Erythroid (red blood) cells are the first cell type to be specified in the postimplantation mammalian embryo and serve highly specialized, essential functions throughout gestation and postnatal life. The existence of 2 developmentally and morphologically distinct erythroid lineages, primitive (embryonic) and definitive (adult), was described for the mammalian embryo more than a century ago. Cells of the primitive erythroid lineage support the transition from rapidly growing embryo to fetus, whereas definitive erythrocytes function during the transition from fetal life to birth and continue to be crucial for a variety of normal physiologic processes. Over the past few years, it has become apparent that the ontogeny and maturation of these lineages are more complex than previously appreciated. In this review, we highlight some common and distinguishing features of the red blood cell lineages and summarize advances in our understanding of how these cells develop and differentiate throughout mammalian ontogeny.

During embryonic development, hematopoiesis functions in the rapid production of large numbers of erythroid cells that support the growth and survival of the embryo/fetus and, later, in the generation of a pool of hematopoietic stem cells (HSCs) that persist throughout the life of the adult animal.1,2  Hematopoiesis in the developing vertebrate embryo occurs in multiple waves and in several different anatomic sites (Figure 1). The first wave occurs in the yolk sac in both mouse and humans3-5  and produces primarily primitive erythroid cells (EryP) as well as macrophages and megakaryocytes.3,6  The second wave also arises in the yolk sac but is “definitive,” composing erythroid, megakaryocyte, and several myeloid lineages.7,8  The third wave emerges from HSCs produced within the major arteries of the embryo, yolk sac, and placenta.1-3,9  HSCs home to and expand within the fetal liver and eventually seed the bone marrow.10,11  Definitive erythroid cells are produced continuously from HSCs in the bone marrow throughout postnatal life.1,2 

Figure 1

Shifts in site of hematopoiesis during mouse and human development. (A) Hematopoietic development in the mouse. Formation of mesoderm during gastrulation (around E6.5), development of yolk sac blood islands (∼ E7.5), emergence of HSCs in the aorta-gonad-mesonephros region (E10.5; other sites such as large arteries and placenta not shown), active fetal liver hematopoiesis (E14.5), and hematopoiesis in the bone marrow of the late gestation fetus (E18.5) and adult animal. Active circulation begins at approximately E9.0.43  (B) Hematopoietic development in the human embryo. An embryo at yolk sac stage of hematopoiesis (day 17), at the time of the first hepatic colonization by HSCs (day 23), arterial cluster formation (day 27), second hepatic colonization (day 30), and bone marrow colonization (10.5 weeks). Active circulation begins at approximately day 21.3 

Figure 1

Shifts in site of hematopoiesis during mouse and human development. (A) Hematopoietic development in the mouse. Formation of mesoderm during gastrulation (around E6.5), development of yolk sac blood islands (∼ E7.5), emergence of HSCs in the aorta-gonad-mesonephros region (E10.5; other sites such as large arteries and placenta not shown), active fetal liver hematopoiesis (E14.5), and hematopoiesis in the bone marrow of the late gestation fetus (E18.5) and adult animal. Active circulation begins at approximately E9.0.43  (B) Hematopoietic development in the human embryo. An embryo at yolk sac stage of hematopoiesis (day 17), at the time of the first hepatic colonization by HSCs (day 23), arterial cluster formation (day 27), second hepatic colonization (day 30), and bone marrow colonization (10.5 weeks). Active circulation begins at approximately day 21.3 

Close modal

Before the initiation of definitive (adult-type) hematopoiesis from multipotent stem cells in the fetal liver, the embryonic circulation is dominated by large, nucleated erythroid cells of the EryP lineage. Primitive erythropoiesis is transient: EryP progenitors are produced in the yolk sac of the embryo for only a brief period (∼ 2 days).7,12  Their terminally differentiated progeny persist in the circulation through the end of gestation and even for a while after birth.13  However, they are rapidly outnumbered by the rapidly expanding population of definitive erythroid cells (EryD) arising from the growing fetal liver.13,14 

Failure in primitive erythropoiesis (as observed, for example, after targeted disruption of genes encoding the transcription factors Gata-1, Gata-2, Lmo2, or Scl) is uniformly associated with embryonic lethality.1,15  The importance of this lineage is underscored by the fact that primitive erythropoiesis is conserved among vertebrates.1,15 

During gastrulation, a single epithelial cell layer (the epiblast) is transformed into the 3 germ layers of the embryo (ectoderm, mesoderm, and endoderm), and the basic body plan of the animal is established.16  In the mouse, gastrulation initiates around embryonic day (E) 6.5 (Figure 1A), when surface ectoderm cells undergo an epithelial-mesenchymal transition and become migratory, moving through the primitive streak and into the extraembryonic region of the embryo. There they form a mesodermal layer that lies directly adjacent to the visceral endoderm, in the early yolk sac.4  In the mouse, EryP are first detected at around E7.5, within “blood islands”5  (Figure 1A). They arise from mesodermal progenitors with restricted hematopoietic potential.

A role for yolk sac (visceral) endoderm in hematopoietic and vascular induction was suggested by classic studies in the chick embryo and was supported by the observation that embryoid bodies formed from Gata4-deficient embryonic stem (ES) cells lacked visceral endoderm and showed greatly reduced primitive erythropoiesis.17  Explant experiments with mouse embryos suggested that soluble signals from visceral endoderm activate hematopoiesis and vascular development during gastrulation17  and function, at least in part, through Indian hedgehog (Ihh), which may in turn regulate expression of BMP4 from the adjacent mesoderm.18,19  Recent coculture experiments have indicated that signals from primitive endoderm-derived extraembryonic endoderm cells20  can stimulate the expansion of EryP progenitors.21  Candidate-secreted factors identified on the basis of a microarray analysis include vascular endothelial growth factor and Ihh.21 

Wnt/β-catenin signaling regulates specification of the EryP lineage from mesoderm in differentiating mouse ES cells22  in a process that requires inhibition of the Notch pathway.23  A microarray analysis of maturing primary mouse EryP isolated at distinct developmental stages indicated that a number of Wnt/β-catenin pathway genes are expressed in EryP progenitors and are rapidly down-regulated as the cells mature.12  That the Wnt pathway is active and functions autonomously in EryP progenitors was suggested by immunostaining for activated (stabilized) β-catenin and by fluorescence of a nuclear-GFP Wnt/β-catenin reporter in the blood islands of the yolk sac.12 

The close spatial and temporal association between primitive erythropoiesis and vasculogenesis (the formation of blood vessels from angioblasts) in the yolk sac led to the concept of a common progenitor termed the “hemangioblast.”1,15  Hemangioblasts were thought to give rise to “blood islands,” clusters of EryP surrounded by endothelial cells within the mesodermal layer of the yolk sac.5  Experimental support for the hemangioblast came from studies of differentiating mouse and human ES cells24,25  and from mouse embryos.26  Although “blast colony”-forming cells (BL-CFCs) were hypothesized to be the in vitro equivalent of the hemangioblast,24  the majority of BL-CFC activity in the mouse embryo was found not in the yolk sac but in the posterior primitive streak.26  BL-CFCs are not strictly bipotential: in addition to primitive and definitive hematopoietic cell types,6,24  they can also give rise to smooth muscle in vitro.27 

On the basis of lineage-tracing experiments, it was concluded that EryP and angioblasts in the yolk sac do not share a common progenitor.28  Analysis of mouse tetrachimeras expressing different fluorescent proteins has suggested that blood islands are polyclonal and, therefore, that hematopoietic and angioblastic lineage commitment in the yolk sac does not involve a 2-potential progenitor.29  However, it may be possible to reconcile these findings with the BL-CFC analyses from mouse embryos26 : the rare hemangioblasts in the posterior streak may produce more restricted progenitors (eg, megakaryocyte-erythroid progenitors, EryP progenitors, angioblasts) that then colonize the yolk sac. In a careful immunohistochemical analysis of the early mouse yolk sac,5  independent blood islands could not be identified. Instead, a “blood band” of CD41-positive hematopoietic progenitor cells was observed (∼ E7.75) to gradually circumscribe the proximal yolk sac.5  It was proposed that this blood band, containing primitive erythroid cells and more sparsely distributed endothelial cells, is later subdivided by layers of endothelial cells.5  Consistent with this idea, 2 waves of angioblast development have been identified in the yolk sac: one closely associated with and a second independent of hematopoiesis.30  Therefore, it remains unclear whether an equivalent of the hemangioblast exists in vivo.

Although it has been proposed that an Flk1-positive hemangioblast undergoes a transition to a “hemogenic” endothelial intermediate before the emergence of hematopoietic cells,31,32  the early wave of blood development in the yolk sac does not progress through a frank endothelial intermediate.5,33 

Embryonic and fetal erythropoiesis has been best studied in the mouse. Primitive and definitive erythroid cells are distinct lineages that arise from different populations of mesoderm (posterior and lateral plate, respectively) generated during gastrulation.4  Cells of the 2 erythroid lineages differ in a number of important characteristics. The “megaloblastic” EryP are much larger than either fetal liver or adult EryD (Figure 2), express distinct globin genes,34  and differ in their O2-carrying capacity and response to low oxygen tension.35  Whereas EryP form only in the yolk sac, progenitors for EryD are found both in the yolk sac and fetal liver.7,36  The primitive and definitive erythroid lineages also differ in their dependence on specific cytokines, transcription factors, and downstream regulatory pathways.

