<<

Ontogeny of erythropoiesis James Palis

University of Rochester Medical Center, Rochester, Purpose of review New York, USA The present study review examines the current understanding of the ontogeny of Correspondence to James Palis, MD, University of erythropoiesis with a focus on the emergence of the embryonic (primitive) erythroid Rochester Medical Center, Department of Pediatrics and Center for Pediatric Biomedical Research, Box lineage and on the similarities and differences between the primitive and the fetal/adult 703, 601 Elmwood Ave., Rochester, NY 14642, USA (definitive) forms of erythroid cell maturation. Tel: +1 585 275 5098; fax: +1 585 276 0232; e-mail: [email protected] Recent findings Primitive erythroid precursors in the mouse embryo and cultured in vitro from embryonic stem cells undergo ‘maturational’ globin switching as they differentiate Current Opinion in Hematology 2008, 15:155– 161 terminally. The appearance of a transient population of primitive ‘pyrenocytes’ (extruded nuclei) in the fetal bloodstream indicates that primitive erythroblasts enucleate by nuclear extrusion. In-vitro differentiation of human embryonic stem cells recapitulates hematopoietic ontogeny reminiscent of the murine yolk sac, including overlapping waves of hemangioblast, primitive, erythroid, and definitive erythroid progenitors. Definitive erythroid potential in zebrafish embryos, like that in mice, initially arises prior to, and independent of, hematopoietic emergence in the region of the aorta. Maturation of definitive erythroid cells within islands promotes erythroblast–erythroblast and erythroblast–stromal interactions that regulate red cell output. Summary The study of embryonic development in several different model systems, as well as in cultured human embryonic stem cells, continues to provide important insights into the ontogeny of erythropoiesis. Contrasting the similarities and differences between primitive and definitive erythropoiesis will lead to an improved understanding of erythroblast maturation and the terminal steps of erythroid differentiation.

Keywords definitive erythropoiesis, hemangioblast, primitive erythropoiesis, pyrenocyte

Curr Opin Hematol 15:155–161 ß 2008 Wolters Kluwer Health | Lippincott Williams & Wilkins 1065-6251

ination of embryonic and fetal cell morphology, Introduction however, revealed that primitive red cells undergo a The red cells of are unique in the synchronous wave of maturation in the bloodstream. Four kingdom as they circulate as enucleated cells. In contrast, years ago, it was discovered that late-stage primitive the fully mature red cells of , amphibians, and fish erythroblasts in the mouse embryo complete their matu- remain nucleated [1]. A century ago, examination of ration by enucleating and continuing to circulate for mammalian embryos revealed the presence of distinct several more days as erythrocytes [3]. The specification nucleated and enucleated red cells [2]. The continuous of hematopoiesis in mammalian embryos was discussed circulation of small, enucleated erythrocytes during fetal last in Current Opinion in Hematology 3 years ago [4]. Here, and postnatal life (‘definitive’ erythropoiesis) was distin- recent insights regarding the ontogeny of erythropoiesis guished from ‘primitive’ erythropoiesis, characterized by and the maturation of the primitive and definitive ery- the transient circulation of large, nucleated red cells that throid lineages will be reviewed. originate in the yolk sac. Similarly, it was recognized that primitive erythropoiesis originates in the yolk sac and intermediate cell mass of chicken and zebrafish embryos, Emergence of primitive erythropoiesis in the respectively. As mammalian primitive erythroblasts cir- early embryo culate as nucleated cells and are confined to the embryo, The initial generation of erythroid cells in the embryo they have been thought of as a ‘primitive’ form of of mammalian and nonmammalian organisms depends erythropoiesis that shares many characteristics with the on the formation of mesoderm cells that migrate through nucleated red cells of nonmammalian vertebrates. Exam- the primitive streak and contribute to the formation of

1065-6251 ß 2008 Wolters Kluwer Health | Lippincott Williams & Wilkins

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. 156 Erythroid system and its diseases

