hierarchical clustering of fractionated Hes1GFP Linneg/cKit+/Sca1− progenitors with other progenitor populations. (E) Heat map of Notch target genes and key genes expressed in the erythroid, granulocyte/monocyte (G/M), and megakaryocyte (MegaK) lineages. Bars denote standard deviation. (F) Lineage potential of sorted pre-MegE progenitors was evaluated in complete methylcellulose seven days after sorting (G) CFU-E assay and (H) CFU-Mk assay of pre-MegE sorted based on Hes1 expression (Hes1+ dark grey bar, Hes1− light grey bar). For (B,C) and (F-H) data are representative of average ± SD of 3 independent experiments. *p<0.05, **p<0.005. See also Figure S4.
To analyze the transcriptional program of progenitors with an active Notch pathway, we sorted cKit+ progenitors based on expression level of Hes1GFP and used them for whole transcriptome analysis. Cluster analysis of microarrays from Hes1GFP mice revealed that Hes1+ progenitors have a transcriptional profile almost identical to PreCFU-E erythroid progenitors (Pronk et al., 2007), whereas Hes1− progenitors clustered closely with PreGM and MkP populations (Figure 4D). Also, we compiled genes important in erythroid, granulocyte/monocyte (GM), and megakaryocyte lineage specific differentiation and found that essential erythroid and Notch target genes were both upregulated in Hes1+ progenitors (Figure 4E). The expression of GM and Mk-specific genes was down-regulated. Changes in expression of selected genes were verified by quantitative qPCR. As expected, in the Hes1GFP+ cells, Hes1 and Nrarp expression was significantly upregulated as was the expression of erythroid genes (Epor, Gypa) (Figure 4E and Figure S4A). The transcription factors Runx1 and the thrombopoietin receptor (Mpl) are essential for generation of normal megakaryocytes and platelets. In GFP+ progenitors, expression of both Runx1 and Mpl were decreased compared to GFP−. Moreover, the expression of transcription factors known to be important in the GM lineage, such as Pu.1 (Sfpi1) and Cebpa were reduced in GFP+ progenitors (Fig 4E and Figure S4A).
To further compare the gene expression profiles of Hes1+ to Hes1− LinnegcKit+ progenitors, we used gene set enrichment analysis (GSEA) and analyzed expression of lineage-affiliated gene sets establish by comparing the transcriptional profile of HSC to more committed progenitors such as LMPP, GMP, and MEP (Ng et al., 2009). Genetic signatures expressed in early (s-ery) and more committed (d-ery) erythroid progenitors were significantly enriched in Hes1+ LinnegcKit+ cells. In contrast, gene sets characteristic of granulocyte/monocyte and lymphoid progenitors (r-myly) showed a negative enrichment (Figure S4B). As previously mentioned, our microarray analysis indicated that key megakaryocyte associated genes were repressed in Hes1+ progenitors. Notably, using similar gene sets (Mercher et al., 2008) we revealed an inverse relationship between the expression of Hes1 and megakaryocyte-related genes. Transcriptional profiling therefore supports the notion that Hes1 expression is characteristic of progenitors committed (or destined to commit) to the erythroid lineage.
To probe the role of Notch signaling in human erythropoiesis we examined public gene expression databases for expression of Notch-related genes. In agreement with the results presented thus far, we found, using meta-analysis of whole transcriptome data (Novershtern et al., 2011), that Notch pathway genes were upregulated during human erythroid differentiation (Figure S4E). More importantly, expression of Notch direct transcriptional targets (including Hes1, Hey1, Gata3 and Tcf7) increased in human erythroid progenitors. We therefore propose that Notch activation is an evolutionarily conserved hallmark of erythroid differentiation not only in mice but also in humans suggesting that our findings may be of broader significance.
Notch2 signaling commits bipotent Pre-MegE progenitors to the erythroid lineage
The bipotent Pre-MegE progenitors generate erythroid and megakaryocyte progenitors both in vitro and in vivo (Pronk et al., 2007). To investigate whether Notch signaling can function at this branch-point we sorted Pre-MegE based on Hes1GFP expression and plated cells in semisolid media. Hes1+ cells generated a significantly higher number of pure erythroid colonies and fewer mixed erythroid-megakaryocyte colonies or pure megakaryocyte colonies (Figure 4F). Conversely, Hes1neg Pre-MegE developed more mixed megakaryocyte-erythrocyte colonies and pure megakaryocyte colonies. This suggested that in the absence of Notch signaling, Pre-MegE retain megakaryocytic potential. Also, in CFU-E assays, Pre-MegE that expressed Hes1 generated more CFU-E than the Hes1neg (Figure 4G). We then focused on Notch2, as it is the only Notch receptor expressed on pre-MegE progenitors and the down-stream early erythroid precursors (Figure 1, Figure S1). In agreement with our previous findings, Pre-MegE sorted for surface Notch2 expression generated more CFU-E erythroid colonies than Notch2low progenitors (Figure S4C). Finally, gene-set enrichment analysis of Pre-MegE, sorted based on Hes1GFP expression demonstrated that erythroid genes are enriched in the Hes1 expressing fraction in comparison to the GFP negative fraction. Analysis of megakaryocyte transcription gene sets (Mercher et al., 2008) revealed an inverse relationship between the expression of Hes1 and megakaryocyte-related genes as we previously found with cKit+ progenitors (Figure S4D).