Figure 2

Primitive red blood cells are megaloblastic. EryP (E10.5) were mixed with (A) fetal (E17.5) or (B) maternal peripheral blood erythrocytes, cytospun, and stained with Giemsa as described by Fraser et al.13  Scale bar represents 20 μm.

Figure 2

Primitive red blood cells are megaloblastic. EryP (E10.5) were mixed with (A) fetal (E17.5) or (B) maternal peripheral blood erythrocytes, cytospun, and stained with Giemsa as described by Fraser et al.13  Scale bar represents 20 μm.

Close modal

More than a century ago, it was observed that mammalian blood contains distinct nucleated and enucleated erythroid cells, leading to the conventional wisdom that a key distinguishing feature of primitive and definitive erythroid cells at all stages was the presence or absence of a nucleus.37  In contrast with EryD, which enucleate extravascularly in the fetal liver or adult bone marrow before entering the bloodstream, EryP were thought to retain their nuclei throughout their development. These large circulating cells were thought to represent a “primitive” form of erythropoiesis because, like the nucleated red cells of nonmammalian vertebrates, they formed in the yolk sac and were confined to embryonic stages of development. However, it is now clear that primitive erythroblasts in the mouse embryo are nucleated for only part of their life span, maturing and expelling their nuclei around midgestation.13,14  They then continue to circulate as enucleated cells until and perhaps, for a short time, past the time of birth.13  In addition to enucleation late in their maturation,13,14  the 2 lineages share a number of other features, including terminal differentiation from unipotential progenitors, progressive stages of maturation as nucleated erythroblasts,13,14,38  accumulation of hemoglobin, decrease in cell size, nuclear condensation, and regulation by erythroid transcription factors, such as GATA1 and EKLF/KLF1.12-14,37,39  The cytologic changes observed during primitive erythroid maturation are shown in Figure 3. EryP display “maturational” gene switching within the β-globin locus40  involving sequential changes in expression from βh1- to εY- (both embryonic) to adult βmaj- (β1) globin. It is evident from Figure 4A that εY-globin remains the predominant transcript throughout EryP development and that, even at late times, βmaj-globin expression remains low. The data are presented in a normalized form in Figure 4B, with peak expression set at 100 for each gene, to emphasize the sequential gene expression.

Figure 3

Cytologic changes during primitive erythroid maturation. Giemsa-stained cytospin preparations of FACS-sorted E8.5 EryP from dispersed ε-globin-H2B-GFP embryos12,42  or peripheral blood from wild-type embryos at E9.5 to E14.5.13  Panels E9.5 to 14.5 were taken from Fraser et al.13  Scale bar represents 20 μm.

Figure 3

Cytologic changes during primitive erythroid maturation. Giemsa-stained cytospin preparations of FACS-sorted E8.5 EryP from dispersed ε-globin-H2B-GFP embryos12,42  or peripheral blood from wild-type embryos at E9.5 to E14.5.13  Panels E9.5 to 14.5 were taken from Fraser et al.13  Scale bar represents 20 μm.

Close modal
Figure 4

Maturational globin gene switching in developing primitive erythroid cells. “Maturational” globin gene switching40  refers to sequential changes in expression from βh1- to εY- (both embryonic) to adult βmaj- (β1) globin within the primitive erythroid lineage during its differentiation. (A) Relative expression is shown for the 3 β-like globin genes. εY-globin remains the predominant transcript throughout EryP development; even at late times, βmaj-globin expression remains low. (B) The data are presented in a normalized form, with peak expression set at 100 for each gene, to emphasize their sequential expression within maturing EryP from E8.5 to E12.5 (J.I., Z. He, M.H.B., unpublished data, February 2009).

Figure 4

Maturational globin gene switching in developing primitive erythroid cells. “Maturational” globin gene switching40  refers to sequential changes in expression from βh1- to εY- (both embryonic) to adult βmaj- (β1) globin within the primitive erythroid lineage during its differentiation. (A) Relative expression is shown for the 3 β-like globin genes. εY-globin remains the predominant transcript throughout EryP development; even at late times, βmaj-globin expression remains low. (B) The data are presented in a normalized form, with peak expression set at 100 for each gene, to emphasize their sequential expression within maturing EryP from E8.5 to E12.5 (J.I., Z. He, M.H.B., unpublished data, February 2009).

Close modal

Primitive erythropoiesis has been difficult to study, in part because of the relative inaccessibility of the mammalian embryo and because EryP form at a time when the embryo is still extremely small. In addition, in the human embryo, the time during embryonic development at which blood islands first emerge (days 16 and 17, Figure 1B) is too early to be accessible from elective terminations of pregnancy.3  This difficulty underscores the value of the ES cell system for investigations into these early embryonic events.41  Past midgestation, maturing EryP and adult-type erythrocytes are present simultaneously within the circulation. The single monoclonal antibody that specifically recognizes mouse erythroid cells, Ter-119, binds to both EryP and EryD. To circumvent these problems, we have used human ε-globin regulatory elements to specifically drive expression of reporters, including lacZ,17,18  a pan-cellular green fluorescent protein (GFP),13,18  and a histone H2B-GFP fusion,42  within the mouse EryP compartment of transgenic mice (Figure 5). The fluorescent reporters have made it possible to isolate EryP progenitors12  and maturing EryP,13,42  to track EryP enucleation,13,42  and to generate a genome-wide transcriptome of this lineage at distinct stages of its development.12 

Figure 5

Nuclear GFP reporter for EryP. (A) Histone H2B-GFP expression within the yolk sac “blood islands” of an E8.5 ε-globin-H2B-GFP transgenic embryo. (B) GFP(+) cells within the yolk sac vasculature of an E9.5 ε-globin-H2B-GFP transgenic embryo. (C) Nuclear expression of H2B-GFP fusion protein permits identification of mitotic figures and actively dividing cells. Blue represents 4,6-diamidino-2-phenylindole. Embryos were imaged as in Isern et al.12,39 

Figure 5

Nuclear GFP reporter for EryP. (A) Histone H2B-GFP expression within the yolk sac “blood islands” of an E8.5 ε-globin-H2B-GFP transgenic embryo. (B) GFP(+) cells within the yolk sac vasculature of an E9.5 ε-globin-H2B-GFP transgenic embryo. (C) Nuclear expression of H2B-GFP fusion protein permits identification of mitotic figures and actively dividing cells. Blue represents 4,6-diamidino-2-phenylindole. Embryos were imaged as in Isern et al.12,39 

Close modal

In the mouse, erythroid progenitors are found only in the yolk sac, but not in the embryo proper, between E7.25 and E9.0.7,12  Primitive erythropoiesis may involve a limited hierarchy of progenitors in which a bipotential megakaryocyte-erythroid progenitor gives rise to unipotential EryP progenitors. In the mouse embryo, the first megakaryocytes, identified by staining for surface GP1bβ, are found in the yolk sac at E9.5 and in the blood at E10.5, substantially later than primitive erythroblasts.6  It is not clear whether the apparent temporal difference in emergence of these 2 lineages reflects a limitation in this method of detection of megakaryocytes or a true kinetic difference in maturation of their progenitors. In any case, EryP are the first morphologically identifiable hematopoietic cells of the embryo.

The signals that initiate differentiation of EryP progenitors, apparently just as the cells begin to enter the bloodstream,7,43  are unknown. EryP progenitors were initially identified retrospectively by their ability to form red-pigmented colonies in cultures containing semisolid medium supplemented with erythropoietin44  and were termed EryP-CFC.7  The identification and isolation of these progenitors have presented challenges because of a lack of specific cell surface markers. Expression of CD31 (PECAM1), Tie-2, CD34, and VE-cadherin45  or low level expression of CD4146  has been used to enrich for EryP progenitors from E7.0 to E7.5 embryos, but these proteins are also expressed on endothelial and/or definitive hematopoietic progenitors.