intraembryonic as well as extraembryonic structures such with, or soon after, the onset of cardiac contractions as the yolk sac and the placenta in mammals. Immature [16,17]. Over the next 8 days, primitive erythroid cells primitive erythroid cells rapidly pool into so-called blood mature in a synchronous cohort as they undergo changes islands soon after the start of gastrulation in the yolk sac of well recognized in maturing definitive erythroid precur- mammalian and avian embryos [5,6]. These blood islands sors, including a limited number of cell divisions, accu- become enveloped by endothelial cells, which form the mulation of increasing amounts of , nuclear initial vascular plexus of the yolk sac (reviewed by condensation, a progressive decrease in cell size, and Ferkowicz and Yoder, [7]). ultimately enucleation [3,18]. Many of these findings have recently been confirmed using a transgenic mouse The appearance of primitive erythroid cells and endo- expressing the enhanced green fluorescent protein thelial cells at the same time and place within the early (eGFP) in primitive erythroid cells [19]. Interestingly, conceptus has long suggested that these lineages share a circulating primitive erythroblasts express several adhe- common developmental origin. A unique blast colony- sion molecules that could mediate interactions with other forming cell (Blast-CFC) containing both hematopoietic cell types [19]. and endothelial cell potential has been identified in both cultured embryonic stem cells and mouse embryos [8,9]. Coincident with primitive erythroblast enucleation, the Consistent with the hypothesis that the first blood appearance of a transient population of very small, cells arise from a hemangioblast precursor, clonal studies nucleated cells with a rim of ey-globin-positive cyto- reveal that a small number of GATA1-expressing cells in plasm was noted in the circulation of mouse embryos blood islands of mouse yolk sacs have endothelial as well [20]. These cells are reminiscent of extruded nuclei as primitive erythroid cell potential [10]. Furthermore, [21], which are the product of enucleation of late-stage the transcription factors GATA2 and endoglin, both of erythroblasts (Fig. 1). The extruded nuclei from defini- which function in hematopoietic stem cells (HSC), have tive erythroid cells undergo rapid loss of the phosphati- each been shown to regulate hemangioblast precursors dylserine asymmetry of the cell membrane and are [11,12]. The close association of the primitive erythroid engulfed by macrophage cells [22]. As the cell membrane and endothelial lineages is further supported by the plays an important role in the biology of these cells, finding that the primitive erythroid lineage in the mouse extruded nucleus is an inadequate term and ‘pyreno- emerges from mesodermal cells that express markers cytes’, derived from the Greek word ‘pyren’ (the pit of a found in adult endothelial cells, including flk-1, vascular stone fruit), has been proposed as a more appropriate endothelial cadherin, tie-2, and PECAM-1 [13]. There is name for this very transient cell [20]. The discovery of an increasing evidence, however, that suggests that primitive pyrenocytes in the fetal bloodstream suggests many, if not most, yolk sac vascular cells in the conceptus that late-stage primitive erythroblasts enucleate by arise from unilineage angioblast precursors and not from nuclear extrusion. Unlike definitive erythroblasts, primi- hemangioblasts [14,15]. Thus, it is likely that all hema- tive erythroblasts do not enucleate spontaneously in vitro topoietic cells, but few endothelial cells, in the mamma- [20]. Nevertheless, they are capable, like definitive lian yolk sac arise from hemangioblast precursors. erythroblasts, of physically interacting with F4/80- positive macrophage cells in vitro,inpartthrougha4- integrin-mediated interactions [20,23]. These findings, Terminal maturation of primitive erythroid supported also by immunohistochemical studies [20], cells raise the intriguing likelihood that primitive erythro- Immature primitive erythroblasts in the blood islands of blasts enucleate while associated with macrophage cells the mouse yolk sac begin to circulate at E8.25 coincident in vivo.

Figure 1 Enucleation of late-stage erythroblasts leads to the formation of two cells – a and a pyrenocyte

n Pyrenocyte

n n

Reticulocyte Late-stage erythroblast

Pyrenocytes lose phosphatidylserine asymmetry and are subsequently phagocytosed by macrophage cells. Reproduced with permission [22].

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. Ontogeny of erythropoiesis Palis 157