Notch2 gain of function promotes early erythroid progenitor differentiation in vivo
If Notch signaling activation characterizes commitment of adult hematopoietic progenitors to the erythrocytic lineage it is conceivable that genetic manipulation of the pathway could promote or impede adult erythropoiesis in vivo. We thus generated transgenic mice expressing the active domain of each Notch receptor from the ubiquitously expressed ROSA26 promoter (Figure 5A). Using these animals, we conditionally expressed intracellular Notch-IC(1-4) (ICN1-4) using inducible Cre-recombinase strains and monitored expression of the transgene using YFP (Kühn et al., 1995; Ruzankina et al., 2007; Seibler et al., 2003). This approach allowed us to compare the functional consequences of expressing each of the four ICNs from the same promoter in hematopoiesis. Interestingly, although we could demonstrate that all transgenes were expressed at comparable levels (Figure S5A and not shown), only expression of ICN1 was sufficient to induce T cell leukemia in vivo (Figure S5B), suggesting that each ICN defines distinct thresholds of signaling strength. Since Notch2 is the only receptor expressed during early erythropoiesis, we focused our subsequent gain of function studies on the effect of Notch2 activation (ICN2) in bone marrow hematopoietic progenitors. Activation of ICN2 expression in hematopoietic cells using the polyI:polyC inducible Mx1-Cre resulted in a significant increase of CD105+ CFU-E erythroid progenitors and CD71+ erythroblasts in the bone marrow and spleen (Figure 5B-D). Platelet counts were significantly reduced in mice when ICN2 was expressed (Figure 5E and Table S1). Histological analysis (Figure 5F) showed that a dramatic increase in nucleated cells was apparent in the splenic red pulp at low power, and the margins of B cell follicles appeared to be blurred. ICN2 nucleated cells clearly displayed the morphology of erythroid blast cells. Numerous early erythroid progenitors and erythroblasts were abundant. In the bone marrow, there was an increase in erythroid progenitor cells, also confirmed by staining and differential counts of bone marrow smears (data not shown). Strikingly, there was 5-fold reduction of megakaryocytes in ICN2 bone marrow (Figure 5F), in agreement to the suggested negative role of the Notch pathway in megakaryopoiesis.
Figure 5. Notch2 gain-of-function enhances erythroid differentiation.
(A) Generation of ROSA26-Notch(1-4)-IC mice for conditional expression of intracellular Notch and IRES-YFP driven by the ROSA26 promoter (B) Representative FACS plot of Erythroid progenitors. CFU-E progenitors were increased in both the spleen and bone marrow upon conditional expression of ICN2. (C) Representative FACS plot of erythroblasts. CD71+ erythroblasts were increased in the spleen and bone marrow with ICN2, (D) Bar graph showing absolute myelo-erythroid progenitor counts per femur (n=4). (E) Platelet counts from peripheral blood of WT (white bar), ICN1+ (blue bar) or ICN2+ (red bar) mice. (F) H&E stained sections of spleen and bone marrow from control and ICN2 mice. Images in are at 10x magnification with 63x magnification inset in lower right. Bone marrow megakaryocytes were counted in 5 bone marrow (10X) high-powered fields (HPF) (n=3). For (D-F) data are representative of mean ± SD, *p<0.05, **p<0.005 (G) Linneg/cKit+/Sca1− progenitors expressing ICN2 were sorted for gene expression arrays and a heat map of genes involved in lineage specific differentiation was generated. (H) GSEA of erythroid gene signatures (d-ery) and myeloid-lymphoid genes (r-myly) in ICN2+ versus WT littermates Linneg/cKit+/Sca1− progenitors. See also Figure S5 and Table S1.
We next sought to examine gene expression of sorted cKit+ progenitors that express activated ICN2 in vivo. As seen in Hes1+ progenitors, ICN2-expressing progenitors upregulated erythroid genes and suppressed GM and megakaryocyte genes (Figure 5G). Transcriptional targets of Notch were upregulated in ICN2 expressing progenitors, confirming the ICN2 functionality. As with Hes1-expressing cKit+ progenitors and Pre-MegE, the expression of erythroid gene sets (dery) was enriched, while the expression of genes expressed in lymphoid or GM progenitors (rmyly) was suppressed (Ng et al., 2009) (Figure 5H). To further address the involvement of Notch in the choice between the erythroid and megakaryocyte lineages, we imposed the expression of ICN2 in liquid cultures of c-Kit+ progenitors using a ROSA26-CreER driver and 4-hydroxytamoxifen (4-OHT). In the ROSA26YFP control, after induction with 4-OHT, both CD71+ erythroblasts and megakaryocytes were generated in culture. However, induction of ICN2 expression directed the differentiation of the progenitors into mostly CD71+ erythroblasts and inhibited the generation of FSChighCD41+ megakaryocytes (Figure S5C). These combined studies further support our main hypothesis as they demonstrated that Notch2 gain-of-function enforces erythrocytic commitment both in vitro and in vivo.
