scholarly journals KIT Is Required for Fetal Liver Hematopoiesis

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.

2021 ◽  
Author(s):  
Alessandro Fantin ◽  
Alice Plein ◽  
Carlotta Tacconi ◽  
Emanuela Villa ◽  
Elena Ceccacci ◽  
...  

AbstractBlood vessels are fundamental to sustain organ growth and tissue metabolism. 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. Thereafter, the vascular network expands by angiogenesis to vascularize developing organs such as the brain. 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 earlier in the hemogenic endothelium, in hematopoietic progenitors that arise via endoHT from hemogenic endothelium and non-hemogenic ECs, such as in the brain. However, it remains unclear whether KIT has essential roles in early hematopoiesis or even blood vessel growth. Here, we have combined transcriptomic analysis to delineate Kit expression with the analysis of knockout mice to show that KIT is expressed during but dispensable for yolk sac endoHT or brain angiogenesis but required for transient definitive erythropoiesis in the fetal liver.


Blood ◽  
2003 ◽  
Vol 101 (2) ◽  
pp. 508-516 ◽  
Author(s):  
Hanna K. A. Mikkola ◽  
Yuko Fujiwara ◽  
Thorsten M. Schlaeger ◽  
David Traver ◽  
Stuart H. Orkin

Murine hematopoietic stem cells (HSCs) originate from mesoderm in a process that requires the transcription factor SCL/Tal1. To define steps in the commitment to blood cell fate, we compared wild-type and SCL−/− embryonic stem cell differentiation in vitro and identified CD41 (GpIIb) as the earliest surface marker missing from SCL−/− embryoid bodies (EBs). Culture of fluorescence-activated cell sorter (FACS) purified cells from EBs showed that definitive hematopoietic progenitors were highly enriched in the CD41+ fraction, whereas endothelial cells developed from CD41− cells. In the mouse embryo, expression of CD41 was detected in yolk sac blood islands and in fetal liver. In yolk sac and EBs, the panhematopoietic marker CD45 appeared in a subpopulation of CD41+ cells. However, multilineage hematopoietic colonies developed not only from CD45+CD41+ cells but also from CD45−CD41+ cells, suggesting that CD41 rather than CD45 marks the definitive culture colony-forming unit (CFU-C) at the embryonic stage. In contrast, fetal liver CFU-C was CD45+, and only a subfraction expressed CD41, demonstrating down-regulation of CD41 by the fetal liver stage. In yolk sac and EBs, CD41 was coexpressed with embryonic HSC markers c-kit and CD34. Sorting for CD41 and c-kit expression resulted in enrichment of definitive hematopoietic progenitors. Furthermore, the CD41+c-kit+ population was missing from runx1/AML1−/− EBs that lack definitive hematopoiesis. These results suggest that the expression of CD41, a candidate target gene of SCL/Tal1, and c-kit define the divergence of definitive hematopoiesis from endothelial cells during development. Although CD41 is commonly referred to as megakaryocyte–platelet integrin in adult hematopoiesis, these results implicate a wider role for CD41 during murine ontogeny.


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.


Blood ◽  
2002 ◽  
Vol 100 (3) ◽  
pp. 1072-1074 ◽  
Author(s):  
Sandra Ziegler ◽  
Kurt Bürki ◽  
Radek C. Skoda

Abstract Thrombopoietin receptor c-mpl is expressed on hematopoietic progenitors and cells of the megakaryocytic lineage. The c-mpl promoter may, therefore, be useful for directing the expression of transgenes. We tested whether a 2-kb genomic DNA fragment comprising the putative c-mpl regulatory elements and most of the 5′-untranslated region of mouse c-mpl is able to direct the expression of a reporter gene to hematopoietic cells in transgenic mice. As a reporter gene we used the human placental alkaline phosphatase (PLAP). In adult transgenic mice, PLAP expression was specifically detected in megakaryocytes and platelets. Embryos showed PLAP reporter gene expression already in the yolk sac at embryonic day 6.5 (E6.5) and in blood islands at E7.5. At E9.5, expression was found in blood vessels of the yolk sac and the embryo proper, followed by high levels of expression in the fetal liver at E11.5. Expression in E6.5 yolk sac is compatible with a function of c-mpl and its ligand, thrombopoietin, in the earliest stages of embryonic hematopoiesis.


Blood ◽  
2009 ◽  
Vol 114 (12) ◽  
pp. 2401-2410 ◽  
Author(s):  
Deborah J. Stumpo ◽  
Hal E. Broxmeyer ◽  
Toni Ward ◽  
Scott Cooper ◽  
Giao Hangoc ◽  
...  