regulate the adult b-globin genes. Recent reexamination Globin regulation in primitive erythroid cells of primitive as well as definitive erythroid cells in EKLF- Hemoglobin molecules contain globin chains derived both null mouse embryos, however, indicates that EKLF also a b from the -globin and -globin gene loci. Although defini- regulates multiple erythroid-specific genes, including tive erythroid cells in the mouse express a1-globin, a2- genes encoding AHSP, heme synthetic pathway proteins globin, b1-globin, and b2-globin, primitive erythroid cells and cytoskeletal proteins such as ankyrin and band in addition express z-globin, bH1-globin, and ey-globin 3 [33,34]. Surprisingly, EKLF-null mouse embryos also [24]. An extensive analysis of globin gene expression in have reduced accumulation of bH1-globin and ey-globin primitive erythroid cells indicates that the initially transcripts in primitive erythroblasts [35]. Furthermore, expressed z- and bH1-globin genes are superseded by double EKLF/-null mouse fetuses have a more the a1-globin, a2-globin and ey-globin genes, respect- severe reduction in embryonic globin gene expression ively, as primitive at E7.5 transition to than either single-null mutant, indicating that these two at E15.5 [25]. This ‘maturational’ globin Kruppel-like factors play nonredundant roles in embryo- switching is associated with changes in RNA polymerase nic globin gene regulation. II density at the promoters of these various globin genes. Furthermore, the bH1-globin and ey-globin genes in is the central cytokine necessary for primitive erythroid cells reside in a single large hyperace- definitive erythroid cell maturation; however, its role in tylated domain, suggesting that this novel form of globin primitive erythropoiesis is less well understood. Tar- switching is regulated by altered transcription factor pre- geted disruption of erythropoietin in mouse leads to a sence instead of chromatin accessibility [25]. significant decrease in primitive erythroid cell numbers, but maturation does not seem to be disrupted for the The GATA1 transcription factor plays an essential role in remainder[36,37].Interestingly,morpholinoknockdown the regulation of erythroid-specific genes in both primi- of erythropoietin in zebrafish embryos leads to a similar tive and definitive erythroid cells and the loss of GATA1 differential phenotype between primitive and definitive leads to the arrest of both lineages at the erythropoiesis [38]. Examination of erythropoietin sig- stage of maturation [26,27]. Interestingly, different func- naling revealed several differences in mouse embryonic tional domains of GATA1 are required for activation of stem cell-derived primitive and definitive erythroid cells target genes in primitive versus definitive erythroid cells [39]. Primitive erythroblasts express higher levels of [28], suggesting that different transcriptional complexes erythropoietin receptor and have a sustained and robust may form in these lineages. Recent examination of mur- phosphorylation of the downstream STAT5 signaling ine erythroleukemia cells has led to the identification of molecule but barely detectable levels of the STAT5 novel transcriptional complexes involving a core complex inhibitory protein SHP-1. These differences predict a (GATA1, Tal1, E2A, Lmo2, and Ldbd1) and newly heightened sensitivity of primitive erythroblasts to ery- identified Ldb1-binding partners Eto-2, Cdk9, and thropoietinsignaling.Althoughthesestudiessuggestthat Lmo4 [29]. Forced downregulation of these latter tran- there may be differential roles for erythropoietin in scription factors in zebrafish embryos reveals functional primitive versus definitive erythropoiesis, it is likely that roles in definitive, but not primitive, hematopoiesis. other cytokine signaling cascades differentially regulate These and other data are leading to increasing complex primitive and definitive erythroid cell maturation. models of genetic regulatory networks in the emerging embryo and definitive erythroid cells [30]. Hematopoiesis in the yolk sac is not confined Several transcription factors have been implicated in the to primitive erythropoiesis regulation of globin gene expression in primitive ery- Examination of hematopoietic progenitors in carefully throid cells, including several Kruppel-like transcription staged mouse embryos as well as cultured embryonic factors. Studies of klf2-null mouse embryos revealed stem cells has revealed two overlapping waves of ery- significant decreases in bH1-globin and ey-globin tran- throid potential [40–42] (Fig. 2). The first wave consists scripts in primitive erythroid precursors [31]. Interest- of primitive erythroid progenitors (EryP-CFC) present in ingly, human e-globin transgenes were also reduced in the yolk sac between E7.25 and E9.0 of gestation that is mice lacking klf2, providing evidence that this transcrip- temporally associated with macrophage and megakaryo- tion factor plays a similar role in . Morpholino cyte progenitors. Recent clonogenic studies in mouse knockdown of klf4 in zebrafish embryos leads to embryos indicate that the primitive erythroid and mega- decreased embryonic globin gene expression [32]. karyocyte lineages are tied to a common bipotential prim- Furthermore, klf4 preferentially binds the CACC sites itive erythroid/ progenitor [43]. Thus, in the promoters of the embryonic compared with the primitive hematopoiesis is at least bilineage in nature. adult b-globin genes. The erythroid-specific Kruppel- The developmental origin of thrombocytes, however, has like factor (EKLF) was originally thought to primarily not yet been examined in other model organisms. The

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. 158 Erythroid system and its diseases