Recently, it has been possible to isolate EryP progenitors prospectively from E7.5 to E8.5 embryos12  on the basis of an EryP-specific GFP reporter expressed in mice.42  In this line, expression of a nuclear histone H2B-GFP fusion protein is driven by a human embryonic ε-globin promoter and sequences from the β-globin locus control region42  (Figure 5). The fusion protein coats chromatin and remains detectable through all phases of the cell cycle (Figure 5C). Thus, it permits tagging and tracking of EryP nuclei and identification of mitotic cells and has proven to be a very useful model for studying embryonic erythropoiesis.12,39,42,47  GFP fluorescence (and endogenous expression of embryonic globin genes) was detected in mouse embryos as early as E7.512 ; therefore, transgene expression is activated toward the end of gastrulation, at the time when the first erythroid cells are specified from nascent mesoderm.28  At E7.5 and E8.5, a substantial proportion (∼ 15%-20% and 50%, respectively) of all cells in the embryo expressed GFP, indicating that significant resources are set aside by the developing embryo for this single lineage.12  EryP progenitor activity, evaluated by quantifying EryP-CFC, was found exclusively in the FACS-sorted GFP-positive population and expanded rapidly between E7.5 and E8.5.12  A subset of GFP-positive cells from E7.5 embryos expressed Flk1, c-kit, VE-cadherin, and Tie-2 proteins, all normally associated with endothelial cells.12  All progenitor activity was found in the fraction of GFP-positive cells that expressed c-kit, whereas Tie-2 expression provided a 4-fold enrichment of progenitors in the GFP-positive population.12  Although progenitor activity is lost once EryP begin to enter the bloodstream,43  circulating EryP do continue to divide until around E13.5 (N. Neriec, S.T.F., M.H.B., unpublished data, August 2006).37 

Interestingly, a variety of cell adhesion proteins (Pecam1, CD44, and the integrins α4, α5, β1, and β3) are found on the surface of EryP progenitors.12  Genes encoding additional adhesion molecules as well as collagens are also expressed in these cells.12  Therefore, within the yolk sac, EryP may form tight associations with each other and/or with surrounding endothelial cells, as suggested by electron microscopy.48  Loss of adhesion proteins from the surface of EryP in the yolk sac, along with expression of metalloproteases, might help to untether the cells and facilitate their entry into the bloodstream.12 

Shortly before midgestation in the mouse embryo, the circulatory systems of the embryo, yolk sac, and placenta connect. As the heart begins to beat (∼ E9.0), proerythroblastic EryP enter the circulation as a synchronous cohort of nucleated cells.43  Circulating EryP in rodents continue to mature within the bloodstream.13,14,38  They display a developmental progression from proerythroblast to orthochromatic erythroblast to reticulocyte that is characterized by loss of nucleoli (E9.5-E10.5), decreased cell diameter and cross-sectional area (E10.5-E11.5), condensation of nuclei to 20% of their original size (E10.5 onwards), expression of cell adhesion proteins (E12.5-E14.5),13,42  and enucleation to primitive reticulocytes (E12.5 onwards)13,14  (Figure 3). Thus, the maturation of primitive erythroblasts is stepwise and developmentally synchronized. EryP maturation is reflected in a progressive loss of CD71 and up-regulation of Ter-119 expression,13  analogous to that observed for fetal liver erythropoiesis.49  Proteins, such as β1 integrin and phosphatidylserine, are selectively partitioned onto the membrane surrounding the expelled nuclei of adult erythroid cells.50-52  In contrast, Ter-119 is partitioned to the reticulocyte membrane.51  Selective reorganization of surface proteins, such as Ter-119, CD71, and several integrins, occurs during enucleation of EryP as well.13,42  We have speculated that loss of adhesion proteins during maturation may facilitate entry of the reticulocyte into the circulation and that preferential coating of the extruded nucleus may facilitate its engulfment by macrophages.42 Figure 6 contains a summary of these changes.

Figure 6

Model for protein redistribution on surface of maturing erythroid cells. As EryP prepare to enucleate, redistribution of surface antigens (Ter119, orange; α4 and β1 integrins, blue) occurs such that the reticulocyte (enucleated erythroid cell) is decorated with Ter119 but displays little, if any, α4 and β1 integrin. Conversely, the extruded nucleus is preferentially coated with α4 and β1 integrins, perhaps facilitating engulfment by macrophages.42  Protein redistribution has been described for maturing adult erythroblasts as well.51 

Figure 6

Model for protein redistribution on surface of maturing erythroid cells. As EryP prepare to enucleate, redistribution of surface antigens (Ter119, orange; α4 and β1 integrins, blue) occurs such that the reticulocyte (enucleated erythroid cell) is decorated with Ter119 but displays little, if any, α4 and β1 integrin. Conversely, the extruded nucleus is preferentially coated with α4 and β1 integrins, perhaps facilitating engulfment by macrophages.42  Protein redistribution has been described for maturing adult erythroblasts as well.51 

Close modal

The possibility that EryP enucleate was suggested by 2 reports from the 1970s describing enucleated megalocytic, hemoglobin-containing cells in the bloodstream.37  Four decades later, an antibody against a mouse embryonic β-like hemoglobin chain was used to demonstrate that EryP begin to undergo nuclear extrusion by E12.5.14  These observations were confirmed using a flow cytometric method in which nucleated and enucleated EryP could be distinguished and quantified in blood on the basis of binding of the cell-permeable DNA-binding fluorescent dye DRAQ5 and EryP-specific expression of GFP.13  Analysis of the kinetics of enucleation revealed a precipitous (50%) reduction in nucleated EryP between E13.5 and E14.5; this process was nearly complete by E15.5.13  However, the total number of GFP-positive EryP cells remained approximately the same from E12.5 to the end of gestation (R. Moore, S.T.F., M.H.B., unpublished data, October 2009).13  Thus, in contrast with the prevailing view that EryP survive for only 4 or 5 days and gradually disappear during gestation, perhaps by apoptosis,53,54  these experiments indicated that EryP are a stable population and that enucleation does not foreshadow their death.13  For at least 3 weeks after birth, approximately 0.5% of all cells in the bloodstream are GFP-positive EryP.13  It seems likely that the life span of a primitive erythrocyte is similar to that of an adult red blood cell and that EryP, like their adult counterparts, are eventually cleared by the spleen.

In contrast with definitive erythrocytes, which are released directly into the circulation from the fetal liver or adult bone marrow as enucleated cells, EryP enter the blood with their nuclei intact, at around E9.0, with the initiation of a strong heartbeat.43  EryP eventually extrude their nuclei, but, surprisingly, the first enucleated EryP are not detectable in the blood for at least 3 days (E12.5) after the cells have begun to circulate.13,14  This phase of primitive erythropoiesis is nearly complete by approximately E15.5.13,14  An explanation for this lag may be that EryP appear to collect and complete their maturation within the fetal liver,42,55  an organ that does not form until midgestation.

Detailed analysis of the surface phenotype of GFP-positive EryP revealed that integrins and other cell surface adhesion proteins are expressed on EryP beginning around E12.5, reflecting a previously unrecognized aspect of their maturation13  and suggesting that they may home to a fetal tissue, such as the liver, where they would continue to mature. Transgenic mouse lines expressing pancellular13  or nuclear42  (Figure 5) GFP were used to test this hypothesis. The fetal liver is the site of development of adult-type red blood cells, which mature within “erythroblastic islands” (EBIs).56  EBIs, which are also found in bone marrow and spleen, are 3-dimensional structures containing a central macrophage surrounded by erythroid cells at various stages of maturation.56  Cytoplasmic extensions from the macrophage make intimate contact with the erythroblasts, leading to the suggestion that the macrophages function as nurse cells and also engulf expelled erythroid nuclei.56 

EryP appear to collect in the fetal liver and to integrate into EBIs, along with developing definitive erythroblasts.42  The EryP found in the fetal liver had uniformly and strongly up-regulated a range of adhesion molecules, including α4, α5, and β1 integrins and CD44, and showed greater adhesion to macrophages in reconstituted EBIs than did circulating EryP.42  The “rosettes” formed by these interactions were disrupted by a blocking antibody against VCAM-1, the macrophage receptor for α4β1 integrin42  and by peptide blocking of fibronectin binding by α4 and α5 integrins (J.I. and M.H.B., unpublished data, April 2008). Disruption of EryP-macrophage interactions using α4 integrin were reported by one group55  but was not observed by us, for blocking antibodies against either α4 or β1 integrin.42  Confocal microscopy and FACS analyses of fetal livers from the ε-globin–H2B-GFP (nuclear GFP) transgenic mice indicated that nuclei extruded by EryP, like those of adult red blood cells,52  undergo phagocytosis by macrophages.42  In both adult and embryonic lineages, nuclear extrusion occurs by budding off of the highly condensed nucleus surrounded by a rim of plasma membrane.55 

The nuclear-GFP reporter made it possible to monitor EryP enucleation at high resolution using flow cytometry. Bright GFP-positive structures with very low granularity were FACS-purified from the bloodstream and fetal liver and displayed the morphology expected for expelled nuclei, with a surrounding remnant of cytoplasm and a cell membrane.42  Using antibodies against mouse embryonic β-hemoglobins to identify EryP, others have also found that these cells can interact with macrophages, which can engulf the expelled nuclei in vitro.55  The GFP fluorescent signal allowed us to sort and analyze the extruded nuclei, which were highly enriched for integrins, in contrast with the enucleated primitive reticulocytes that showed little or no expression of these proteins.42  Therefore, adhesion molecules are specifically redistributed onto the membrane surrounding the expelled nucleus, perhaps causing it to become more attractive for engulfment by fetal liver macrophages42  (Figure 6). Protein sorting has also been described for enucleating adult erythoblasts.51  The stages of primitive erythropoietic development are summarized in Figure 7.

Figure 7

Stages of primitive erythropoiesis. Summary of different phases of EryP development, from progenitor to bloodstream (where the cells continue to undergo limited proliferation) to terminal maturation and enucleation. The images of EryP at different stages were cropped from photographs of actual Giemsa-stained cells.

Figure 7

Stages of primitive erythropoiesis. Summary of different phases of EryP development, from progenitor to bloodstream (where the cells continue to undergo limited proliferation) to terminal maturation and enucleation. The images of EryP at different stages were cropped from photographs of actual Giemsa-stained cells.