Notch loss-of-function inhibits early erythroid progenitor differentiation
To study effects of Notch loss-of-function, we used either deletion of Notch1 and Notch2 or Nicastrin (a non-redundant part of the γ-secretase complex) to avoid putative compensatory receptor functions (Klinakis et al., 2011). We conditionally deleted Notch signaling in adult mice using the Mx1-Cre deleter strain and analyzed bone marrow progenitor distribution focusing on erythroblasts. In agreement with previous reports no overt anemia under steady-state conditions was noticed (Maillard et al., 2008; Mancini et al., 2005). However, in the absence of Notch signaling, we found that both the frequency and absolute numbers of CD105+ CFU-E progenitors and CD45−CD71+ erythroblasts were significantly reduced in comparison to control mice (Figure 6A-C). Furthermore, CFU-E and CFU-Mk assays using flow purified Linneg/cKit+/Sca1− progenitors showed decrease ability of Notch deficient progenitors to generate CFU-E and increased ability to generate CFU-Mk (Figure S6A). In contrast to the aforementioned gain-of-function analysis using ICN2 ectopic expression, Notch loss-of-function (in both Notch1−/−2−/− and Ncstn−/− mice) lead to increased platelet counts and numbers of splenic megakaryocytes (Figure 6D, Figure S6B). Cell cycle and apoptosis analysis of pre-MegE, pre-CFU-E and CFU-E populations revealed no significant differences between Ncstn−/− and littermate control (Figure S6C-D), suggesting that the reduction of CFU-E population observed in Notch loss of function models is mainly due to differentiation bias. These data are consistent with our previous in vitro and in vivo studies, enforcing the idea that in a subset of progenitors Notch signaling is important for early stages of lineage commitment and erythroid progenitor differentiation.
Figure 6. Notch loss-of-function affects early erythroid differentiation and recovery from erythroid stress.
(A) Adult mice were analyzed one week after 3 injections of polyI:polyC to induce compound deletion of Notch1 and Notch2 using the Mx1-Cre strain (Mx1cre+Notch1−/−2−/−). The frequency of CFU-E progenitors and CD45−CD71+ erythroblasts is shown. (B) Absolute numbers of early erythroid progenitors in control and Notch1−/−2−/− mice (n=5) from bone marrow and (C) spleen. Data represent mean ± SD. (D) H&E stained paraffin sections of spleens from control, Notch1−/−2−/− and Ncstn−/− mice. Megakaryocytes are indicated with black arrows. (E) Erythropoietic response to acute hemolytic anemia in control (Ncstnf/f) and Ncstn−/− (Ncstnf/f Vav1-cre+) (n=5) after PHZ-induced hemolysis. Data represent mean ± SD. (F) Representative FACS plot (left panel) and quantification of proportion of GFP+ cells in Hes1GFP/wt mice before (blue) and 4 days after (green) sublethal 4Gy irradiation. Data represent mean ± SD of 3 biological replicates (G) Representative FACS plots and (H) absolute quantification of erythroid progenitors from bone marrow of control (Ncstnf/f) and Ncstn−/− (Ncstnf/f Vav1-cre+) (n=3) littermates 4 days after sublethal 4Gy irradiation. Data represent mean ± SD. *p<0.05, **p<0.005, ***p<0.001. See also Figure S6 and Figure S7
Notch signaling is essential for optimal progenitor responses during stress erythropoiesis
To further test the importance of Notch signaling for the generation of adult red blood cells (RBC) and their progenitors, we deleted Ncstn specifically in the hematopoietic system usingNcstnf/f Vav1-cre+animals and studied response to stress caused initially by the administration of phenylhydrazine (PHZ), an oxidative agent able to cause severe hemolytic anemia. Whereas no significant difference in the peripheral blood red cell compartment was observed at steady state (Figure S6B), Ncstn-deficient animals showed a significant delay of recovery from stress as demonstrated by lower numbers of total RBC (Figure 6E) circulating in the peripheral blood. As PHZ mainly targets mature RBC and we have shown that the activation of the Notch pathway is only transient and not evident in the later stages of RBC differentiation we focused on an additional stress stimulus. We selected sublethal ionizing radiation, which rapidly eliminates marrow and splenic erythroid progenitors while sparing mature peripheral RBC (Peslak et al., 2012). Using this stress stimulus we initially demonstrated that irradiation significantly increased the abundance of Hes1GFP erythrocytic progenitors (Figure 6F) in the bone marrow, a response initiated as early as the pre-MegE stage. Interestingly, Notch2 receptor cell surface expression was increased in pre-MegE cells in response to stress (Figure S6D). These studies suggested an enhanced activation of the Notch pathway in response to erythrocytic stress. Most importantly, irradiation of Ncstn−/− animals revealed a profound inability to mount a response to radiation-induced erythroid stress in the absence of Notch signaling. Indeed, both early (CFU-E, pre-CFU-E, pre-MegE) and late (ProE, BasoE, OrthoE) stages of red blood cell differentiation were virtually absent in mice lacking Notch signaling (Figure 6G-H). On the other hand, recovery of the pre-GM population was not significantly altered in Ncstn−/− suggesting that the defects are specific to the erythroid lineage.
As Notch2 is the only Notch receptor expressed on the surface of erythrocytic progenitors we have also tested whether Notch2 deficiency is sufficient to impede differentiation of erythrocyte progenitors in response to stress. To exclude contributions from the microenvironment we have transplanted Vav1-cre+Notch2f/fCD45.2+ bone marrow to irradiated congenic CD45.1+ recipients. As control we have used Vav1-cre−Notch2f/fCD45.2+ bone marrow from littermate animals. Five weeks post-transplant and after we verified that recipient animal bone marrow was comprised by more than 90% Notch2neg donor cells (Figure S7A-B), we have sub-lethaly irradiated the animals and analyzed erythrocytic stress response as previously described. Four days post-irradiation Vav1-cre+Notch2f/f reconstituted animals showed signs of defective stress response including smaller spleens and significantly decreased numbers of pre-CFU-E and CFU-E progenitors (Figure S7C-E) in the spleen. On the other hand there were no significant differences in the recovery of granulocyte/monocyte and megakaryocyte progenitors suggesting specificity for the erythrocytic lineage. All these studies highlight the importance of Notch signaling, and specifically Notch2, in stress erythropoiesis.
