scholarly journals Definitive EMP and Pre-HSC Emerge in Myb-Null Murine Embryos and Retain Macrophage Potential

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2556-2556
Author(s):  
Kathleen E. McGrath ◽  
Jenna M Frame ◽  
Katherine H Fegan ◽  
Emanuele Azzoni ◽  
Paul D. Kingsley ◽  
...  

Abstract Myb-null murine embryos lack definitive erythropoiesis but can produce primitive erythroid cells, allowing their survival to embryonic day 15 (E15) (Mucenski et al., Cell, 1991). Myb expression has been used to detect emerging HSC and is required for HSC maintenance (Lieu et al., PNAS 2009; North et al., Cell 2009). These data have led to the model that myb function is required for definitive hematopoiesis. Interestingly, macrophages and megakaryocytes are still detected in myb-null embryos and, as both of these lineages are also components of primitive hematopoiesis, it is proposed that these cells are not definitive in origin. Myb-independent macrophages infiltrate fetal tissues and have been implicated as a self-renewing source for several adult tissue-resident macrophage populations (Schulz et al., Nature 2012; Gomez Perdiguero et al., Glia 2013; Hoeffel et al., Immunity 2015). We tested the hypothesis that definitive hematopoiesis is entirely myb-dependent by examining two distinct sources of definitive erythroid/myeloid potential: 1) HSCs that emerge from hemogenic endothelium, including the AGM region at E10.5 in mice and 2) HSC-independent definitive EMP that emerge after primitive hematopoiesis from yolk sac hemogenic endothelium beginning at E8.25 (Frame et al., Stem Cells 2016). By E9.5, EMP can be prospectively isolated based on immunophenotype and contain all the erythroid/myeloid progenitor activity present at this time (McGrath et al., Cell Reports 2015). Surprisingly, we found normal numbers of immunophenotypic EMP in E9.5 Myb-null yolk sacs, and immunohistochemical analysis confirmed their emergence from Runx1+ hemogenic endothelium. At E10.5, reduced numbers of Myb-null EMP were found not only in the yolk sac, but also in the bloodstream and the liver. This decrease correlated with fewer hemogenic clusters in the yolk sacs of Myb-null embryos, as well as alterations in their cell-cycle status. The presence of significant numbers of immunophenotypic EMP suggests they could serve as an alternate source of Myb-null macrophages. Clonal analysis of sorted EMP confirmed that Myb function is necessary for erythroid and granulocyte progenitors, but Myb-null EMP retain normal plating efficiencies for macrophage progenitors. Indeed, Myb-null EMP generate only macrophages in liquid culture, lacking not only erythroid and granulocyte cells, but also Ly6C+ monocytes. Consistent with these results, RNA sequencing analysis of Myb-null versus wildtype EMP demonstrated decreased expression of genes in pathways associated with cellular growth, as well as erythroid and granulocyte fates. We further determined that Myb is not required for the emergence of immunophenotypic pre-HSC in the AGM region of E10.5 embryos. In addition, there were normal numbers of clusters in the aorta of E10.5 myb-null embryos. We were also able to detect lineage-, Kit+ (LK) cells in E14.5 livers as previously reported (Sumner et al., Oncogene 2000). While LK numbers were reduced, the Sca1+ (LSK) subset was present in normal numbers. Like EMP, sorted Myb-null E10.5 pre-HSC, as well as E14.5 liver LK and LSK, lacked erythroid or granulocyte CFC activity, but retained normal CFC-M plating efficiencies. In addition, Ly6C+ monocytes were not observed in liquid cultures of sorted Myb-null E10.5 pre-HSC, which produced only macrophages in vitro, or in Myb-null E14.5 livers. Together these data indicate that Myb is not required for hematopoietic emergence of definitive EMP or HSC, but does facilitate the expansion of these definitive stem/progenitor cells and is required for erythroid and granulocyte differentiation. Additionally, EMP and HSC contain Myb-independent macrophage potential, which does not appear to differentiate from a monocyte intermediate. Disclosures Palis: Rubius Therapeutics: Consultancy.