Abstract Members of the tristetraprolin family of tandem CCCH finger proteins can bind to AU-rich elements in the 3′-untranslated region of mRNAs, leading to their deadenylation and subsequent degradation. Partial deficiency of 1 of the 4 mouse tristetraprolin family members, Zfp36l2, resulted in complete female infertility because of early embryo death. We have now generated mice completely deficient in the ZFP36L2 protein. Homozygous Zfp36l2 knockout (KO) mice died within approximately 2 weeks of birth, apparently from intestinal or other hemorrhage. Analysis of peripheral blood from KO mice showed a decrease in red and white cells, hemoglobin, hematocrit, and platelets. Yolk sacs from embryonic day 11.5 (E11.5) Zfp36l2 KO mice and fetal livers from E14.5 KO mice gave rise to markedly reduced numbers of definitive multilineage and lineage-committed hematopoietic progenitors. Competitive reconstitution experiments demonstrated that Zfp36l2 KO fetal liver hematopoietic stem cells were unable to adequately reconstitute the hematopoietic system of lethally irradiated recipients. These data establish Zfp36l2 as a critical modulator of definitive hematopoiesis and suggest a novel regulatory pathway involving control of mRNA stability in the life cycle of hematopoietic stem and progenitor cells.


Blood ◽  
2001 ◽  
Vol 98 (1) ◽  
pp. 6-12 ◽  
Author(s):  
Sahoko Matsuoka ◽  
Kohichiro Tsuji ◽  
Hiroaki Hisakawa ◽  
Ming-jiang Xu ◽  
Yasuhiro Ebihara ◽  
...  

Abstract There is controversy as to whether murine definitive hematopoiesis originates from yolk sac (YS) or the intraembryonic region. This study reports the generation of definitive hematopoietic stem cells (HSCs) from both early YS and intraembryonic paraaortic splanchnopleures (P-Sp) on AGM-S3 stromal cells derived from the aorta-gonad-mesonephros (AGM) region at 10.5 days post coitum (dpc). YS and P-Sp cells at 8.5 dpc generated no definitive hematopoiesis-derived colony-forming cells in cocultures with AGM-S3 cells, but spleen colony-forming cells and HSCs capable of reconstituting definitive hematopoiesis in adult mice simultaneously appeared on day 4 of coculture. Precursors for definitive HSCs were present in YS and P-Sp at 8.0 dpc, a time when YS and embryo were not connected by blood vessels. It is proposed that precursors with the potential to generate definitive HSCs appear independently in YS and intraembryonic P-Sp and that the P-Sp or AGM region affords the microenvironment that facilitates generation of definitive hematopoiesis from precursors.


Blood ◽  
2003 ◽  
Vol 101 (5) ◽  
pp. 1669-1675 ◽  
Author(s):  
Kathleen E. McGrath ◽  
Anne D. Koniski ◽  
Jeffrey Malik ◽  
James Palis

To better understand the relationship between the embryonic hematopoietic and vascular systems, we investigated the establishment of circulation in mouse embryos by examining the redistribution of yolk sac–derived primitive erythroblasts and definitive hematopoietic progenitors. Our studies revealed that small numbers of erythroblasts first enter the embryo proper at 4 to 8 somite pairs (sp) (embryonic day 8.25 [E8.25]), concomitant with the proposed onset of cardiac function. Hours later (E8.5), most red cells remained in the yolk sac. Although the number of red cells expanded rapidly in the embryo proper, a steady state of approximately 40% red cells was not reached until 26 to 30 sp (E10). Additionally, erythroblasts were unevenly distributed within the embryo's vasculature before 35 sp. These data suggest that fully functional circulation is established after E10. This timing correlated with vascular remodeling, suggesting that vessel arborization, smooth muscle recruitment, or both are required. We also examined the distribution of committed hematopoietic progenitors during early embryogenesis. Before E8.0, all progenitors were found in the yolk sac. When normalized to circulating erythroblasts, there was a significant enrichment (20- to 5-fold) of progenitors in the yolk sac compared with the embryo proper from E9.5 to E10.5. These results indicated that the yolk sac vascular network remains a site of progenitor production and preferential adhesion even as the fetal liver becomes a hematopoietic organ. We conclude that a functional vascular system develops gradually and that specialized vascular–hematopoietic environments exist after circulation becomes fully established.