Close modal

Although the numbers of free GFP-positive EryP nuclei detected in the circulation were lower than in the fetal liver, suggesting that a small fraction briefly escape engulfment by macrophages and enter the bloodstream, it is also possible that some EryP enucleate while in circulation. The free nuclei presumably are phagocytosed later, when they circulate through the fetal liver or other tissues,42  possibly including the placenta, as reported for the human embryo.57  Human primitive erythroblasts also enucleate, but the site is apparently the placental villi rather than the fetal liver.57  Microscopic analysis suggests that the nuclei expelled by human EryP are engulfed by macrophages, although this finding remains to be confirmed more directly57  (eg, using FACS). Interestingly, macrophage progenitors are present in the human placenta before the onset of circulation.57  Whether enucleation of EryP also occurs in the mouse placenta is not known.

Collectively, these results indicate that one of the major roles of fetal liver macrophages is the engulfment and destruction of erythroid nuclei. Although a role for macrophages in definitive erythroid cell maturation is well accepted,56  several lines of evidence suggest that macrophages are not essential for their enucleation in vivo.58  Enhancement of EryP enucleation during coculture on fetal liver macrophages was detected in one study55  but not in another.42  Nevertheless, interactions with macrophages may facilitate other aspects of erythroid maturation. For example, erythroid progenitors expand and mature more efficiently in vitro in the presence of stromal cells,59  and definitive erythroblast proliferation is stimulated by coculture with macrophages.60 

The fetal liver provides a microenvironment(s) for the expansion and differentiation of definitive HSCs, from which definitive erythroid cells differentiate via a hierarchy of progenitors.1,2  For many years, the first definitive erythroid cells were thought to originate from HSCs that colonize the fetal liver.1,2,61  However, a second wave of hematopoiesis was found to emerge from the yolk sac at around E8.25 to E8.5; it produces cells of the definitive erythroid lineage, identified in vitro using an assay for erythroid burst-forming units, as well as megakaryocyte and myeloid lineages.6-8,62  The second wave of hematopoiesis also includes multipotential, highly proliferative progenitors (HPP-CFC) identified in vitro.62  HPP-CFC and more extensively self-renewing erythroid progenitors that expand in culture63  are both found in the yolk sac at about the same time of development. These progenitors are maintained in culture under different growth factor and cytokine conditions,62,63  however, and their lineage relationship in vivo (if any) is unclear. In addition, HPP-CFC lack HSC activity and are found in the yolk sac 12 to 24 hours earlier than the first newborn repopulating HSCs64  are detected (M. Yoder, personal oral communication, October 2011).

Recently, it has been demonstrated that erythropoiesis in the fetal liver begins before its colonization by HSCs.36  Early definitive erythroblasts were isolated by flow cytometry on the basis of expression of c-kit (which is not found on EryP at this stage of their development12,13 ) and shown to display a distinct and unique profile of endogenous mouse and transgenic human globin gene expression.36  Maturing definitive erythroblasts isolated from E9.5 yolk sac show the same pattern of β-globin gene expression as the early (E11.5) definitive fetal liver erythroblasts. This observation and several other lines of circumstantial evidence suggest that the first definitive erythroblasts in the fetal liver arise from yolk sac erythroid/myeloid progenitors,36  although this remains to be demonstrated directly. It has been proposed that erythroid/myeloid progenitor-derived definitive erythropoiesis bridges the transition between primitive and HSC-derived erythropoiesis.2,36  It is tempting to speculate that the first hepatic colonization described for the human fetus (Figure 1B) might correspond to the second wave of hematopoiesis identified in the mouse embryo.

Definitive HSCs arise in the mouse embryo in the para-aortic splanchnopleure (E7.5-E9.5), the aorta-gonad-mesonephros region (E10.5-E11.5), the major blood vessels, and in the placenta.1,2,9  The HSCs that form in these tissues do not differentiate there but seed the developing fetal liver, thymus, and bone marrow, presumably as niches form and acquire the ability to support HSC self-renewal.1,2,9  The final wave of definitive hematopoiesis produces all lineages, including B and T lymphoid potentials.1,2 

Development of the hematopoietic system in humans is less well studied but is broadly similar to that of the mouse.3,9  In the human embryo, erythroid burst-forming unit activity is first detected at 7 to 8 weeks, after the second wave of colonization of the fetal liver by HSCs3  (Figure 1B). Fetal and adult human erythrocytes can be distinguished by their production of distinct forms of hemoglobin (hemoglobin F or A, respectively).34  The switch in expression of fetal to adult β-like globin genes begins at approximately 32 weeks and is completed after birth.34  In the human embryo, as in the mouse, HSCs have been identified in the aorta-gonad-mesonephros region, large vessels, and placenta.65 

Little is known about the growth factor requirements for primitive erythropoiesis in vivo. The hormone erythropoietin plays an essential role in definitive erythropoiesis, providing a key survival signal for late progenitors (colony-forming units-E).66  Mouse embryos deficient for the erythropoietin receptor have measurable but somewhat late defects in the proliferation and maturation of primitive erythroblasts but not progenitors, becoming evident only around E10.5 and later.67  Embryonic death from severe anemia occurs by E13.5, with a nearly complete ablation of definitive erythropoiesis.67,68  The differential effects of loss of erythropoietin receptor signaling in the 2 lineages suggests a role for additional cytokines in EryP development.37 

Stem cell factor (or kit ligand) also has a differential effect on EryP and EryD in mouse. Stem cell factor receptor (c-kit) mutants die of severe anemia because of a failure in definitive erythropoiesis.69  Although c-kit is expressed on the surface of EryP progenitors,12,45  their proliferation, survival, and maturation are unaffected by exogenous stem cell factor in culture.12  EryP progenitors express another receptor tyrosine kinase receptor, Tie-2,12,45  but their growth was not consistently stimulated by addition of a recombinant Tie2 ligand, angiopoietin-1.12  It is possible that functions for c-kit and/or Tie-2 signaling could be unmasked by development of serum-free culture conditions for EryP progenitors.12 

The TGF-β pathway has been implicated in embryonic erythroid development. On certain genetic backgrounds, embryos deficient for TGF-β1 die by midgestation from hematopoietic and vascular defects70 ; this phenotype is regulated by a genetic modifier.71  It is not known whether the hematopoietic defects observed70  reflect abnormalities in the formation and/or proliferation of EryP progenitors or their maturation. Targeted mutation of the Tgfbr1 receptor gene also results in embryonic lethality, but in this case it was concluded that the defect involved angiogenesis and that hematopoiesis was normal.72  However, the mutant embryos were clearly anemic and EryP progenitor assays were not performed.72  Deletion of Tgfbr2 also resulted in hematopoietic and vascular defects in the yolk sac.73  These embryos clearly had greatly reduced numbers of EryP in the vessels of sectioned tissue, but no further analysis was performed. Findings from our gene-expression profiling study prompted us to reevaluate the possibility of a role for TGF-β signaling in the primitive erythroid lineage. EryP progenitors express mRNAs for receptors (Tgfbr1 and Tgfbr3, Acvr2b) and downstream signaling components (eg, Smad3, Smad4, and Smad5) of these pathways; transcription of these genes begins to decrease by E8.5.12  To determine whether the TGF-β signaling pathway can modulate the expansion of EryP progenitors, we tested recombinant forms of the 3 Tgfbr1 ligands TGF-β1, TGF-β2, and TGF-β3 in clonogenic assays. TGF-β1 (but not TGF-β2 or TGF-β3) showed a dose-dependent effect on EryP progenitor activity in vitro: growth of EryP progenitors was inhibited at a low concentration and stimulated at higher concentrations of this cytokine.12  The TGF-β signaling pathway may function via an autocrine or paracrine mechanism, as both the receptor and its ligand, Tgfβ1, are transcribed in EryP.12  In contrast, TGF-β1 has been viewed as an inhibitor of definitive erythropoiesis in vitro, decreasing proliferation of progenitors and inducing their differentiation.74  A more detailed evaluation of the function of this pathway in EryP is warranted.

Fibroblast growth factor receptor-1 is required for hematopoietic development in differentiating mouse ES cells75  but whether the absence of the receptor affects primitive and/or definitive erythroid lineages was not defined. Sprouty (Spry) proteins antagonize signaling by receptor tyrosine kinases, such as fibroblast growth factors and epidermal growth factor.76  A recent study concluded that Spry1 negatively regulates primitive erythropoiesis in mouse embryos, but EryP progenitor assays were not performed77  and the cellular basis for the defect needs to be clarified.