Blood ◽  
2006 ◽  
Vol 109 (4) ◽  
pp. 1433-1441 ◽  
Author(s):  
Joanna Tober ◽  
Anne Koniski ◽  
Kathleen E. McGrath ◽  
Radhika Vemishetti ◽  
Rachael Emerson ◽  
...  

Abstract In the adult, platelets are derived from unipotential megakaryocyte colony-forming cells (Meg-CFCs) that arise from bipotential megakaryocyte/erythroid progenitors (MEPs). To better define the developmental origin of the megakaryocyte lineage, several aspects of megakaryopoiesis, including progenitors, maturing megakaryocytes, and circulating platelets, were examined in the murine embryo. We found that a majority of hemangioblast precursors during early gastrulation contains megakaryocyte potential. Combining progenitor assays with immunohistochemical analysis, we identified 2 waves of MEPs in the yolk sac associated with the primitive and definitive erythroid lineages. Primitive MEPs emerge at E7.25 along with megakaryocyte and primitive erythroid progenitors, indicating that primitive hematopoiesis is bilineage in nature. Subsequently, definitive MEPs expand in the yolk sac with Meg-CFCs and definitive erythroid progenitors. The first GP1bβ-positive cells in the conceptus were identified in the yolk sac at E9.5, while large, highly reticulated platelets were detected in the embryonic bloodstream beginning at E10.5. At this time, the number of megakaryocyte progenitors begins to decline in the yolk sac and expand in the fetal liver. We conclude that the megakaryocyte lineage initially originates from hemangioblast precursors during early gastrulation and is closely associated both with primitive and with definitive erythroid lineages in the yolk sac prior to the transition of hematopoiesis to intraembryonic sites.


Blood ◽  
1999 ◽  
Vol 94 (1) ◽  
pp. 127-138 ◽  
Author(s):  
Agnieszka M. Lichanska ◽  
Catherine M. Browne ◽  
Gregory W. Henkel ◽  
Kathleen M. Murphy ◽  
Michael C. Ostrowski ◽  
...  

During mouse embryogenesis, macrophage-like cells arise first in the yolk sac and are produced subsequently in the liver. The onset of liver hematopoiesis is associated with the transition from primitive to definitive erythrocyte production. This report addresses the hypothesis that a similar transition in phenotype occurs in myelopoiesis. We have used whole mount in situ hybridization to detect macrophage-specific genes expressed during mouse development. The mouse c-fms mRNA, encoding the receptor for macrophage colony-stimulating factor (CSF-1), was expressed on phagocytic cells in the yolk sac and throughout the embryo before the onset of liver hematopoiesis. Similar cells were detected using the mannose receptor, the complement receptor (CR3), or the Microphthalmia transcription factor (MITF) as mRNA markers. By contrast, other markers including the F4/80 antigen, the macrophage scavenger receptor, the S-100 proteins, S100A8 and S100A9, and the secretory product lysozyme appeared later in development and appeared restricted to only a subset of c-fms–positive cells. Two-color immunolabeling on disaggregated cells confirmed that CR3 and c-fmsproteins are expressed on the same cells. Among the genes appearing later in development was the macrophage-restricted transcription factor, PU.1, which has been shown to be required for normal adult myelopoiesis. Mice with null mutations in PU.1 had normal numbers of c-fms–positive phagocytes at 11.5dpc. PU.1(−/−) embryonic stem cells were able to give rise to macrophage-like cells after cultivation in vitro. The results support previous evidence that yolk sac–derived fetal phagocytes are functionally distinct from those arising in the liver and develop via a different pathway.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2372-2372
Author(s):  
Jenna M. Frame ◽  
Katherine H. Fegan ◽  
Seana C. Catherman ◽  
Joanna Tober ◽  
Anne D. Koniski ◽  
...  