Blood ◽  
1998 ◽  
Vol 91 (9) ◽  
pp. 3134-3143 ◽  
Author(s):  
Tsukasa Okuda ◽  
Zhongling Cai ◽  
Shouli Yang ◽  
Noel Lenny ◽  
Chuhl-joo Lyu ◽  
...  

Abstract The t(8;21)-encoded AML1-ETO chimeric product is believed to be causally involved in up to 15% of acute myelogenous leukemias through an as yet unknown mechanism. To directly investigate the role of AML1-ETO in leukemogenesis, we used gene targeting to create anAML1-ETO “knock-in” allele that mimics the t(8;21). Unexpectedly, embryos heterozygous for AML1-ETO(AML1-ETO/+) died around E13.5 from a complete absence of normal fetal liver–derived definitive hematopoiesis and lethal hemorrhages. This phenotype was similar to that seen following homozygous disruption of either AML1 orCBFβ. However, in contrast to AML1- or CBFβ-deficient embryos, fetal livers from AML1-ETO/+ embryos contained dysplastic multilineage hematopoietic progenitors that had an abnormally high self-renewal capacity in vitro. To further document the role of AML1-ETO in these growth abnormalities, we used retroviral transduction to express AML1-ETO in murine adult bone marrow–derived hematopoietic progenitors. AML1-ETO–expressing cells were again found to have an increased self-renewal capacity and could be readily established into immortalized cell lines in vitro. Taken together, these studies suggest that AML1-ETO not only neutralizes the normal biologic activity of AML1 but also directly induces aberrant hematopoietic cell proliferation.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1599-1599
Author(s):  
Kathleen E McGrath ◽  
Jenna M Frame ◽  
Anne Koniski ◽  
Paul D Kingsley ◽  
James Palis

Abstract Abstract 1599 The ontogeny of hematopoiesis in mammalian embryos is complicated by the requirement for functional blood cells prior to the emergence of hematopoietic stem cells or the bone marrow microenvironment. In the murine embryo, transplantable HSC are first evident at embryonic day (E) 10.5 and the first few HSC are found in the fetal liver hematopoietic environment by E12.5. However, two overlapping waves of hematopoietic potential arise in the yolk sac before E10.5. The first “primitive” wave produces progenitors from E7.25 to E8.5 with primitive erythroid, megakaryocyte and macrophage potentials. The resulting primitive erythroid cells mature within the circulation and support embryonic growth past E9.5. At E8.5, a second wave of hematopoiesis begins in the yolk sac and generates definitive erythroid and multiple myeloid progenitors that are the proposed source of the hematopoietic progenitors seeding the fetal liver before HSC colonization. We have identified a cell population displaying a unique cell surface immunophenotype in the E9.5 yolk sac that contains the potential to form definitive erythroid cells, megakaryocytes, macrophages and all forms of granulocytes within days of in vitro culture. Furthermore, all definitive hematopoietic colony-forming cells (BFU-E, CFC-myeloid and HPP-CFC) in the E9.5 yolk sac have this immunophenotype. These erythro-myeloid progenitors (EMP) are lineage-negative and co-express ckit, CD41, CD16/32 and Endoglin. Interestingly, this is not an immunophenotype evident in the adult bone marrow. Other markers that have been associated with HSC formation (AA4.1, ScaI) or with lymphoid potential (IL7R, Flt3) are not present on these cells at E9.5. Consistent with the lack of lymphoid markers, we also do not observe short-term development of B-cells (CD19+B220+ expressing Rag2 RNA) in cultures of the E9.5 sorted EMP, while bone marrow Lin-/ckit+/ScaI- cells do form B-cells under the same conditions. Clonal analysis of sorted EMP cells revealed single cells with both erythroid and granulocyte potential, similar to the common myeloid progenitors in adult bone marrow. Though these EMP are enriched at E9.5 in the yolk sac, they are also found at low levels in the fetal blood, embryo proper and placenta, consistent with their entrance into the circulation. By E10.5, EMP were most highly enriched in the newly formed fetal liver. Additionally by E12.5, a time when the first few HSCs are detected in the fetal liver, we find active erythropoiesis and granulopoiesis in the liver and the first definitive red blood cells and neutrophils in the bloodstream. Therefore, we believe the yolk sac definitive progenitors' fate is to populate the fetal liver and thus provide the first definitive erythrocytes and granulocytes for the embryo. The differentiation of embryonic stem cells (ES) and induced pluripotent stem cells (iPS) cells into mature cells types offers the hope of cell-based therapies. Analysis of differentiating murine ES cells reveals overlapping waves of primitive and definitive hematopoietic colony forming potential. We demonstrate the appearance of an EMP-like (ckit+/CD41+/FcGR+) population coincident with the emergence of definitive hematopoietic progenitors during murine ES cell differentiation as embryoid bodies. We have confirmed with colony forming assays that definitive hematopoietic potential is associated with this immunophenotypic group. Our studies support the concept that blood cell emergence during ES cell differentiation closely mimics pre-HSC hematopoiesis in the yolk sac. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 910-910
Author(s):  
Kathleen E McGrath ◽  
Katherine H Fegan ◽  
Jenna M Frame ◽  
Paul D Kingsley ◽  
James Palis