Figure 2 Simplified model of erythroid ontogeny in the mammalian embryo

Current data support a model in which three waves of erythroid progenitors emerge in the mammalian embryo. The first wave consists of primitive erythroid progenitors (EryP-CFC) that originate in the yolk sac from hemangioblast precursors (HB) and generate a synchronous cohort of primitive erythroid precursors that mature in the bloodstream and enucleate to form reticulocytes and pyrenocytes. The second wave consists of a transient wave of definitive erythroid progenitors (BFU-E) that emerge from the yolk sac and seed the fetal . There they generate maturing definitive erythroid precursors that enucleate to become the first circulating definitive erythrocytes (RBC) of the fetus. The third wave consists of a continuous stream of definitive erythroid progenitors in the late gestation liver and postnatal marrow that originate from adult-repopulating hematopoietic stem cells (HSC). Unlike primitive erythroid cells, definitive erythroid precursors mature extravascularly within erythroblast islands. AGM, aorta–gonad–mesonephros region; mf, macrophage cell.

second wave of definitive erythroid progenitors (BFU-E) Further evidence in support of the developmental origin emerges and expands in the yolk sac between E8.25 and of definitive erythroid potential comes from the mouse E10.5 (see below), and is temporally linked to the expan- models lacking a functional circulation, as for exam- sion of multiple unilineage and multilineage myeloid ple, vascular endothelial cadherin-null embryos [45]. A progenitors [42]. more recent model involves loss of NCX1, a sodium– calcium exchanger whose expression is confined to the heart in the mouse embryo. NCX1-null embryos lack Emergence of definitive erythroid potential in a heartbeat and thus have no functional circulation, and the embryo yet have normal numbers of primitive and definitive Primitiveerythroidcellsfulfillthefunctionscriticalforearly erythroid progenitors emerging in the yolk sac [46]. postimplantation embryonic survival and growth, however Few definitive erythroid progenitors, however, are found the fetus requires even more red cells to meet the demands within the embryo proper of these mutant mice, support- of growth at later stages of development. Prior to the ing the hypothesis that all definitive hematopoietic pro- formation of the marrow, the liver serves as the site genitors are initially generated in the yolk sac between ofmaturationofdefinitiveerythroidcellsinthemammalian E8.25 and E10 and are redistributed to the embryo proper fetus. The liver in the murine embryo is colonized by at the onset of embryonic circulation. The NCX1-mutant external hematopoietic elements at E9.5, soon after it embryo should continue to serve as a useful model to begins to form as an organ. BFU-E and CFU-E sub- dissect the emergence of hematopoietic potential within sequently expand exponentially in numbers for several the embryo proper and the placenta without the con- days and generate definitive erythroid cells [44]. The founding factor of movement within the circu- developmental origin of the BFU-E that initially colonize lation. In aggregate, these studies indicate that a transient the liver has been postulated to be the yolk sac, as BFU-E wave of definitive erythroid progenitors emerges from the first emerge in the yolk sac at E8.25, before the onset of yolk sac of mammalian embryos prior to the emergence of the circulation, and continue to preferentially expand in adult-repopulating hematopoietic stem cells and colo- numberswithintheyolksacforthenext48 h[40,42](Fig.2). nizes the fetal liver (Fig. 2). Interestingly, a wave of

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. Ontogeny of erythropoiesis Palis 159

definitive ‘erythro-myeloid’ progenitors has recently podocalyxin, and thus regulating reticulocyte egress from been found in the posterior blood islands of zebrafish the marrow [56]. These studies highlight an emerging role embryos after the appearance of primitive erythroid cells for adhesion factors in the terminal differentiation of in the intermediate cell mass but before emergence of definitive erythroid cells. HSC in the region of the aorta [47]. These findings provide evidence that hematopoietic ontogeny is quite MicroRNAs are endogenous molecules that target highly conserved between disparate species. mRNAs in a sequence-specific manner and regulate various cellular functions. Recent screens have identified Toward the end of gestation, definitive erythroid progen- numerous microRNAs in definitive erythroid cells that itors in the mammalian embryo transition from the liver to are either upregulated or downregulated with maturation the newly formed . Although fundamentally [57–59]. Initial functional studies in murine erythroleu- similar to their adult counterparts, fetal erythroid progeni- kemia cells indicate that the abundantly expressed miR- tors also have some distinctive features. First, fetal BFU-E 451 facilitates erythroid maturation [57]. The continued have a greater and more rapid proliferative capacity. Sec- study of microRNAs and their targets will lead to the ond, fetal, but not steady-state adult, BFU-E proliferate identification of genes critical for hematopoietic differ- invitroinresponsetoerythropoietinintheabsenceofadded entiation and erythroid lineage maturation throughout colony-stimulating factors [48,49]. Finally, CFU-E in ontogeny. the murine fetus are more sensitive to erythropoietin [50].