EryP must adapt to changes in oxygen and nutrient supplies during their maturation within successive microenvironments (yolk sac, circulation, and fetal liver) within the embryo.42  The expression profile of EryP is consistent with high glycolytic activity in progenitors, followed by a metabolic switch as a group of genes known to play crucial roles in glucose metabolism are rapidly down-regulated around the time when these cells begin to circulate.12  In culture, not only are EryP progenitor numbers increased under conditions of low oxygen but the colonies that form in methylcellulose are also larger, apparently because of increased EryP-CFC proliferation.12  EryP progenitors also display a signature characteristic of the “Warburg effect” associated with rapidly proliferating cells (eg, cancer cells) that catabolize glucose aerobically and produce high levels of lactate in the cytosol, rather than using the more energy efficient mitochondrial pathway.78,79  We propose that EryP switch from aerobic glycolysis to mitochondrial oxidation as they enter the circulation. The rapid proliferation of EryP progenitors and their cellular response to hypoxia are reminiscent of stress erythropoiesis in the fetus and adult.80,81  It is not known whether the aerobic glycolytic profile of EryP progenitors simply reflects the particular energy demands of these rapidly dividing cells or is a more unique feature of primitive erythropoiesis.12 

The specification and differentiation of erythroid cells have been well studied for the definitive lineage and are regulated at transcriptional, posttranscriptional, and epigenetic levels. This subject has been extensively reviewed by others.1,82-85  In this section, we emphasize key points and insights from the recent literature. The zinc finger protein GATA1 was one of the first cell type-restricted transcription factors to be identified and cloned for any lineage.1  Since that time, a battery of additional erythroid regulators have been discovered; the reader is referred to excellent “primers” on core erythroid transcription factors (notably, GATA1, EKLF/KLF1, NFE2, and SCL) and cofactors (FOG1, CBP/p300, TRAP200, BRG1, and others).82-84 

Genome-wide expression profiling revealed that transcription in maturing EryP is characterized by 2 discrete waves that correlate with key developmental hallmarks.12  GATA1 and EKLF are central players in both primitive and definitive erythropoiesis. GATA1 and GATA2 overlap in function in primitive erythropoiesis, as demonstrated by analysis of double-knockout embryos.86  EKLF was initially thought to function primarily, if not exclusively, in definitive erythropoiesis,87,88  although its expression had been detected in mouse embryos as early as E7.5.89  It was later appreciated that Eklf functions in primitive erythropoiesis as well.53,90  We were able to analyze the impact of loss of one or both alleles of Eklf during primitive erythropoiesis by crossing the ε-globin:H2B-GFP transgenic mouse42  onto an Eklf mutant background.88  Haploinsufficiency of Eklf had a profound effect on the surface phenotype of EryP but not EryD, suggesting that the differentiation of EryP may be more sensitive to gene dosage than that of EryD.39  Indeed, the levels of Eklf are approximately 3-fold lower in EryP than in EryD.91  However, fetal-to-adult globin gene switching is also sensitive to Eklf gene dosage91,92 : haploinsufficiency of EKLF in humans causes hereditary persistence of fetal hemoglobin.92 

EryP and EryD differ in their requirements for a variety of other transcriptional regulators. For example, in contrast with fetal liver erythropoiesis, which is dependent on the function of the proto-oncogene c-Myb, the core binding factor Ruxn1, and the Kruppel-type zinc finger protein ZBP-89, primitive erythropoiesis is not.93-96  However, although it is not crucial for primitive erythropoiesis, Runx1 does function in the development of EryP.97  The ε-globin:H2B-GFP transgenic mouse42  should be useful in identifying functions for additional transcription factors that may not be essential in EryP. Conversely, lineage-specific ablation of genes in EryP will permit the identification of genes that are critical for primitive but not definitive erythropoiesis.

Both the primitive and definitive erythroid and megakaryocyte lineages share a common progenitor (megakaryocyte-erythroid progenitor).98 Eklf had been shown to play a dual role in definitive hematopoiesis by promoting erythropoiesis and suppressing megakaryopoiesis.98,99  We found that null mutation of Eklf resulted in a partial “identity crisis”39  within maturing EryP in the circulation, with up-regulation of a subset of megakaryocyte-related surface proteins, such as CD41 and Pecam-1/CD31, and transcription factor genes, such as Fli-1.39  Therefore, loss of Eklf in EryP causes a partial change in cellular identity.39  Additional observations12  suggest that Eklf not only regulates lineage divergence from the megakaryocyte-erythroid progenitor but also controls various aspects of EryP maturation.12  Findings such as these underscore the utility of the ε-globin–H2B-GFP transgenic mouse line42  for evaluating the effects of targeted mutations specifically in the primitive erythroid lineage.

It is now appreciated that not only the absolute and relative levels of these proteins but also their order of action is critical in the segregation of erythroid and megakaryocytic lineages as well as in their differentiation.1,84,100,101  For example, conditional deletion of Gata1 and Zfpm1 (which encodes FOG1) revealed that FOG1 and GATA1 function sequentially in the formation and lineage divergence of erythroid-megakaryocytic progenitors during definitive hematopoiesis.101  GATA1 and EKLF form large complexes with many other proteins to regulate the expression of erythroid-specific genes.102,103 

ChIP-Seq, in which chromatin immunoprecipitation is coupled with high-throughput DNA sequencing, has made it possible to generate genome-wide maps of binding by transcription factors, such as GATA1, GATA2, and NF-E2,104,105  and thereby define the full repertoire of loci occupied by these factors. Advances in this technology have allowed the identification of much wider combinatorial interactions among transcription factors than was previously possible.102,103  Although detailed analysis of individual transcription factors has provided a wealth of information about their functions in erythroid cells, there has been increasing emphasis on their coordinated binding. It would be of interest to compare the transcription complexes that form at different stages of EryP development (Figure 4) with those found in differentiating EryD.

In conclusion, although erythropoiesis has traditionally been described as encompassing embryonic (primitive) and “adult-type” (definitive) phases, it now appears that there are not 2, but 3 distinct erythroid lineages, each with a unique pattern of β-globin gene expression.36  Thus, the erythroid burst-forming units that arise from HSCs in the fetal liver actually constitute a third wave of erythropoiesis. In addition, at least in the mouse, switching in globin gene expression during development reflects maturational switches within a single erythroid lineage36,40  (Figure 4) as well as lineage switching (Figure 1). The progenitors that produce EryP are the first hematopoietic cells of the embryo and are found in the yolk sac only briefly but terminally differentiated primitive erythrocytes are present in the circulation through the end of gestation and probably for a short time after birth13  (primitive erythropoiesis is summarized in Figure 7). The ontogeny of erythropoiesis is much more complex than previously appreciated. It will be important to take this complexity into consideration when interpreting observations made not only in the embryo/fetus but in cell culture systems, such as ES and iPS cells. Comparative studies of erythropoiesis at different stages of development, in different tissues within the same stage, and in different species will probably continue to lead to refinements in our understanding of the biology of the red blood cell lineage.

Some of the pathways involved in the maturation of EryP and EryD are probably conserved. Surprisingly little is known about how red blood cells enucleate, even in the adult. Similarly, the mechanisms underlying the redistribution of proteins on the surface of erythroid cells as they mature (Figure 6) have barely been explored. Comparisons between primitive and definitive erythropoiesis may provide useful insights into these processes, our understanding of which remains largely descriptive. Elucidation of the common as well as the distinguishing features of embryonic versus adult erythroid development will be a prerequisite for the directed differentiation of human stem or progenitor cells for therapeutic purposes and for the efficient production of pure populations of red blood cells for transfusion in patients. The study of red blood cells will probably continue to provide fascinating insights into fundamental questions in biology and information of considerable practical value.

This work was supported in part by the National Institutes of Health (grants RO1 HL62248, DK52191, and EB02209), the Roche Foundation for Anemia Research (grant 9699367999, cycle X), and the New York State Department of Health (NYSTEM grant N08G-024, M.H.B.).

National Institutes of Health

Contribution: M.H.B. created figures and wrote the paper; J.I. created figures; and S.T.F. wrote an initial draft of the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

The current affiliation for J.I. is Cardiovascular Development and Repair Department, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain. The current affiliation for S.T.F. is Disciplines of Physiology, Anatomy and Histology, School of Medical Sciences, University of Sydney, Camperdown NSW, Australia.

Correspondence: Margaret H. Baron, Mount Sinai School of Medicine, Box 1079, 1468 Madison Ave, Annenberg 24-04E, New York, NY 10029-6574; e-mail: margaret.baron@mssm.edu.