DISCUSSION
Our lineage tracing experiments provide a window into the mechanisms utilized by distinct Notch receptors in adult and fetal hematopoiesis. They offer in vivo mapping of receptor expression and activity in the bone marrow and extra-medullary sites (Figure 7). Indeed, they identify a remarkable division of labor between Notch receptors, as they connect Notch1 expression to commitment to the lymphoid lineage, while Notch2 expression corresponds to the initiation of erythrocytic differentiation. On the other hand, Notch3 and Notch4 were not associated with bone marrow stem/progenitor cells. Although CreER-mediated target loci deletion is expression-level dependent and recombination may not occur at low levels of gene expression, our studies strongly suggest that Notch1 and Notch2 are the main regulators of early adult hematopoiesis. In agreement with this notion and our lineage tracing findings, antibody staining for Notch1 and Notch2 in progenitors further confirmed our genetic fate mapping. Furthermore qRT-PCR analysis and antibody staining for Notch4 confirmed the results obtained with the lineage tracing, as mRNA and cell surface expression of this receptor was undetectable in all bone marrow stem and progenitor subsets. Finally, in vivo reporter activity together with loss- and gain-of-function genetic studies demonstrated the crucial role for Notch signaling in early stages of hematopoietic differentiation and stress erythropoiesis. This demonstrates the role of Notch in red blood cells differentiation in vivo. It is noteworthy that, meta-analysis of human progenitor gene expression datasets demonstrated that Notch activity is also increased when human stem and progenitor cells commit to the erythroid lineage, suggesting that the involvement of Notch signaling during erythropoiesis is evolutionary conserved. Finally, we were able to show that Notch-dependent regulation of erythropoiesis occurs only in adult hematopoiesis, and is crucial for efficient recovery from erythrocytic stress and life-threatening anemia induced by exposure to ionizing irradiation or blood loss.
Figure 7. “Road map” of Notch signaling pathway in hematopoiesis.
General overview of level of Notch1 receptor expression (blue gradient), Notch2 receptor expression (red gradient), Notch signaling pathway activation reported by Hes1GFP expression (green gradient) as well as known niches and Notch ligands involved in adult hematopoiesis. LT-HSC: Long Term Hematopoietic Stem Cell; MPP: Multi-Potential Progenitor; CMP: Common Myeloid Progenitor; GMP: Granulocyte/Monocyte Progenitor; Pre-MegE: Pre-Megakaryocyte/Erythrocyte Progenitor; MkP: Megakaryocyte Progenitor; CFU-E: Colony Forming Unit Erythrocyte; ProE: Pro-Erythroblast; LMPP: Lymphocyte-Primed Multi-Potential Progenitor; CLP: Common Lymphocyte Progenitor; ProB: Pro B-cell; FoB: Follicular B-cell; MzB: Marginal zone B-cell; ETP: Early Thymic Progenitor; DN: Double Negative (CD4−8−) T cell progenitor 3.
Our studies also provide insights into the role of Notch signaling in the differentiation of adult HSC. We were able to clearly identify Notch receptors that are expressed on LT-HSC, mainly Notch2, and show that the transcriptional target Hes1 is activated in a subset of HSC under homeostatic conditions. Purification and subsequent serial transplantations of Notch2RFP+ and Hes1GFP+ LSK demonstrated that they contain bona-fide HSC (not shown). In agreement with this notion, analysis of sorted Hes1GFP+ LSK reveals molecular correlation to HSC gene expression signatures. However, we were also able to show that Hes1GFP+ LSK cells have a molecular priming towards the erythrocytic lineage using in vitro assays and transcriptome analysis. This differentiation bias towards the erythrocytic lineage becomes even more apparent at subsequent stages of differentiation: Multipotent LinnegcKit+ or bipotent pre-MegE progenitors expressing Hes1GFP have a propensity to differentiate into erythrocytes and are characterized by erythrocytic gene signatures. On the other hand, without Notch activation, progenitors (Hes1GFP-negative) mainly differentiate towards the granulocyte/monocyte or megakaryocyte lineages. There are significant consequences of these findings, as they suggest that Notch signaling could have distinct roles in different blood lineages. In agreement with this notion, it was shown that Notch could turn from an oncogene (T cell leukemia, B cell lymphoma)(Grabher et al., 2006; Puente et al., 2011) to a tumor suppressor (myelo-monocytic leukemia) (Klinakis et al., 2011) depending on the type of hematopoietic progenitor initiating each disease. Our current studies would suggest that in addition to Notch inhibition, receptor-specific pathway agonism could also be a promising therapeutic alternative, in tumors characterized by silencing of the pathway (Lobry et al., 2013; Lobry et al., 2011).
It would be intriguing to integrate our findings to previously published reports on Notch function in early hematopoiesis. The discovery of a role for Notch signaling in erythrocytic differentiation and stress response is novel and could open clinically important areas of future investigation. In agreement with a role in the promotion of erythropoiesis, we clearly show that Notch signaling suppresses megakaryocytic differentiation, a finding inconsistent with a previous report suggesting that Notch4 can promote differentiation towards this lineage (Mercher et al., 2008). However, those studies were based largely on in vitro cultures performed in the presence of transfected Notch ligands and exogenous Thrombopoietin. Nevertheless, our findings are in agreement with a recent in vitro study focusing on human hematopoiesis and suggesting a negative role for the Notch pathway in megakaryopoiesis (Poirault-Chassac et al., 2010). Although it is difficult to reconcile all these studies, it is possible that they are all correct as at different stages of differentiation Notch signaling could have distinct effects on megakaryocyte-erythrocyte progenitor commitment/maintenance or even mature megakaryocyte differentiation and maturation.