Human embryonic stem cells serve as a new Maturation of definitive erythroid cells model of embryonic erythropoiesis In contrast to primitive erythroid cells that mature in the The culture of human embryonic stem cells has emerged bloodstream, definitive erythroid precursors in the fetal as an increasingly important model of early embryonic liver and postnatal marrow mature while attached to events. Recent studies indicate that the in-vitro differ- macrophage cells of erythroblastic islands [51] (Fig. 2). entiation of human embryonic stem cells recapitulates Although macrophage cells engulf and digest pyrenocytes hematopoietic ontogeny reminiscent of the in-vivo yolk following enucleation [22], it is unclear what other func- sac, including overlapping waves of hemangioblast, tions macrophage cells perform to enhance erythroid primitive erythroid, and definitive erythroid potential maturation. It has recently been shown that coculture of [60–62]. Interestingly, culture of differentiating human maturing Friend leukemia virus infected erythroid cells embryonic stem cells in liquid culture has recently pro- with macrophage cells in vitro enhanced their proliferation vided evidence that human primitive erythroid cells, like but did not alter their intrinsic ability to enucleate [52]. their murine counterparts, undergo ‘maturational’ z-glo- Potential ‘nurse’ functions of macrophage cells, such as the bin to a-globin switching [63]. Thus, human embryonic provision of iron and cytokines, remain to be proven. stem cells can provide access to hematopoietic cells that are not otherwise available for study. Erythroblast islands bring maturing definitive erythroid precursors in contact not only with macrophage cells but also with other erythroblasts. Fas and FasL signaling Conclusion between maturing erythroblasts has been invoked as a Primitive erythropoiesis serves as a useful model of mam- mechanism controlling erythroid cell output. Both nega- malian erythroid differentiation, as primitive erythroblasts tive feedback of more mature erythroblasts on immature in mammals mature in a synchronous cohort in the blood- erythroblasts as well as autoregulatory loops at the proer- stream and ultimately enucleate. In contrast, definitive ythroblast stage of maturation have been proposed erythroid precursors mature attached to macrophage cells [53,54]. As definitive erythroblasts mature extravascu- in erythroblast islands within the complex cellular milieu larly, they are also in contact with stromal elements. of the fetal liver and postnatal bone marrow (Fig. 2). Recent evidence suggests that definitive erythroblasts Erythroblastic islands can thus facilitate erythroblast– transition from an erthropoietin-dependent phase to a erythroblast and erythroblast–stromal interactions that fibronectin-dependent phase, and that both erythropoietin play important roles in the regulation of definitive ery- and fibronectin deliver antiapoptotic signals to maturing thropoiesis. The continued study of the similarities erythroblasts [55]. Alpha 4 integrin was shown to mediate and differences between primitive and definitive erythro- this erythroblast–fibronectin interaction, raising the intri- poiesis will lead to an improved understanding of eryth- guing question of whether similar signals are mediated to roblast maturation and the terminal steps of erythroid erythroblasts by VCAM1 on macrophage cells. Surpris- differentiation. ingly, erythropoietin was recently shown to play a role at very late stages of erythroid maturation by specifically The comparative investigation of embryonic development altering the expression of adhesion molecules, including in multiple model organisms, including mouse, chicken,

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. 160 Erythroid system and its diseases