Abstract Abstract 910 In the mammalian embryo, hematopoietic stem cells (HSC) emerge from vascular beds and colonize the fetal liver. The first HSC are found in the murine fetal liver by embryonic day 12.5 (E12.5). However blood function is required before HSC have formed, and two earlier waves of hematopoietic potential arise to sustain the embryo. The first wave of hematopoietic progenitors are formed in the yolk sac between E7.25 and E8.5 and are termed “primitive” because, along with megakaryocyte and macrophage potential, they differentiate into primitive erythroid cells that mature in the circulation and express embryonic globins. A second lineage of hematopoietic potential has been characterized in the murine and human yolk sac as well as in zebrafish. These cells have been termed “EMP”, erythro-myeloid progenitors, that generate definitive erythroid and myeloid lineages, including granulocytes. We find that EMP emerging in the mouse embryo express many of the markers associated with HSC emergence from hemogenic endothelium. At early stages of their emergence (E8.5), EMP express not only kit and CD41, but also VE-cadherin, CD31 and CD34. A day later, EMP constitute a robust population of over 1,000 cells in the yolk sac and display diminished expression of VE-cadherin and increased expression of CD45. However, unlike HSC, EMP do not express Sca1, but do express the myeloid progenitor marker CD16/32 (low affinity FCgamma receptor II/III). Like CD45, CD16/32 is expressed on a subset of the CD41+/kit+ cells at E8.5. Colony forming assays confirm that EMP potential is found in both CD16/32 positive and negative CD41+/kit+ cells. By E9.5, over 90% of CD41+/kit+ cells also express CD16/32 and all of the hematopoietic colony-forming potential at E9.5 is found in this triple-positive population. The transition from endothelial-associated to hematopoietic-associated genes suggests that EMP may emerge from hemogenic endothelial intermediates. Between E9.5 and E11.5, cells with the EMP immunophenotype are found in the bloodstream and become concentrated in the liver, where evidence of very robust erythro-myeloid differentiation precedes HSC colonization. In culture, EMP rapidly expand, dividing twice daily, and within 6 days generate predominately erythroid cells as well as smaller numbers of megakaryocyte, macrophage and mast cells, but only rare granulocytes. This is in contrast to lin-/kit+ ScaI- bone marrow progenitors grown in the same culture conditions that generate predominately myeloid cells, particularly granulocytes, and rarely mast cells. In agreement with data in the zebrafish, we also do not see evidence of lymphoid potential or RAG2 expression in EMP cultures or expression of the lymphoid markers Flt3 and IL7 receptor on the cell surface of EMP. In order to better understand the lineage potential of EMP, we examined the expression of known transcriptional regulators of bone marrow hematopoiesis. In the adult, relative levels of GATA1 versus Pu.1 are proposed to determine fate between megakaryocyte/erythroid progenitors (MEP- GATA1 hi) and granulocyte/macrophage progenitors (GMP-Pu.1 hi). Consistent with their erythro-myeloid potential, EMP expressed both GATA1 and Pu.1 at intermediate levels compared to adult marrow-derived MEP and GMP. In the adult, Gfi-1 and C/EBPalpha are both proposed to upregulate granulocyte versus macrophage differentiation. We found lower levels of these regulators in EMP compared to GMP, consistent with EMP cultures generating small numbers of granulocytes versus macrophages. In addition, the GATA1 expression within the Pu.1+ GMP is found to increase mast cell potential and, thus, the high GATA1 and Pu.1 expression in EMP may account for their high mast cell potential. Taken together, these data suggest that, like HSC, EMP emerge from hemogenic endothelium and their erythro-myeloid potential is governed by the action of shared regulatory networks. However, the transcription factors and markers are present in the EMP in unique combinations consistent with their specific role in providing a transient initial wave of definitive hematopoiesis in the embryo. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document