Abstract In the adult, the proto-oncogene Myb critically regulates both the maintenance of hematopoiesis and the differentiation of several hematopoietic lineages. Myb-/- mouse embryos die by embryonic day (E) 15 with severe anemia due to the absence of definitive erythropoiesis (Mucenski et al., Cell, 1991). Similarly, zebrafish embryos lacking myb do not express adult globin genes, have a reduction in other mature hematopoietic lineages by 48 hours post fertilization, and maintain a bloodless phenotype through adulthood (Soza-Ried et al., PNAS, 2010). These and other data have led to the concept that Myb-/- embryos entirely lack definitive hematopoiesis. In both mouse and zebrafish embryos, the first definitive hematopoietic potential arises as a hematopoietic stem cell (HSC)-independent wave of erythro-myeloid progenitors (EMPs). EMPs emerge in the murine yolk sac beginning at E8.25, partially overlapping with an earlier wave of primitive hematopoietic progenitors. We previously demonstrated that EMPs are multipotent progenitors, and are the major source of definitive erythroid potential in the early fetal liver, prior to the colonization of adult-repopulating HSCs (McGrath et al., Blood, 2011). Recently, we identified a unique cell surface phenotype that facilitates the prospective isolation of murine definitive EMPs, distinguishing them from primitive hematopoietic progenitors and maturing populations of megakaryocytes and macrophages in the yolk sac (McGrath et al., Cell Reports, 2015). We detected expression of Myb in sorted EMPs, suggesting that Myb may regulate the emergence and/or differentiation of EMPs. We tested this hypothesis by assessing the emergence, hematopoietic potential and expansion capacity of EMPs, compared with other maturing primitive hematopoietic lineages, in Myb-/- mouse embryos. Consistent with the proposed Myb-independence of the earlier wave of primitive progenitors, we observed normal numbers of maturing macrophages in E9.5 Myb-/- yolk sacs. Interestingly, E9.5 Myb-/- yolk sacs also contained normal numbers of immunophenotypic EMPs. These EMPs were present in hemogenic endothelial-derived clusters expressing Runx1, similar to littermate controls, suggesting that Myb is dispensable for EMP emergence from hemogenic endothelium. We next assessed the differentiation capability of Myb-/- EMPs in vitro. E9.5 Myb-/- yolk sacs lacked high proliferative colony-forming potential (HPP-CFC), a hallmark of immature definitive hematopoietic progenitors. In addition, both definitive erythroid and granulocyte colony-forming potential were absent in methylcellulose cultures of sorted Myb-/- EMPs, in contrast to littermate controls. Surprisingly, however, sorted Myb-/- EMPs gave rise to macrophage progenitors in colony-forming assays, and CD11b+ F4/80+ macrophages in differentiation cultures. These data indicate that Myb is not required for the differentiation of primary definitive EMPs into macrophages. Analysis of Myb-/- fetal liversalso confirmed the presence of F4/80+ macrophages. While these fetal liver macrophages have been previously proposed to be of primitive hematopoietic origin, our data raise the possibility that they may also be derived from EMPs. Further analysis of in vitro differentiation cultures demonstrated an inability of sorted Myb-/- EMPs to proliferate when compared with normal littermates, although these cultures still generated small numbers of macrophages. It is not yet clear whether this reduction in proliferation is due solely to the loss of differentiation of multiple hematopoietic lineages, or is also due to defective maintenance or expansion of EMPs. However, consistent with a role for Myb in continued emergence and/or expansion of EMPs, we observed a reduction in the total number of EMPs by E10.5 in yolk sacs of Myb-/- embryos compared with normal littermates. Taken together, these data indicate that Myb is a critical regulator not only of HSCs, but also of HSC-independent definitive hematopoietic progenitors (EMPs). While Myb is dispensable for the initial emergence of EMPs, it is required for their subsequent differentiation into erythroid and granulocyte lineages. Surprisingly, the persistence of EMPs, while reduced, may provide a source of definitive macrophages in Myb-/- embryos. Disclosures No relevant conflicts of interest to declare.


2018 ◽  
Vol 11 (5) ◽  
pp. 1061-1074 ◽  
Author(s):  
Eva Garcia-Alegria ◽  
Sara Menegatti ◽  
Muhammad Z.H. Fadlullah ◽  
Pablo Menendez ◽  
Georges Lacaud ◽  
...  