Xenopus and zebrafish, will continue to provide important 18 Fantoni A, de la Chapelle A, Marks PA. Synthesis of embryonic during erythroid cell development in fetal mice. J Biol Chem 1969; 244:675– insights into the ontogeny of erythropoiesis. The in-vitro 681. culture of human embryonic system cells is now estab- 19 Fraser ST, Isern J, Baron MH. Maturation and enucleation of primitive lished as an important model system to investigate the erythroblasts during mouse embryogenesis is accompanied by changes in cell-surface antigen expression. Blood 2007; 109:343–352. early ontogeny of human hematopoiesis and offers the 20 McGrath KE, Kingsley PD, Koniski AD, et al. Enucleation of primitive erythroid potential for cell-based therapies for the treatment of cells generates a transient population of ‘‘pyrenocytes’’ in the mammalian hematopoietic disorders. fetus. Blood 2008; 111:2408–2417. The appearance of primitive pyrenocytes provides evidence that primitive erythro- blasts enucleate by nuclear extrusion. Interestingly, primitive erythroblasts can physically interact with macrophage cells and reconstitute erythroblast islands Acknowledgements in vitro. I thank coworkers and colleagues for many stimulating discussions. 21 Skutelsky E, Danon D. An electron microscopic study of nuclear elimination This study was supported in part by the National Institutes of Health from the late erythroblast. J Cell Biol 1967; 33:625–635. grants DK09361 and DK071116. 22 Yoshida H, Kawane K, Koike M, et al. Phosphatidylserine-dependent engulf- ment by of nuclei from erythroid precursor cells. Nature 2005; 437:754–758. 23 Sadahira Y, Yoshino T, Monobe Y. Very late activation antigen 4-vascular cell References and recommended reading adhesion molecule 1 interaction is involved in the formation of erythroblastic Papers of particular interest, published within the annual period of review, have islands. J Exp Med 1995; 181:411–415. been highlighted as: 24 Trimborn T, Gribnau J, Grosveld F, Fraser P. Mechanisms of developmental of special interest control of transcription in the murine a- and b-globin loci. Genes Dev 1999; of outstanding interest 13:112–124. Additional references related to this topic can also be found in the Current World Literature section in this issue (pp. 256–257). 25 Kingsley PD, Malik J, Emerson RL, et al. ‘‘Maturational’’ globin switching in primary primitive erythroid cells. Blood 2006; 107:1667–1672. 1 Gulliver G. Observations on the sizes and shapes of red corpuscles of the 26 Pevny L, Lin C-S, D’Agati V, et al. Development of hematopoietic cells lacking blood of vertebrates, with drawings of them to a uniform scale, and extended the transcription factor GATA-1. Development 1995; 121:163–172. and revised tables of measurements. Proc Zool Soc London 1875; 474-495. 27 Fujiwara Y, Browne CP, Cunniff K, et al. Arrested development of embryonic 2 Maximow AA. The earliest developmental stages of blood and connective red cell precursors in mouse embryos lacking transcription factor GATA-1. tissue cells in the mammalian embryo until blood formation in the liver [in Proc Natl Acad Sci U S A 1996; 93:12355–12358. German]. Arch Mikroskop Anat 1909; 73:444–561. 28 Shimizu R, Takahashi S, Ohneda K, et al. In vivo requirements for GATA-1 3 Kingsley PD, Malik J, Fantauzzo K, Palis J. Yolk sac-derived primitive erythro- functional domains during primitive and definitive erythropoiesis. EMBO J blasts enucleate during mammalian embryogenesis. Blood 2004; 104:19– 2001; 20:5250–5260. 25. 29 Meier N, Krpic S, Rodriguez P, et al. Novel binding partners of Ldb1 are 4 Baron MH, Frazer ST. The specification of early hematopoiesis in the . required for haematopoietic development. Development 2006; 133:4913– Curr Opin Hematol 2005; 12:217–221. 4924. 5 Sabin FR. Studies on the origin of blood-vessels and red blood-corpuscles as 30 Swiers G, Patient R, Loose M. Genetic regulatory networks programming seen in the living blastoderm of chicks during the second day of incubation. hematopoietic stem cells and erythroid lineage specification. Dev Biol 2006; Contr Embryol 1920; 9:215–262. 