Our data also suggest the existence of progenitor cell niches characterized by Notch ligand expression (Figure 7) responsible for commitment to distinct cell fates. Indeed, our work suggests that in the bone marrow there are distinct Notch ligand-expressing niches, responsible for HSC function, as well as lymphocytic and erythrocytic differentiation. Although it would be intriguing to correlate Notch signaling to specific ligand microenvironments, development of in vivo genetic reporters in combination to advanced imaging is required to further characterize and analyze specific ligand expression and role. Furthermore, our finding that activation of Notch2-mediated signaling stimulates erythroid differentiation and is essential in the response to erythrocytic stress could also be clinically important. Anemia is a common feature of patients with renal disease, chronic heart failure, as well as the majority of cancer patients (Melnikova, 2006). Currently, recombinant EPO is used to treat anemia and provides significant clinical benefit (Eschbach et al., 1987). However, resistance to EPO treatment or an insufficiency of erythroid progenitors is found in many patients (van der Putten et al., 2008). It is possible that such patients will benefit by simultaneous activation of Notch2 and EPO receptors. Bio-available peptide ligands or specific Notch2 receptor agonists could thus be important in such clinical settings.
EXPRERIMENTAL PROCEDURES
Animals
Notch(1-4)CreER and Hes1-GFP knock-in mice were recently described (Fre et al., 2011). ROSA26-ICN(1-4) mice were generated by insertion of a loxP flanked splice acceptor NEO-ATG cassette with two polyA sites followed by ICN(1-4)-IRES-YFP into the ROSA26 locus, allowing the ROSA26 promoter to drive expression of the NEO-ATG cassette. Cre recombinase mediated excision of NEO-ATG results in use of the splice acceptor in the ICN(1-4)-IRES-YFP cassette and irreversible expression of the transgene and the IRES-YFP bi-cistronic mRNA allows expression to be monitored by YFP expression. ROSA26-RFP mice (gift from H-J. Fehling, Ulm University) and ROSA26-YFP animals were (gift from D. Littman, NYU School of Medicine) have been described (Luche et al., 2007; Srinivas et al., 2001). Inducible Cre animals used include: the tamoxifen inducible human ubiquitin C promoter driven CreER (Ubc-CreER) (Ruzankina et al., 2007)(gift from D. Littman, NYU School of Medicine), tamoxifen inducible ROSA26-CreER (Seibler et al., 2003)(gift from D. Littman, NYU School of Medicine), and polyI:polyC inducible Mx1Cre (Jackson Labs). N1f/f N2f/f and Ncstnf/f mice were previously described (Klinakis et al., 2011). Hematopoietic specifique Vav1-cre was previously described (Stadtfeld and Graf, 2005). All animal experiments were done in accordance to the guidelines of the NYU School of Medicine Institutional Animal Care and Use Committee.
Notch lineage tracing and LSK transplants
Tamoxifen (Sigma Aldrich) was solubilized in corn oil (Sigma Aldrich) at a concentration of 20mg/mL and injected intraperitoneally at 0.2mg/g body weight. Following (1 or 3) daily injections and a 3 day, 7 day, or 20 weeks chase, animals were euthanized for analysis of peripheral blood and tissues by FACS. For transplantation of RFP labeled LSK cells, ROSA26CreER, Notch1CreER, and Notch2CreER lineage tracer mice were crossed to ROSA26-RFP reporter mice and injected with tamoxifen daily for 3 days (0.2mg/g mouse). Two days after the last injection, bone marrow was isolated from femurs and tibias. Lineage depleted cells were stained as described and RFP+ LSK cells were sorted and 500-750 cells were transplanted with 2.5×105 helper bone marrow cells into lethally irradiated recipients. Peripheral blood was analyzed 4 weeks and 24 weeks post-transplant.
Stress erythropoiesis
Phenylhydrazine was dissolved in PBS and injected intraperitoneally at 50 mg/kg on two consecutive days to induce acute anemia. Peripheral blood (50μl) was collected and analyzed using the Hemavet 950 (Drew Scientific) hematology system. Sublethal total body irradiation of 4 Gy radiation was used to model endogenous stress erythropoiesis as previously described (Peslak et al., 2012). Progenitor responses were analyzed at day 4 following sublethal irradiation.
Antibodies and Flow Cytometry
Freshly dissected femurs and tibias were dissected and bone marrow was flushed with a 3cc syringe and 25g needle into PBS with 3% fetal bovine serum. The bone marrow suspension was centrifuged at 400 rcf for 10min at 4°C, washed and resuspended in PBS with 3% FBS. Antibody staining and FACS analysis was performed as previously described (Klinakis et al., 2011). All antibodies were purchased from BD-Pharmingen or e-Bioscience. We used the following fluorochrome or biotin conjugated antibodies: CD117 (2B8), Sca-1 (D7), CD11b (M1/70), Gr-1 (RB6-8C5), NK1.1 (PK136), TER-119, CD3 (145-2C11), CD19 (1D3), CD21 (7E9), CD23 (B3B4), CD127 (A7R34), CD34 (RAM34), FcγRII/III (2.4G2 or 93), CD135 (A2F10.1), CD4 (RM4-5), CD8 (53-6.7), CD150 (9D1), CD41 (MWReg30), B220 (RA3-6B2), CD48 (HM481), CD105 (MJ7/18), F4/80 (BM8), CD71 (Rl7217). Bone marrow lineage antibody cocktail included CD11b, Gr-1, NK1.1, TER-119, CD4, CD8, CD3, B220, Il7Rα. For analysis of erythroid progenitors, TER-119 was not included in lineage cocktail as previously described (Pronk et al., 2007).