Blood ◽  
1997 ◽  
Vol 90 (5) ◽  
pp. 1799-1806 ◽  
Author(s):  
Jay L. Hess ◽  
Benjamin D. Yu ◽  
Bin Li ◽  
Rob Hanson ◽  
Stanley J. Korsmeyer

Abstract Translocations involving the mixed lineage leukemia gene (MLL ), the human homolog of the Drosophila gene trithorax, are one of the most common genetic alterations in human acute leukemias. Each translocation involving MLL results in loss of one functional copy of MLL and the generation of a chimeric fusion protein with potential dominant negative or neomorphic activity. Mll is a positive regulator of Hox genes, which have been implicated in both axial skeleton patterning and hematopoietic development. Previous studies indicated that Hox gene expression is altered in Mll heterozygous (+/−) and homozygous (−/−) deficient mice. To study the role of Mll in hematopoiesis and to obtain insights into leukemogenesis, we have examined the effects of haplo-insufficiency or absence of Mll by in vitro differentiation of Mll +/+, +/−, and −/− yolk sac progenitor cells. Mll −/− colonies were fewer in number, took longer to develop, and contained fewer cells than their wild-type and heterozygous counterparts. Formation of colony-forming unit-granulocyte, erythroid, macrophage, megakaryocyte (CFU-GEMM), colony-forming unit-macrophage (CFU-M), and burst-forming unit-erythroid (BFU-E) was markedly decreased in Mll −/− cultures, while numbers of colony-forming unit-erythroid (CFU-E), colony-forming unit-granulocyte (CFU-G), and colony-forming unit-granulocyte macrophage (CFU-GM) were essentially unaffected. Despite the decreased numbers of colonies present, Mll −/− cultures showed all cell types without morphologic evidence of maturation arrest. These studies indicate that Mll is required for normal numbers of hematopoietic progenitors and their proper differentiation, especially along the myeloid and macrophage pathways.


Stem Cells ◽  
2015 ◽  
Vol 34 (2) ◽  
pp. 431-444 ◽  
Author(s):  
Jenna M. Frame ◽  
Katherine H. Fegan ◽  
Simon J. Conway ◽  
Kathleen E. McGrath ◽  
James Palis

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2434-2434
Author(s):  
R. Katherine Hyde ◽  
Ling Zhao ◽  
Lemlem Alemu ◽  
Pu Paul Liu