1
Orkin
 
SH
Zon
 
LI
Hematopoiesis: an evolving paradigm for stem cell biology.
Cell
2008
, vol. 
132
 
4
(pg. 
631
-
644
)
2
Dzierzak
 
E
Speck
 
NA
Of lineage and legacy: the development of mammalian hematopoietic stem cells.
Nat Immunol
2008
, vol. 
9
 
2
(pg. 
129
-
136
)
3
Tavian
 
M
Peault
 
B
Embryonic development of the human hematopoietic system.
Int J Dev Biol
2005
, vol. 
49
 
2
(pg. 
243
-
250
)
4
Baron
 
MH
Early patterning of the mouse embryo: implications for hematopoietic commitment and differentiation.
Exp Hematol
2005
, vol. 
33
 
9
(pg. 
1015
-
1020
)
5
Ferkowicz
 
MJ
Yoder
 
MC
Blood island formation: longstanding observations and modern interpretations.
Exp Hematol
2005
, vol. 
33
 
9
(pg. 
1041
-
1047
)
6
Tober
 
J
Koniski
 
A
McGrath
 
KE
, et al. 
The megakaryocyte lineage originates from hemangioblast precursors and is an integral component both of primitive and of definitive hematopoiesis.
Blood
2007
, vol. 
109
 
4
(pg. 
1433
-
1441
)
7
Palis
 
J
Robertson
 
S
Kennedy
 
M
Wall
 
C
Keller
 
G
Development of erythroid and myeloid progenitors in the yolk sac and embryo proper of the mouse.
Development
1999
, vol. 
126
 
22
(pg. 
5073
-
5084
)
8
Bertrand
 
JY
Jalil
 
A
Klaine
 
M
Jung
 
S
Cumano
 
A
Godin
 
I
Three pathways to mature macrophages in the early mouse yolk sac.
Blood
2005
, vol. 
106
 
9
(pg. 
3004
-
3011
)
9
Lee
 
LK
Ueno
 
M
Van Handel
 
B
Mikkola
 
HK
Placenta as a newly identified source of hematopoietic stem cells.
Curr Opin Hematol
2010
, vol. 
17
 
4
(pg. 
313
-
318
)
10
Kumaravelu
 
P
Hook
 
L
Morrison
 
AM
, et al. 
Quantitative developmental anatomy of definitive haematopoietic stem cells/long-term repopulating units (HSC/RUs): role of the aorta-gonad-mesonephros (AGM) region and the yolk sac in colonisation of the mouse embryonic liver.
Development
2002
, vol. 
129
 
21
(pg. 
4891
-
4899
)
11
Cumano
 
A
Godin
 
I
Ontogeny of the hematopoietic system.
Annu Rev Immunol
2007
, vol. 
25
 (pg. 
745
-
785
)
12
Isern
 
J
He
 
Z
Fraser
 
ST
, et al. 
Single lineage transcriptome analysis reveals key regulatory pathways in primitive erythroid progenitors in the mouse embryo.
Blood
2011
, vol. 
117
 
18
(pg. 
4924
-
4934
)
13
Fraser
 
ST
Isern
 
J
Baron
 
MH
Maturation and enucleation of primitive erythroblasts is accompanied by changes in cell surface antigen expression patterns during mouse embryogenesis.
Blood
2007
, vol. 
109
 
1
(pg. 
343
-
352
)
14
Kingsley
 
PD
Malik
 
J
Fantauzzo
 
KA
Palis
 
J
Yolk sac-derived primitive erythroblasts enucleate during mammalian embryogenesis.
Blood
2004
, vol. 
104
 
1
(pg. 
19
-
25
)
15
Lensch
 
MW
Daley
 
GQ
Origins of mammalian hematopoiesis: in vivo paradigms and in vitro models.
Curr Top Dev Biol
2004
, vol. 
60
 (pg. 
127
-
196
)
16
Arnold
 
SJ
Robertson
 
EJ
Making a commitment: cell lineage allocation and axis patterning in the early mouse embryo.
Nat Rev Mol Cell Biol
2009
, vol. 
10
 
2
(pg. 
91
-
103
)
17
Belaoussoff
 
M
Farrington
 
SM
Baron
 
MH
Hematopoietic induction and respecification of A-P identity by visceral endoderm signaling in the mouse embryo.
Development
1998
, vol. 
125
 
24
(pg. 
5009
-
5018
)
18
Dyer
 
MA
Farrington
 
SM
Mohn
 
D
Munday
 
JR
Baron
 
MH
Indian hedgehog activates hematopoiesis and vasculogenesis and can respecify prospective neurectodermal cell fate in the mouse embryo.
Development
2001
, vol. 
128
 
10
(pg. 
1717
-
1730
)
19
Baron
 
MH
Induction of embryonic hematopoietic and endothelial stem/progenitor cells by hedgehog-mediated signals.
Differentiation
2001
, vol. 
68
 
4
(pg. 
175
-
185
)
20
Kunath
 
T
Arnaud
 
D
Uy
 
GD
, et al. 
Imprinted X-inactivation in extra-embryonic endoderm cell lines from mouse blastocysts.
Development
2005
, vol. 
132
 
7
(pg. 
1649
-
1661
)
21
Artus
 
J
Douvaras
 
P
Piliszek
 
A
Isern
 
J
Baron
 
MH
Hadjantonakis
 
A-K
BMP signalling directs primitive endoderm-derived XEN cells to an extraembryonic visceral endoderm identity.
Dev Biol
2012
, vol. 
361
 
2
(pg. 
245
-
262
)
22
Nostro
 
MC
Cheng
 
X
Keller
 
GM
Gadue
 
P
Wnt, activin, and BMP signaling regulate distinct stages in the developmental pathway from embryonic stem cells to blood.
Cell Stem Cell
2008
, vol. 
2
 
1
(pg. 
60
-
71
)
23
Cheng
 
X
Huber
 
TL
Chen
 
VC
Gadue
 
P
Keller
 
GM
Numb mediates the interaction between Wnt and Notch to modulate primitive erythropoietic specification from the hemangioblast.
Development
2008
, vol. 
135
 
20
(pg. 
3447
-
3458
)
24
Choi
 
K
Kennedy
 
M
Kazarov
 
A
Papadimitriou
 
JC
Keller
 
G
A common precursor for hematopoietic and endothelial cells.
Development
1998
, vol. 
125
 
4
(pg. 
725
-
732
)
25
Zambidis
 
ET
Peault
 
B
Park
 
TS
Bunz
 
F
Civin
 
CI
Hematopoietic differentiation of human embryonic stem cells progresses through sequential hematoendothelial, primitive, and definitive stages resembling human yolk sac development.
Blood
2005
, vol. 
106
 
3
(pg. 
860
-
870
)
26
Huber
 
TL
Kouskoff
 
V
Fehling
 
HJ
Palis
 
J
Keller
 
G
Haemangioblast commitment is initiated in the primitive streak of the mouse embryo.
Nature
2004
, vol. 
432
 
7017
(pg. 
625
-
630
)
27
Ema
 
M
Faloon
 
P
Zhang
 
WJ
, et al. 
Combinatorial effects of Flk1 and Tal1 on vascular and hematopoietic development in the mouse.
Genes Dev
2003
, vol. 
17
 
3
(pg. 
380
-
393
)
28
Kinder
 
SJ
Tsang
 
TE
Quinlan
 
GA
Hadjantonakis
 
AK
Nagy
 
A
Tam
 
PP
The orderly allocation of mesodermal cells to the extraembryonic structures and the anteroposterior axis during gastrulation of the mouse embryo.
Development
1999
, vol. 
126
 
21
(pg. 
4691
-
4701
)
29
Ueno
 
H
Weissman
 
IL
Clonal analysis of mouse development reveals a polyclonal origin for yolk sac blood islands.
Dev Cell
2006
, vol. 
11
 
4
(pg. 
519
-
533
)
30
Furuta
 
C
Ema
 
H
Takayanagi
 
S
, et al. 
Discordant developmental waves of angioblasts and hemangioblasts in the early gastrulating mouse embryo.
Development
2006
, vol. 
133
 
14
(pg. 
2771
-
2779
)
31
Lancrin
 
C
Sroczynska
 
P
Stephenson
 
C
Allen
 
T
Kouskoff
 
V
Lacaud
 
G
The haemangioblast generates haematopoietic cells through a haemogenic endothelium stage.
Nature
2009
, vol. 
457
 
7231
(pg. 
892
-
895
)
32
Eilken
 
HM
Nishikawa
 
S
Schroeder
 
T
Continuous single-cell imaging of blood generation from haemogenic endothelium.
Nature
2009
, vol. 
457
 
7231
(pg. 
896
-
900
)
33
Iacovino
 
M
Chong
 
D
Szatmari
 
I
, et al. 
HoxA3 is an apical regulator of haemogenic endothelium.
Nat Cell Biol
2011
, vol. 
13
 
1
(pg. 
72
-
78
)
34
Sankaran
 
VG
Xu
 
J
Orkin
 
SH
Advances in the understanding of haemoglobin switching.
Br J Haematol
2010
, vol. 
149
 
2
(pg. 
181
-
194
)
35
He
 
Z
Russell
 
JE
Expression, purification, and characterization of human hemoglobins Gower-1 (zeta(2)epsilon(2)), Gower-2 (alpha(2)epsilon(2)), and Portland-2 (zeta(2)beta(2)) assembled in complex transgenic-knockout mice.
Blood
2001
, vol. 
97
 
4
(pg. 
1099
-
1105
)
36
McGrath
 
KE
Frame
 
JM
Fromm
 
GJ
, et al. 
A transient definitive erythroid lineage with unique regulation of the beta-globin locus in the mammalian embryo.
Blood
2011
, vol. 
117
 
17
(pg. 
4600
-
4608
)
37
Palis
 
J
Malik
 
J
McGrath
 
KE
Kingsley
 
PD
Primitive erythropoiesis in the mammalian embryo.
Int J Dev Biol
2010
, vol. 
54
 
6
(pg. 
1011
-
1018
)
38
Morioka
 
K
Minamikawa-Tachino
 
R
Temporal characteristics of the differentiation of embryonic erythroid cells in fetal peripheral blood of the Syrian hamster.
Dev Growth Differ
1993
, vol. 
35
 (pg. 
569
-
582
)
39
Isern
 