Microarray and Gene set enrichment analysis
Human hematopoietic population micro array data have been previously described (Novershtern et al., 2011) and are available at GEO database (http://www.ncbi.nlm.nih.gov/geo/) under accession number GSE24759. Data were normalized using the Robust Multi-array Average algorithm using Genespring GX software (Agilent) and gene expression were leveled according to GAPDH expression. Mouse erythoid and myeloid progenitor population micro arrays have been previously described (Pronk et al., 2007) and are available at GEO database under accession number GSE8407.
Microarray analysis was performed as previously described (Klinakis et al., 2011). For each experiment, freshly isolated cells from three individual mice were sorted by surface marker expression and GFP expression, total RNA was extracted using the RNeasy Plus Micro kit (Qiagen). The Ovation RNA Amplification System V2 or Ovation Pico amplification (NuGEN) kits were used for amplification. Amplified RNA was labeled and hybridized to the Mouse 430.2 microarrays (Affymetrix). The Affymetrix gene expression profiling data were normalized using the previously published robust multi-array average algorithm using the GeneSpring GX software (Agilent). The gene-expression intensity presentations were generated with Multi Experiment Viewer software (http://www.tm4.org/mev/). Gene set enrichment analysis was performed using Gene Set Enrichment Analysis software (http://www.broadinstitute.org/gsea) using gene set as permutation type, 1,000 permutations and log2 ratio of classes as metric for ranking genes. Genesets used in this study have been previously published (Mercher et al., 2008; Ng et al., 2009; Pronk et al., 2007). Newly generated microarray data are available at GEO database under accession number GSE46726.
In vitro differentiation assays
Sorted LSK, Linneg/cKit+/Sca1− progenitors (500) or Pre-MegE (500) were plated in triplicate into cytokine-supplemented methylcellulose medium (MethoCult 3434, Stem Cell Technologies). Colonies were scored after 10 days of culture and cells were collected for analysis by FACS. For CFU-E assays cells were plated in methylcellulose medium supplemented with EPO (MethoCult 3334, Stem Cell Technologies) and colonies were counted 2 days after plating. Collagen gel assays for CFU-Mk were performed using Megacult-C (supplemented TPO and IL-3, Peprotech). Collagen gels were acetone fixed and acetylcholinesterase staining was performed according to manufacturers guidelines (MegaCult-C, Stem Cell Technologies).
Statistical analysis
The means of each data set were analyzed using Student's t-test, with a two-tailed distribution and assuming equal sample variance.
Supplementary Material
Highlights.
Notch receptor lineage tracing reveals division of labor during early hematopoiesis
Signaling through Notch2-Hes1 axis promotes commitment to the erythroid lineage
Notch signaling is essential for progenitor response during stress erythropoiesis
ACKNOWLEDGEMENTS
We are grateful to H-J. Fehling for the ROSA26-RFP mice; Linheng Li, Paul Frenette and Amy Wagers for sharing experimental tools and advice. We would like to thank the NYU Genome Technology Center (supported in part by NIH/NCI P30 CA016087-30 grant) for expert assistance with micro-array experiments, and the NYU Flow Cytometry facility (supported in part by NIH/NCI 5 P30CA16087-31) for expert cell sorting, the NYU Histology Core (5P30CA16087-31), the Transgenic Mouse Core (NYU Cancer Institute Center Grant (5P30CA16087-31), and Jacquelyn Freund for technical assistance. I.A. is supported by the National Institutes of Health (RO1CA133379, RO1CA105129, RO1CA149655, and RO1GM088847), the Leukemia & Lymphoma Society (TRP program grants), The V Foundation for Cancer Research, the Irma T. Hirschl Trust, and the St. Baldrick's Foundation for Cancer Research. P.O. was supported by the NYU MSTP Program. C.L. was supported by the Helen and Martin Kimmel Center for Stem Cell Research and is currently a Leukemia and Lymphoma Society Fellow. The research in the S.A-T. laboratory was supported by the NIH (RO1 CA 098402 ). I.A. is a Howard Hughes Medical Institute Early Career Scientist.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
REFERENCES
- Artavanis-Tsakonas S, Matsuno K, Fortini ME. Notch signaling. Science. 1995;268:225–232. doi: 10.1126/science.7716513. [DOI] [PubMed] [Google Scholar]
- Butler JM, Nolan DJ, Vertes EL, Varnum-Finney B, Kobayashi H, Hooper AT, Seandel M, Shido K, White IA, Kobayashi M, et al. Endothelial Cells Are Essential for the Self-Renewal and Repopulation of Notch-Dependent Hematopoietic Stem Cells. Stem Cell. 2010;6:251–264. doi: 10.1016/j.stem.2010.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ciofani M, Zúñiga-Pflücker JC. Notch promotes survival of pre–T cells at the β-selection checkpoint by regulating cellular metabolism. Nature immunology. 2005;6:881–888. doi: 10.1038/ni1234. [DOI] [PubMed] [Google Scholar]