294:525–540. 6 Haar JL, Ackerman GA. Ultrastructural changes in mouse yolk sac associated 31 Basu P, Morris PE, Haar JL, et al. KLF2 is essential for primitive erythropoiesis with the initiation of vitelline circulation. Anat Rec 1971; 170:437–456. and regulates the human and murine embryonic beta-like globin genes in vivo. 7 Ferkowicz MJ, Yoder MC. Blood island formation: longstanding observations Blood 2005; 106:2566–2571. and modern interpretations. Exp Hematol 2005; 33:1041–1049. 32 Gardiner MR, Gongora MM, Grimmond SM, Perkins AC. A global role for 8 Choi K, Kennedy M, Kazarov A, et al. A common precursor for hematopoietic zebrafish klf4 in embryonic erythropoiesis. Mech Dev 2007; 124:762–774. and endothelial cells. Development 1998; 125:725–732. 33 Hodge D, Coghill E, Keys J, et al. A global role for EKLF in definitive and 9 Huber TL, Kouskoff V, Fehling HJ, et al. Haemangioblast commitment is primitive erythropoiesis. Blood 2006; 107:3359–3370. initiated in the primitive streak of the mouse embryo. Nature 2004; 432:625– 34 Nilson DG, Sabatino DE, Bodine DM, Gallagher PG. Major erythrocyte 630. membrane protein genes in EKLF-deficient mice. Exp Hematol 2006; 10 Yokomizo T, Takahashi S, Mochizuki N, et al. Characterization of GATA-1(þ) 34:705–712. hemangioblastic cells in the mouse embryo. EMBO J 2007; 26:184– 35 Basu P, Lung TK, Lemsaddek W, et al. EKLF and KLF2 have compensatory 196. roles in embryonic {beta}-globin gene expression and primitive erythropoiesis. GATA-1(þ) cells isolated from E7.0–7.5 mouse embryos include a common Blood 2007; 110:3417–3425. precursor for hematopoietic and endothelial cells, as well as primitive and definitive erythroid progenitors. 36 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 11 Lugus JJ, Chung YS, Mills JC, et al. GATA2 functions at multiple steps in receptor. Cell 1995; 83:59–67. hemangioblast development and differentiation. Development 2007; 134:393–405. 37 Lin C-S, Lim S-K, D’Agati V, Constantini F. Differential effects of an erythro- poietin receptor gene disruption on primitive and definitive erythropoiesis. 12 Perlingeiro RC. Endoglin is required for hemangioblast and early hemato- Genes Dev 1996; 10:154–164. poietic development. Development 2007; 134:3041–3048. 38 Paffett-Lugassy N, Hsia N, Fraenkel PG, et al. I. Functional conservation of 13 Ema M, Yokomizo T, Wakamatsu A, et al. Primitive erythropoiesis from erythropoietin signaling in zebrafish. Blood 2007; 110:2718–2726. mesodermal precursors expressing VE-cadherin, PECAM-1, Tie2, endoglin, 39 Tsuji-Takayama K, Otani T, Inoue T, et al. Erythropoietin induces sustained and CD34 in the mouse embryo. Blood 2006; 108:4018–4024. phosphorylation of STAT5 in primitive but not definitive erythrocytes gener- 14 Ueno H, Weissman IL. Clonal analysis of mouse development reveals a ated from mouse embryonic stem cells. Exp Hematol 2006; 34:1323–1332. polyclonal origin for yolk sac blood islands. Dev Cell 2006; 11:519– 40 Wong PMC, Chung SW, Chui DHK, Eaves CJ. Properties of the earliest 533. clonogenic hemopoietic precursors to appear in the developing murine yolk 15 Furuta C, Ema H, Takayanagi S, et al. Discordant developmental waves of sac. Proc Natl Acad Sci U S A 1986; 83:3851–3854. angioblasts and hemangioblasts in the early gastrulating mouse embryo. 41 Keller G, Kennedy M, Papayannopoulou TH, Wiles MV. Hematopoietic Development 2006; 133:2771–2779. commitment during embryonic stem cell differentiation in culture. Mol Cell 16 McGrath KE, Koniski AD, Malik J, Palis J. Circulation is established in a step- Biol 1993; 13:473–486. wise pattern in the mammalian embryo. Blood 2003; 101:1669–1676. 42 Palis J, Robertson S, Kennedy M, et al. Development of erythroid and myeloid 17 Lucitti JL, Jones EA, Huang C, et al. Vascular remodeling of the mouse yolk sac progenitors in the yolk sac and embryo proper of the mouse. Development requires hemodynamic force. Development 2007; 134:3317–3326. 1999; 126:5073–5084.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited. Ontogeny of erythropoiesis Palis 161