Abstract Abstract 2434 Acute myeloid leukemia (AML) is often associated with specific, recurrent chromosomal abnormalities, such as the inversion of chromosome 16 (Inv(16)) which is associated with subtype M4 with eosinophilia. This inversion creates a fusion between CBFB and MYH11, which encode Core Binding Factor beta and Smooth Muscle Myosin Heavy Chain, respectively. The resulting fusion gene, CBFB-MYH11, is known to be the initiating factor in Inv(16) AML, but its mechanism is not clear. Previous studies indicated that repression of RUNX1 is a potential mechanism. However, we found that Cbfb-MYH11 has activities independent of Runx1 repression. During primitive hematopoiesis, we showed that expression of Cbfb-MYH11 in knockin mouse embryos (Cbfb+/MYH11) caused defects in differentiation that were not seen in embryos nullizygous for Runx1 (Runx1−/−), indicating that Cbfb-MYH11 has activities in addition to the repression of Runx1. Moreover, we found that the defects in the primitive hematopoiesis were rescued in the Cbfb+/MYH11; Runx1−/− embryos, which suggests that Runx1 is required for Cbfb-MYH11 activity during primitive hematopoiesis. We next asked whether Cbfb-MYH11 was similarly dependent on Runx1 during definitive hematopoiesis. For this purpose we used mice expressing another allele of Runx1 in which a 3'-truncated Runx1 is fused to the b-galactosidase gene, lacZ (Runx1lzd). This Runx1 allele has been reported to have dominant negative activities. Using an in vitro promoter assay, we found that co-expression of Cbfβ with Runx1 and Runx1-lzd resulted in decreased activation of the MCSFR promoter as compared to co-expressing Cbfβ and Runx1, indicating that Runx1-lzd has dominant negative activities. In addition, we found that expression of a single Runx1-lzd allele rescued the primitive blood defect in the Cbfb+/MYH11 embryos. Runx1+/lzd; Cbfb+/MYH11 embryos showed almost normal definitive hematopoiesis providing further evidence that Runx1-lzd has dominant negative activity. Previously we showed that induction of Cbfb-MYH11 results in a distinct population of pre-leukemic cells. By combining the Runx1-lzd allele with an inducible allele of Cbfb-MYH11, we examined the requirement for Runx1 activity in the production of pre-leukemic cells. We found that 7 days after induction of Cbfb-MYH11, Runx1+/lzd; Cbfb+/MYH11 mice showed a statistically significant decrease in the number of pre-leukemic cells as compared to Runx1+/+; Cbfb+/MYH11 mice. We also found a statistically significant decrease in BrdU incorporation in the bone marrow of Runx1+/lzd; Cbfb+/MYH11 mice as compared to Runx1+/+; Cbfb+/MYH11 mice. This indicates that Runx1 is important for Cbfb-MYH11 activity in adult hematopoietic cells. Consistent with this idea, we found that adult mice expressing Cbfb-MYH11 and the Runx1-lzd allele showed a significant delay in the development of leukemia as compared to their Cbfb+/MYH11; Runx1+/+ littermates. Collectively, this work implies that RUNX1 is important for CBFB-MYH11 activity and that inhibitors of RUNX1 have potential use for the treatment of Inv(16) leukemia. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1996 ◽  
Vol 88 (10) ◽  
pp. 3720-3730 ◽  
Author(s):  
M Bielinska ◽  
N Narita ◽  
M Heikinheimo ◽  
SB Porter ◽  
DB Wilson

During mouse embryogenesis the first hematopoietic and endothelial cells form in blood islands located between layers of visceral endoderm and mesoderm in the yolk sac. The role of visceral endoderm in primitive hematopoiesis and vasculogenesis is not well understood. We have assessed the consequences of a lack of visceral endoderm on blood cell and vessel formation using embryoid bodies derived from mouse embryonic stem (ES) cells deficient in GATA-4, a transcription factor expressed in yolk sac endoderm. When differentiated in vitro, these mutant embryoid bodies do not develop an external visceral endoderm layer. We found that Gata4-/-embryoid bodies, grown either in suspension culture or attached to a substratum, are defective in primitive hematopoiesis and vasculogenesis as evidenced by a lack of recognizable blood islands and vascular channels and a reduction in the expression of the primitive erythrocyte marker epsilon y-globin. Expression of the endothelial cell transcripts FIk-1, FIt-1, and platelet-endothelial cell adhesion molecule (PECAM) was not affected in the mutant embryoid bodies. Gata4-/-ES cells retained the capacity to differentiate into primitive erythroblasts and endothelial cells when cultured in methylcellulose or matrigel. Analysis of chimeric mice, generated by injecting Gata4-/-ES cells into 8-cell stage embryos of ROSA26 transgenic animals, showed that Gata4-/-ES cells can form blood islands and vessels when juxtaposed to visceral endoderm in vivo. We conclude that the visceral endoderm is not essential for the differentiation of primitive erythrocytes or endothelial cells, but this cell layer plays an important role in the formation and organization of yolk sac blood islands and vessels.


Author(s):  
Alessandro Fantin ◽  
Carlotta Tacconi ◽  
Emanuela Villa ◽  
Elena Ceccacci ◽  
Laura Denti ◽  
...  