J
Fraser
 
ST
He
 
Z
Zhang
 
H
Baron
 
MH
Dose-dependent regulation of primitive erythroid maturation and identity by the transcription factor Eklf.
Blood
2010
, vol. 
116
 
19
(pg. 
3972
-
3980
)
40
Kingsley
 
PD
Malik
 
J
Emerson
 
RL
, et al. 
“Maturational” globin switching in primary primitive erythroid cells.
Blood
2006
, vol. 
107
 
4
(pg. 
1665
-
1672
)
41
Murry
 
CE
Keller
 
G
Differentiation of embryonic stem cells to clinically relevant populations: lessons from embryonic development.
Cell
2008
, vol. 
132
 
4
(pg. 
661
-
680
)
42
Isern
 
J
Fraser
 
ST
He
 
Z
Baron
 
MH
The fetal liver is a niche for maturation of primitive erythroid cells.
Proc Natl Acad Sci U S A
2008
, vol. 
105
 
18
(pg. 
6662
-
6667
)
43
McGrath
 
KE
Koniski
 
AD
Malik
 
J
Palis
 
J
Circulation is established in a stepwise pattern in the mammalian embryo.
Blood
2003
, vol. 
101
 
5
(pg. 
1669
-
1676
)
44
Wong
 
PM
Chung
 
SW
Reicheld
 
SM
Chui
 
DH
Hemoglobin switching during murine embryonic development: evidence for two populations of embryonic erythropoietic progenitor cells.
Blood
1986
, vol. 
67
 
3
(pg. 
716
-
721
)
45
Ema
 
M
Yokomizo
 
T
Wakamatsu
 
A
Terunuma
 
T
Yamamoto
 
M
Takahashi
 
S
Primitive erythropoiesis from mesodermal precursors expressing VE-cadherin, PECAM-1, Tie2, endoglin, and CD34 in the mouse embryo.
Blood
2006
, vol. 
108
 
13
(pg. 
4018
-
4024
)
46
Ferkowicz
 
MJ
Starr
 
M
Xie
 
X
, et al. 
CD41 expression defines the onset of primitive and definitive hematopoiesis in the murine embryo.
Development
2003
, vol. 
130
 
18
(pg. 
4393
-
4403
)
47
Fraser
 
ST
Isern
 
J
Baron
 
MH
Wasserman
 
P
The use of transgenic fluorescent reporter mouse lines to monitor hematopoietic and erythroid development during embryogenesis.
Guide to Techniques in Mouse Development: Methods in Enzymology
2010
, vol. 
Vol 476
 
Pasadena, CA
Elsevier Press
(pg. 
403
-
427
)
48
Haar
 
J
Ackerman
 
GA
A phase and electron microscopic study of vasculogenesis and erythropoiesis in the yolk sac of the mouse.
Anat Rec
1971
, vol. 
170
 
2
(pg. 
199
-
224
)
49
Zhang
 
J
Socolovsky
 
M
Gross
 
AW
Lodish
 
HF
Role of Ras signaling in erythroid differentiation of mouse fetal liver cells: functional analysis by a flow cytometry-based novel culture system.
Blood
2003
, vol. 
102
 
12
(pg. 
3938
-
3946
)
50
Geiduschek
 
JB
Singer
 
SJ
Molecular changes in the membranes of mouse erythroid cells accompanying differentiation.
Cell
1979
, vol. 
16
 
1
(pg. 
149
-
163
)
51
Lee
 
JC
Gimm
 
JA
Lo
 
AJ
, et al. 
Mechanism of protein sorting during erythroblast enucleation: role of cytoskeletal connectivity.
Blood
2004
, vol. 
103
 
5
(pg. 
1912
-
1919
)
52
Yoshida
 
H
Kawane
 
K
Koike
 
M
Mori
 
Y
Uchiyama
 
Y
Nagata
 
S
Phosphatidylserine-dependent engulfment by macrophages of nuclei from erythroid precursor cells.
Nature
2005
, vol. 
437
 
7059
(pg. 
754
-
758
)
53
Drissen
 
R
von Lindern
 
M
Kolbus
 
A
, et al. 
The erythroid phenotype of EKLF-null mice: defects in hemoglobin metabolism and membrane stability.
Mol Cell Biol
2005
, vol. 
25
 
12
(pg. 
5205
-
5214
)
54
Bieker
 
JJ
An unexpected entry into the globin real estate market.
Blood
2005
, vol. 
106
 
7
(pg. 
2230
-
2231
)
55
McGrath
 
KE
Kingsley
 
PD
Koniski
 
AD
Porter
 
RL
Bushnell
 
TP
Palis
 
J
Enucleation of primitive erythroid cells generates a transient population of “pyrenocytes” in the mammalian fetus.
Blood
2008
, vol. 
111
 
4
(pg. 
2409
-
2417
)
56
Chasis
 
JA
Mohandas
 
N
Erythroblastic islands: niches for erythropoiesis.
Blood
2008
, vol. 
112
 
3
(pg. 
470
-
478
)
57
Van Handel
 
B
Prashad
 
SL
Hassanzadeh-Kiabi
 
N
, et al. 
The first trimester human placenta is a site for terminal maturation of primitive erythroid cells.
Blood
2010
, vol. 
116
 
17
(pg. 
3321
-
3330
)
58
Spike
 
BT
Dibling
 
BC
Macleod
 
KF
Hypoxic stress underlies defects in erythroblast islands in the Rb-null mouse.
Blood
2007
, vol. 
110
 
6
(pg. 
2173
-
2181
)
59
Giarratana
 
MC
Kobari
 
L
Lapillonne
 
H
, et al. 
Ex vivo generation of fully mature human red blood cells from hematopoietic stem cells.
Nat Biotechnol
2005
, vol. 
23
 
1
(pg. 
69
-
74
)
60
Rhodes
 
MM
Kopsombut
 
P
Bondurant
 
MC
Price
 
JO
Koury
 
MJ
Adherence to macrophages in erythroblastic islands enhances erythroblast proliferation and increases erythrocyte production by a different mechanism than erythropoietin.
Blood
2008
, vol. 
111
 
3
(pg. 
1700
-
1708
)
61
Ema
 
H
Nakauchi
 
H
Expansion of hematopoietic stem cells in the developing liver of a mouse embryo.
Blood
2000
, vol. 
95
 
7
(pg. 
2284
-
2288
)
62
Palis
 
J
Chan
 
RJ
Koniski
 
A
Patel
 
R
Starr
 
M
Yoder
 
MC
Spatial and temporal emergence of high proliferative potential hematopoietic precursors during murine embryogenesis.
Proc Natl Acad Sci U S A
2001
, vol. 
98
 
8
(pg. 
4528
-
4533
)
63
England
 
SJ
McGrath
 
KE
Frame
 
JM
Palis
 
J
Immature erythroblasts with extensive ex vivo self-renewal capacity emerge from the early mammalian fetus.
Blood
2011
, vol. 
117
 
9
(pg. 
2708
-
2717
)
64
Yoder
 
MC
Hiatt
 
K
Engraftment of embryonic hematopoietic cells in conditioned newborn recepients.
Blood
1997
, vol. 
89
 
6
(pg. 
2176
-
2183
)
65
Ivanovs
 
A
Rybtsov
 
S
Welch
 
L
Anderson
 
RA
Turner
 
ML
Medvinsky
 
A
Highly potent human hematopoietic stem cells first emerge in the intraembryonic aorta-gonad-mesonephros region.
J Exp Med
2011
, vol. 
208
 
12
(pg. 
2417
-
2427
)
66
Lodish
 
H
Flygare
 
J
Chou
 
S
From stem cell to erythroblast: regulation of red cell production at multiple levels by multiple hormones.
IUBMB Life
2010
, vol. 
62
 
7
(pg. 
492
-
496
)
67
Lin
 
C-S
Lim
 
S-K
D'Agati
 
V
Costantini
 
F
Differential effects of an erythropoietin receptor gene disruption on primitive and definitive erythropoiesis.
Genes Dev
1996
, vol. 
10
 
2
(pg. 
154
-
164
)
68
Wu
 
H
Liu
 
X
Jaenisch
 
R
Lodish
 
HF
Generation of committed erythroid BFU-E and CFU-E progenitors does not require erythropoietin or the erythropoietin receptor.
Cell
1995
, vol. 
83
 
1
(pg. 
59
-
67
)
69
Metcalf
 
D
Hematopoietic cytokines.
Blood
2008
, vol. 
111
 
2
(pg. 
485
-
491
)
70
Dickson
 
MC
Martin
 
JS
Cousins
 
FM
Kulkarni
 
AB
Karlsson
 
S
Akhurst
 
RJ
Defective haematopoiesis and vasculogenesis in transforming growth factor-beta1 knock out mice.
Development
1995
, vol. 
121
 
6
(pg. 
1845
-
1854
)
71
Bonyadi
 
M
Rusholme
 
SA
Cousins
 
FM
, et al. 
Mapping of a major genetic modifier of embryonic lethality in TGF-beta 1 knockout mice.
Nat Genet
1997
, vol. 
15
 