- Clements WK, Kim AD, Ong KG, Moore JC, Lawson ND, Traver D. A somitic Wnt16/Notch pathway specifies haematopoietic stem cells. Nature. 2011;474:220–224. doi: 10.1038/nature10107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dzierzak E, Speck NA. Of lineage and legacy: the development of mammalian hematopoietic stem cells. Nature immunology. 2008;9:129–136. doi: 10.1038/ni1560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eschbach JW, Egrie JC, Downing MR, Browne JK, Adamson JW. Correction of the anemia of end-stage renal disease with recombinant human erythropoietin. Results of a combined phase I and II clinical trial. N Engl J Med. 1987;316:73–78. doi: 10.1056/NEJM198701083160203. [DOI] [PubMed] [Google Scholar]
- Fiorini E, Merck E, Wilson A, Ferrero I, Jiang W, Koch U, Auderset F, Laurenti E, Tacchini-Cottier F, Pierres M, et al. Dynamic Regulation of Notch 1 and Notch 2 Surface Expression during T Cell Development and Activation Revealed by Novel Monoclonal Antibodies. The Journal of Immunology. 2009;183:7212–7222. doi: 10.4049/jimmunol.0902432. [DOI] [PubMed] [Google Scholar]
- Fre S, Hannezo E, Sale S, Huyghe M, Lafkas D, Kissel H, Louvi A, Greve J, Louvard D, Artavanis-Tsakonas S. Notch lineages and activity in intestinal stem cells determined by a new set of knock-in mice. PLoS ONE. 2011;6:e25785. doi: 10.1371/journal.pone.0025785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grabher C, von Boehmer H, Look AT. Notch 1 activation in the molecular pathogenesis of T-cell acute lymphoblastic leukaemia. Nat Rev Cancer. 2006;6:347–359. doi: 10.1038/nrc1880. [DOI] [PubMed] [Google Scholar]
- Hirata H, Yoshiura S, Ohtsuka T, Bessho Y, Harada T, Yoshikawa K, Kageyama R. Oscillatory expression of the bHLH factor Hes1 regulated by a negative feedback loop. Science. 2002;298:840–843. doi: 10.1126/science.1074560. [DOI] [PubMed] [Google Scholar]
- Jackson CW. Cholinesterase as a possible marker for early cells of the megakaryocytic series. Blood. 1973;42:413–421. [PubMed] [Google Scholar]
- Kleinmann E, Geimer Le Lay A-S, Sellars M, Kastner P, Chan S. Ikaros represses the transcriptional response to Notch signaling in T-cell development. Molecular and cellular biology. 2008;28:7465–7475. doi: 10.1128/MCB.00715-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klinakis A, Lobry C, Abdel-Wahab O, Oh P, Haeno H, Buonamici S, van De Walle I, Cathelin S, Trimarchi T, Araldi E, et al. A novel tumour-suppressor function for the Notch pathway in myeloid leukaemia. Nature. 2011;473:230–233. doi: 10.1038/nature09999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kopan R, Ilagan MX. The canonical Notch signaling pathway: unfolding the activation mechanism. Cell. 2009;137:216–233. doi: 10.1016/j.cell.2009.03.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kühn R, Schwenk F, Aguet M, Rajewsky K. Inducible gene targeting in mice. Science. 1995;269:1427–1429. doi: 10.1126/science.7660125. [DOI] [PubMed] [Google Scholar]
- Kumano K, Chiba S, Kunisato A, Sata M, Saito T, Nakagami-Yamaguchi E, Yamaguchi T, Masuda S, Shimizu K, Takahashi T, et al. Notch1 but not Notch2 is essential for generating hematopoietic stem cells from endothelial cells. Immunity. 2003;18:699–711. doi: 10.1016/s1074-7613(03)00117-1. [DOI] [PubMed] [Google Scholar]
- Lobry C, Ntziachristos P, Ndiaye-Lobry D, Oh P, Cimmino L, Zhu N, Araldi E, Hu W, Freund J, Abdel-Wahab O, et al. Notch pathway activation targets AML-initiating cell homeostasis and differentiation. The Journal of experimental medicine. 2013;210:301–319. doi: 10.1084/jem.20121484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lobry C, Oh P, Aifantis I. Oncogenic and tumor suppressor functions of Notch in cancer: it's NOTCH what you think. J Exp Med. 2011;208:1931–1935. doi: 10.1084/jem.20111855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luche H, Weber O, Nageswara Rao T, Blum C, Fehling HJ. Faithful activation of an extra-bright red fluorescent protein in “knock-in” Cre-reporter mice ideally suited for lineage tracing studies. In Eur J Immunol. 2007:43–53. doi: 10.1002/eji.200636745. [DOI] [PubMed] [Google Scholar]
- Maillard I, Koch U, Dumortier A, Shestova O, Xu L, Sai H, Pross SE, Aster JC, Bhandoola A, Radtke F, et al. Canonical notch signaling is dispensable for the maintenance of adult hematopoietic stem cells. Cell stem cell. 2008;2:356–366. doi: 10.1016/j.stem.2008.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mancini SJC, Mantei N, Dumortier A, Suter U, Macdonald HR, Radtke F. Jagged1-dependent Notch signaling is dispensable for hematopoietic stem cell self-renewal and differentiation. Blood. 2005;105:2340–2342. doi: 10.1182/blood-2004-08-3207. [DOI] [PubMed] [Google Scholar]
- Melnikova I. Anaemia therapies. Nat Rev Drug Discov. 2006;5:627–628. doi: 10.1038/nrd2113. [DOI] [PubMed] [Google Scholar]