In the mouse embryo, endothelial cell (EC) progenitors almost concomitantly give rise to the first blood vessels in the yolk sac and the large vessels of the embryo proper. Although the first blood cells form in the yolk sac before blood vessels have assembled, consecutive waves of hematopoietic progenitors subsequently bud from hemogenic endothelium located within the wall of yolk sac and large intraembryonic vessels in a process termed endothelial-to-hematopoietic transition (endoHT). The receptor tyrosine kinase KIT is required for late embryonic erythropoiesis, but KIT is also expressed in hematopoietic progenitors that arise via endoHT from yolk sac hemogenic endothelium to generate early, transient hematopoietic waves. However, it remains unclear whether KIT has essential roles in early hematopoiesis. Here, we have combined single-cell expression studies with the analysis of knockout mice to show that KIT is dispensable for yolk sac endoHT but required for transient definitive hematopoiesis in the fetal liver.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 730-730 ◽  
Author(s):  
Kathleen E. McGrath ◽  
Jenna M. Cacciatori ◽  
Anne D. Koniski ◽  
James Palis

Abstract In the mouse embryo, hematopoietic function is required by E10.5 (embryonic day 10.5) before adult-repopulating hematopoietic stem cells (HSC) exist. The earliest erythroid function is provided by a wave of primitive erythroid progenitors that arise at E7.5, in association with megakaryocyte and macrophage progenitors. Intriguingly, a second wave of hematopoietic potential arises between the first primitive hematopoietic wave and functional HSC formation. This second progenitor wave also forms in the yolk sac but is distinguished from the primitive wave by its slightly later onset (E8.25), generation of definitive erythroid cells, and its additional association with granulocyte and mast cell progenitors. The proposed function of these “wave 2” progenitors is to colonize the newly formed fetal liver (beginning at E10) and differentiate into the first mature definitive erythroid cells observed in circulation at E12. However, it is unclear how much definitive hematopoiesis arising in the yolk sac recapitulates the paradigm of later HSC-derived myeloid potential, progenitor hierarchy and immunophenotype. To investigate this question, we examined markers of adult myeloid progenitor maturation in the yolk sac and early fetal liver. As previously described by others, all definitive hematopoietic progenitors in the yolk sac, unlike those in the bone marrow, express CD41, which we found associated with Fc gamma receptor expression (FcγR, CD16/32) beginning at E8.5. By E9.5, definitive hematopoietic progenitors can be identified by their surface co-expression of ckit, CD41, FcγR, as well as endoglin. When cultured in vitro, these cells can differentiate into all myeloid lineages, including neutrophils, eosinophils, basophils and mast cells as identified by morphology, immunophenotype and gene expression. Preliminary clonal analysis confirms that a common erythroid/granulocyte progenitor exists in this population. Consistent with adult myelopoiesis, we found a qualitative association of higher FcγR expression with granulocyte fate and higher endoglin expression associated with erythroid fate. However, the unusual co-expression of these four markers and the prevalence of erythroid fate, even in FcγRhi cells, suggest the definitive hematopoietic progenitors in the yolk sac may be quite plastic and highly predisposed to an erythroid fate. Consistent with the concept that these “wave 2” progenitors colonize the fetal liver, we also found similar ckit+CD41+FcγR+endoglin+ cells in the early liver (E11.5) with the potential to produce a variety of myeloid cells when cultured in vitro. The emergence of enucleated definitive erythrocytes by E12, within 24 hours HSC fetal liver colonization, implies that these first erythrocytes are derived from the yolk sac definitive progenitors found in the liver by E10.5. We therefore asked whether the multiple myeloid potentials associated with “wave 2” progenitors are similarily realized the early fetal liver. Beginning at E11.5 we found a population of Gr1+Mac1+ cells in the liver with morphological and histological characteristics of neutrophils, and increase 100-fold in number between E12.5 and E14.5. In contrast, we did not observe eosinophils, basophils or mast cells by morphology, immunophenotype or by RT-PCR for lineage-specific messages. We conclude that complete definitive myeloid potential first arises in the yolk sac from a unique population of ckit+FcγR+CD41+endoglin+ progenitors. Our data suggest that these progenitors then enter the fetal liver and differentiate into a subset of their potential fates producing the first mature definitive erythromyeloid cells. This second wave of hematopoietic progenitors emerging from the yolk sac thus serves as a novel model of mulitpotential definitive hematopoiesis.


Sign in / Sign up

Export Citation Format

Share Document