2
(pg. 
207
-
211
)
72
Larsson
 
J
Goumans
 
MJ
Sjostrand
 
LJ
, et al. 
Abnormal angiogenesis but intact hematopoietic potential in TGF-beta type I receptor-deficient mice.
EMBO J
2001
, vol. 
20
 
7
(pg. 
1663
-
1673
)
73
Oshima
 
M
Oshima
 
H
Taketo
 
MM
TGF-β receptor type II deficiency results in defects of yolk sac hematopoiesis and vasculogenesis.
Dev Biol
1996
, vol. 
179
 
1
(pg. 
297
-
302
)
74
Carotta
 
S
Pilat
 
S
Mairhofer
 
A
, et al. 
Directed differentiation and mass cultivation of pure erythroid progenitors from mouse embryonic stem cells.
Blood
2004
, vol. 
104
 
6
(pg. 
1873
-
1880
)
75
Magnusson
 
PU
Ronca
 
R
Dell'Era
 
P
, et al. 
Fibroblast growth factor receptor-1 expression is required for hematopoietic but not endothelial cell development.
Arterioscler Thromb Vasc Biol
2005
, vol. 
25
 
5
(pg. 
944
-
949
)
76
Edwin
 
F
Anderson
 
K
Ying
 
C
Patel
 
TB
Intermolecular interactions of Sprouty proteins and their implications in development and disease.
Mol Pharmacol
2009
, vol. 
76
 
4
(pg. 
679
-
691
)
77
Yang
 
X
Gong
 
Y
Friesel
 
R
Spry1 is expressed in hemangioblasts and negatively regulates primitive hematopoiesis and endothelial cell function.
PLoS One
2011
, vol. 
6
 
4
pg. 
e18374
 
78
Vander Heiden
 
MG
Cantley
 
LC
Thompson
 
CB
Understanding the Warburg effect: the metabolic requirements of cell proliferation.
Science
2009
, vol. 
324
 
5930
(pg. 
1029
-
1033
)
79
Levine
 
AJ
Puzio-Kuter
 
AM
The control of the metabolic switch in cancers by oncogenes and tumor suppressor genes.
Science
2010
, vol. 
330
 
6009
(pg. 
1340
-
1344
)
80
Porayette
 
P
Paulson
 
RF
BMP4/Smad5 dependent stress erythropoiesis is required for the expansion of erythroid progenitors during fetal development.
Dev Biol
2008
, vol. 
317
 
1
(pg. 
24
-
35
)
81
Paulson
 
RF
Shi
 
L
Wu
 
DC
Stress erythropoiesis: new signals and new stress progenitor cells.
Curr Opin Hematol
2011
, vol. 
18
 
3
(pg. 
139
-
145
)
82
Kim
 
SI
Bresnick
 
EH
Transcriptional control of erythropoiesis: emerging mechanisms and principles.
Oncogene
2007
, vol. 
26
 
47
(pg. 
6777
-
6794
)
83
Tsiftsoglou
 
AS
Vizirianakis
 
IS
Strouboulis
 
J
Erythropoiesis: model systems, molecular regulators, and developmental programs.
IUBMB Life
2009
, vol. 
61
 
8
(pg. 
800
-
830
)
84
Dore
 
LC
Crispino
 
JD
Transcription factor networks in erythroid cell and megakaryocyte development.
Blood
2011
, vol. 
118
 
2
(pg. 
231
-
239
)
85
Hattangadi
 
S
Wong
 
P
Zhang
 
L
Flygare
 
J
Lodish
 
HF
From stem cell to red cell: regulation of erythropoiesis at multiple levels by multiple proteins, RNAs and chromatin modifications.
Blood
2011
, vol. 
118
 
24
(pg. 
6258
-
6268
)
86
Fujiwara
 
Y
Chang
 
AN
Williams
 
AM
Orkin
 
SH
Functional overlap of GATA-1 and GATA-2 in primitive hematopoietic development.
Blood
2004
, vol. 
103
 
2
(pg. 
583
-
585
)
87
Nuez
 
B
Michalovich
 
D
Bygrave
 
A
Ploemacher
 
R
Grosveld
 
F
Defective haematopoiesis in fetal liver resulting from inactivation of the EKLF gene.
Nature
1995
, vol. 
375
 
6529
(pg. 
316
-
318
)
88
Perkins
 
AC
Sharpe
 
AH
Orkin
 
SH
Lethal beta-thalas-saemia in mice lacking the erythroid CACCC-transcription factor EKLF.
Nature
1995
, vol. 
375
 
6529
(pg. 
318
-
322
)
89
Southwood
 
CM
Downs
 
KM
Bieker
 
JJ
Erythroid Kruppel-like factor exhibits an early and sequentially localized pattern of expression during mammalian erythroid ontogeny.
Dev Dyn
1996
, vol. 
206
 
3
(pg. 
248
-
259
)
90
Hodge
 
D
Coghill
 
E
Keys
 
J
, et al. 
A global role for EKLF in definitive and primitive erythropoiesis.
Blood
2006
, vol. 
107
 
8
(pg. 
3359
-
3370
)
91
Zhou
 
D
Liu
 
K
Sun
 
CW
Pawlik
 
KM
Townes
 
TM
KLF1 regulates BCL11A expression and gamma- to beta-globin gene switching.
Nat Genet
2010
, vol. 
42
 
9
(pg. 
742
-
744
)
92
Borg
 
J
Papadopoulos
 
P
Georgitsi
 
M
, et al. 
Haploinsufficiency for the erythroid transcription factor KLF1 causes hereditary persistence of fetal hemoglobin.
Nat Genet
2010
, vol. 
42
 
9
(pg. 
801
-
805
)
93
Mucenski
 
ML
McLain
 
K
Kier
 
AB
, et al. 
A functional c-myb gene is required for normal murine fetal hepatic hematopoiesis.
Cell
1991
, vol. 
65
 
4
(pg. 
677
-
689
)
94
Okuda
 
T
van Deursen
 
J
Hiebert
 
SW
Grosveld
 
G
Downing
 
JR
AML1, the target of multiple chromosomal translocations in human leukemia, is essential for normal fetal liver hematopoiesis.
Cell
1996
, vol. 
84
 
2
(pg. 
321
-
330
)
95
Wang
 
Q
Stacy
 
T
Binder
 
M
Marin-Padilla
 
M
Sharpe
 
AH
Speck
 
NA
Disruption of the Cbfa2 gene causes necrosis and hemorrhaging in the central nervous system and blocks definitive hematopoiesis.
Proc Natl Acad Sci U S A
1996
, vol. 
93
 
8
(pg. 
3444
-
3449
)
96
Woo
 
AJ
Moran
 
TB
Schindler
 
YL
, et al. 
Identification of ZBP-89 as a novel GATA-1-associated transcription factor involved in megakaryocytic and erythroid development.
Mol Cell Biol
2008
, vol. 
28
 
8
(pg. 
2675
-
2689
)
97
Yokomizo
 
T
Hasegawa
 
K
Ishitobi
 
H
, et al. 
Runx1 is involved in primitive erythropoiesis in the mouse.
Blood
2008
, vol. 
111
 
8
(pg. 
4075
-
4080
)
98
Siatecka
 
M
Bieker
 
JJ
The multifunctional role of EKLF/KLF1 during erythropoiesis.
Blood
2011
, vol. 
118
 
8
(pg. 
2044
-
2054
)
99
Frontelo
 
P
Manwani
 
D
Galdass
 
M
, et al. 
Novel role for EKLF in megakaryocyte lineage commitment.
Blood
2007
, vol. 
110
 
12
(pg. 
3871
-
3880
)
100
Graf
 
T
Enver
 
T
Forcing cells to change lineages.
Nature
2009
, vol. 
462
 
7273
(pg. 
587
-
594
)
101
Mancini
 
E
Sanjuan-Pla
 
A
Luciani
 
L
, et al. 
FOG-1 and GATA-1 act sequentially to specify definitive megakaryocytic and erythroid progenitors.
EMBO J
2011
, vol. 
31
 
2
(pg. 
351
-
365
)
102
Wilson
 
NK
Foster
 
SD
Wang
 
X
, et al. 
Combinatorial transcriptional control in blood stem/progenitor cells: genome-wide analysis of ten major transcriptional regulators.
Cell Stem Cell
2010
, vol. 
7
 
4
(pg. 
532
-
544
)
103
Soler
 
E
Andrieu-Soler
 
C
de Boer
 
E
, et al. 
The genome-wide dynamics of the binding of Ldb1 complexes during erythroid differentiation.
Genes Dev
2010
, vol. 
24
 
3
(pg. 
277
-
289
)
104
Steiner
 
LA
Maksimova
 
Y
Schulz
 
V
, et al. 
Chromatin architecture and transcription factor binding regulate expression of erythrocyte membrane protein genes.
Mol Cell Biol
2009
, vol. 
29
 
20
(pg. 
5399
-
5412
)
105
Fujiwara
 
T
O'Geen
 
H
Keles
 
S
, et al. 
Discovering hematopoietic mechanisms through genome-wide analysis of GATA factor chromatin occupancy.
Mol Cell
2009
, vol. 
36
 
4
(pg. 
667
-
681
)
Sign in via your Institution