- Mercher T, Cornejo MG, Sears C, Kindler T, Moore SA, Maillard I, Pear WS, Aster JC, Gilliland DG. Notch signaling specifies megakaryocyte development from hematopoietic stem cells. Cell stem cell. 2008;3:314–326. doi: 10.1016/j.stem.2008.07.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moriyama Y, Sekine C, Koyanagi A, Koyama N, Ogata H, Chiba S, Hirose S, Okumura K, Yagita H. Delta-like 1 is essential for the maintenance of marginal zone B cells in normal mice but not in autoimmune mice. Int Immunol. 2008;20:763–773. doi: 10.1093/intimm/dxn034. [DOI] [PubMed] [Google Scholar]
- Ng SY-M, Yoshida T, Zhang J, Georgopoulos K. Genome-wide lineage-specific transcriptional networks underscore Ikaros-dependent lymphoid priming in hematopoietic stem cells. Immunity. 2009;30:493–507. doi: 10.1016/j.immuni.2009.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Novershtern N, Subramanian A, Lawton LN, Mak RH, Haining WN, McConkey ME, Habib N, Yosef N, Chang CY, Shay T, et al. Densely interconnected transcriptional circuits control cell states in human hematopoiesis. Cell. 2011;144:296–309. doi: 10.1016/j.cell.2011.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ntziachristos P, Tsirigos A, Van Vlierberghe P, Nedjic J, Trimarchi T, Sol Flaherty M, Ferres-Marco D, da Ros V, Tang Z, Siegle J, et al. Genetic inactivation of the polycomb repressive complex 2 in T cell acute lymphoblastic leukemia. Nat Med. 2012 doi: 10.1038/nm.2651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Orkin SH, Zon LI. Hematopoiesis: an evolving paradigm for stem cell biology. Cell. 2008;132:631–644. doi: 10.1016/j.cell.2008.01.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Peslak SA, Wenger J, Bemis JC, Kingsley PD, Koniski AD, McGrath KE, Palis J. EPO-mediated expansion of late-stage erythroid progenitors in the bone marrow initiates recovery from sublethal radiation stress. Blood. 2012;120:2501–2511. doi: 10.1182/blood-2011-11-394304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Poirault-Chassac S, Six E, Catelain C, Lavergne M, Villeval JL, Vainchenker W, Lauret E. Notch/Delta4 signaling inhibits human megakaryocytic terminal differentiation. Blood. 2010;116:5670–5678. doi: 10.1182/blood-2010-05-285957. [DOI] [PubMed] [Google Scholar]
- Pronk CJH, Rossi DJ, Månsson R, Attema JL, Norddahl GL, Chan CKF, Sigvardsson M, Weissman IL, Bryder D. Elucidation of the phenotypic, functional, and molecular topography of a myeloerythroid progenitor cell hierarchy. Cell stem cell. 2007;1:428–442. doi: 10.1016/j.stem.2007.07.005. [DOI] [PubMed] [Google Scholar]
- Puente XS, Pinyol M, Quesada V, Conde L, Ordonez GR, Villamor N, Escaramis G, Jares P, Bea S, Gonzalez-Diaz M, et al. Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia. Nature. 2011;475:101–105. doi: 10.1038/nature10113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ruzankina Y, Pinzon-Guzman C, Asare A, Ong T, Pontano L, Cotsarelis G, Zediak VP, Velez M, Bhandoola A, Brown EJ. Deletion of the developmentally essential gene ATR in adult mice leads to age-related phenotypes and stem cell loss. Cell stem cell. 2007;1:113–126. doi: 10.1016/j.stem.2007.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seibler J, Zevnik B, Kuter-Luks B, Andreas S, Kern H, Hennek T, Rode A, Heimann C, Faust N, Kauselmann G, et al. Rapid generation of inducible mouse mutants. Nucleic Acids Res. 2003;31:e12. doi: 10.1093/nar/gng012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sekine C, Moriyama Y, Koyanagi A, Koyama N, Ogata H, Okumura K, Yagita H. Differential regulation of splenic CD8-dendritic cells and marginal zone B cells by Notch ligands. Int Immunol. 2009;21:295–301. doi: 10.1093/intimm/dxn148. [DOI] [PubMed] [Google Scholar]
- Srinivas S, Watanabe T, Lin CS, William CM, Tanabe Y, Jessell TM, Costantini F. Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Dev Biol. 2001;1:4. doi: 10.1186/1471-213X-1-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stadtfeld M, Graf T. Assessing the role of hematopoietic plasticity for endothelial and hepatocyte development by non-invasive lineage tracing. Development. 2005;132:203–213. doi: 10.1242/dev.01558. [DOI] [PubMed] [Google Scholar]
- Tanigaki K, Han H, Yamamoto N, Tashiro K, Ikegawa M, Kuroda K, Suzuki A, Nakano T, Honjo T. Notch-RBP-J signaling is involved in cell fate determination of marginal zone B cells. Nature immunology. 2002;3:443–450. doi: 10.1038/ni793. [DOI] [PubMed] [Google Scholar]
- van der Putten K, Braam B, Jie KE, Gaillard CA. Mechanisms of Disease: erythropoietin resistance in patients with both heart and kidney failure. Nat Clin Pract Nephrol. 2008;4:47–57. doi: 10.1038/ncpneph0655. [DOI] [PubMed] [Google Scholar]
- Varnum-Finney B, Halasz LM, Sun M, Gridley T, Radtke F, Bernstein ID. Notch2 governs the rate of generation of mouse long- and short-term repopulating stem cells. J Clin Invest. 2011:1–10. doi: 10.1172/JCI43868. [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.