43 Tober J, Koniski A, McGrath KE, et al. The megakaryocyte lineage originates 53 De Maria R, Testa U, Luchetti L, et al. Apoptotic role of Fas/Fas ligand system from hemangioblast precursors and is an integral component both of primitive in the regulation of erythropoiesis. Blood 1999; 93:796–803. and of definitive hematopoiesis. Blood 2007; 109:1433–1441. 54 Socolovsky M, Murrell M, Liu Y, et al. Negative autoregulation by FAS 44 Kurata H, Mancini GC, Alespieti G, et al. Stem cell factor induces proliferation mediates robust fetal erythropoiesis. PLoS Biol 2007; 5:2296–2311. and differentiation of fetal progenitor cells in the mouse. Br J Haematol 1998; 55 Eshghi S, Vogelzang MG, Hynes RO, et al. a4b1 integrin and erythropoietin 101:676–687. mediate temporally distinct steps in erythropoiesis: integrins in red cell 45 Rampon C, Huber P. Multilineage hematopoietic progenitor activity generated development. J Cell Biol 2007; 177:871–880. autonomously in the mouse yolk sac: analysis using angiogenesis-defective 56 Sathyanarayana P, Menon MP, Bogacheva O, et al. Erythropoietin modulation embryos. Int J Dev Biol 2003; 47:273–280. of podocalyxin, and a proposed erythroblast niche. Blood 2007; 110:509– 46 Lux C, Yoshimoto M, McGrath KE, et al. All primitive and definitive hemato- 518. poietic progenitor cells emerging prior to E10 in the mouse embryo are The authors identify several adhesion molecules in early and late stages of products of the yolk sac. Blood 2007; 11 Oct [Epub ahead of print]. erythroid precursor maturation that are targets of erythropoietin signaling and NCX1-null mouse embryos lack a heartbeat but survive to E10. The lack of a the modulation of these target genes can regulate reticulocyte egress from the heartbeat permits the study of the emergence of hematopoietic potential in the yolk marrow. sac, embryo proper and placenta without the complicating factor of circulating 57 Zhan M, Miller C, Papayannopoulou TH, et al. MicroRNA expression dynamics blood cells. Here the authors confirm that all the definitive erythroid progenitors in during murine and human erythroid differentiation. Exp Hematol 2007; the E9.5 mouse embryo are derived from the yolk sac. 35:1015–1025. 47 Bertrand JY, Kim AD, Violette EP, et al. Definitive hematopoiesis initiates 58 Choong ML, Yang HH, McNiece I. MicroRNA expression profiling during through a committed erythromyeloid progenitor in the zebrafish embryo. human cord blood-derived CD34 cell erythropoiesis. Exp Hematol 2007; Development 2007; 134:4147–4156. 35:551–564. The authors show that there is a wave of definitive ‘erythro-myeloid’ progenitors that emerge from posterior blood islands of the zebrafish embryo prior to the 59 Bruchova H, Yoon D, Agarwal AM, et al. Regulated expression of microRNAs emergence of HSC in the para-aortic region. This study indicates that ‘definitive’ in normal and vera erythropoiesis. Exp Hematol 2007; erythropoiesis in zebrafish embryos, like that in the mouse, emerges prior to 35:1657–1667. emergence in the AGM. 60 Zambidis ET, Peault B, Park TS, et al. Hematopoietic differentiation of human 48 Emerson SG, Shanti T, Ferrara JL, Greenstein JL. Developmental regulation of embryonic stem cells progresses through sequential hematoendothelial, erythropoiesis by hematopoietic growth factors: analysis on populations of primitive, and definitive stages resembling human yolk sac development. BFU-E from bone marrow, peripheral blood, and fetal liver. Blood 1989; Blood 2005; 106:860–870. 74:49–55. 61 Chang K-H, Nelson AM, Cao H, et al. Definitive-like erythroid cells derived 49 Migliaccio AR, Migliaccio G. Human embryonic hemopoiesis: control me- from human embryonic stem cells coexpress high levels of embryonic and chanisms underlying progenitor differentiation in vitro. Dev Biol 1988; fetal globins with little or no adult globin. Blood 2006; 108:1515–1523. 125:127–134. 62 Kennedy M, D’Souza SL, Lynch-Kattman M, et al. Development of the 50 Rich IN, Kubanek B. Erythroid colony formation in foetal liver and adult bone hemangioblast defines the onset of hematopoiesis in human ES cell differ- marrow and from the mouse. Blood 1976; 33:171–180. entiation cultures. Blood 2007; 109:2679–2687. 51 Chasis JA. Erythroblastic islands: specialized microenvironmental niches for 63 Qiu C, Olivier EN, Velho M, Bouhassira EE. Globin switches in yolk-sac-like erythropoiesis. Curr Opin Hematol 2006; 13:137–141. primitive and fetal-like definitive red blood cells produced from human em- 52 Rhodes MM, Kopsombut P, Bondurant MC, et al. Adherence to macrophages bryonic stem cells. Blood 2008; 111:2400–2408. in erythroblastic islands enhances erythroblast proliferation and increases Culture of human embryonic stem cells generates large numbers of primitive erythrocyte production by a different mechanism than erythropoietin. Blood erythroblasts that mature in vitro and undergo a ‘maturational’ switch from Hb 2008; 111:1700–1708. Gower I (z2e2) to Gower II (a2e2) hemoglobin.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.