Diverse Myeloid Lineage Potential Arises in the Yolk Sac of the Mammalian Embryo.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1666-1666
Author(s):  
James Palis ◽  
Anne Koniski ◽  
Timothy P. Bushnell ◽  
Kathleen E. McGrath

Abstract The emergence of the hematopoietic system in the mammalian embryo is characterized by two temporally overlapping waves of distinct “primitive” and “definitive” erythroid progenitors that peak in the murine yolk sac at E8.5 and E10.5, respectively. We have recently determined that each wave contains megakaryocyte potential (Tober, et al., submitted), but the initial emergence of distinct myeloid lineages is less well understood. Temporal and spatial analysis of hematopoietic progenitors in the murine embryo suggest that the macrophage lineage is associated with both waves, while neutrophil and mast cell lineages are restricted to the second, definitive wave (Palis, et al., Development126:5073, 1999). To better define the emergence of myelopoiesis in the murine embryo, we cultured dissociated E9.5 yolk sac cells in a broad array of cytokines (SCF, IL3, IL5, IL6, IL7, IL9, IL11 and GM-CSF) for seven days and identified morphologically distinguishable macrophages, neutrophils, eosinophils, basophils, and mast cells. The presence of eosinophils was further confirmed by expression of eosinophil peroxidase transcripts. As expected, this multilineage myeloid potential was restricted to c-kit++ cells. However, unlike the bone marrow, these c-kit++ cells in the E9.5 yolk sac were predominately a single population that also express CD41 and Fc gamma receptor (FcγR, CD16/32). FcγR+ cells first emerge in the yolk sac between E8.5 and E9.5 and are not found in the embryo proper until E10.5, when they are restricted to the liver. These findings suggest that the expression of FcγR is restricted in the yolk sac to definitive hematopoiesis and tracks its transition to the fetal liver. While mature definitive erythroid cells first emerge from the fetal liver at E12.5, to our knowledge the onset of granulopoiesis has not been investigated in the early murine embryo. We found approximately 1000 GR1+/Mac1+ cells in the fetal liver at E12.5 and this population expands 100-fold during the next two days of development. These GR1+/Mac1+ cells consisted predominantly of progressively maturing neutrophils. We conclude that the potential to generate multiple myeloid lineages first arises in the yolk sac along with definitive erythroid progenitors and that a robust granulocyte population subsequently emerges in the fetal liver at midgestation.

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 ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2216-2216
Author(s):  
Zachary C. Murphy ◽  
Kathleen E. McGrath ◽  
James Palis

The mammalian embryo requires the sequential circulation of primitive and definitive erythroid cells for survival and growth. The central cytokine regulator of primitive and definitive erythropoiesis is erythropoietin (EPO). We have previously determined that primitive erythroid progenitors, unlike their definitive (CFU-E) counterparts, do not require EPO for survival. However, the mechanisms regulating the EPO-independent emergence of primitive erythropoiesis in the yolk sac remains poorly understood. Interestingly, maturing primitive erythroblasts in the murine embryo, unlike definitive erythroblasts in the fetal liver and adult bone marrow, express not only STAT5 but also STAT3, which is tyrosine phosphorylated at baseline and in response to EPO. These initial findings led is to hypothesize that STATs 5 and 3 differentially regulate terminal differentiation of primitive erythroid cells. To analyze the function of these two STATs in primary erythroid cells, we developed an imaging flow cytometry-based methodology to quantitate total and phosphorylated levels of STAT proteins in small numbers of cells isolated from staged murine embryos. We found that STAT5 plays conserved roles in primitive and definitive erythroblast survival and surface CD71 expression. In contrast, STAT3 regulates cell cycle progression and mitochondrial polarization specifically in primitive erythroblasts. In addition, STAT3, unlike STAT5, was phosphorylated in the absence of cytokine stimulation. We asked if reactive oxygen species (ROS) may be activating STAT3, since primary primitive erythroblasts, unlike definitive erythroblasts, specifically express Aquaporins 3 and 8, which can transport hydrogen peroxide, as well as water. Indeed, hydrogen peroxide exposure increased endogenous ROS, as well as phosphorylated STAT3, levels both in wild-type and EPOR-null primitive erythroblasts. Consistent with these findings, inhibition of aquaporin channel transport prevented STAT3 phosphorylation by exogenous hydrogen peroxide, but not by EPO. Taken together, our data support the concept that the primitive erythroid lineage, emerging in the hypoxic and EPO-low environment of the yolk sac in the pre-circulation murine embryo, uniquely integrates ROS-mediated STAT3 activation to regulate key aspects of terminal erythroid maturation. Disclosures Palis: Rubies Therapeutics: Consultancy.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 566-566
Author(s):  
James Palis ◽  
Joanna Tober ◽  
Radhika Vemishetti ◽  
Anne Koniski ◽  
Richard Waugh

Abstract Two distinct waves of hematopoietic progenitors originate in the yolk sac of the mammalian embryo. The first “primitive” wave contains primitive erythroid and macrophage progenitors that generate the embryo’s first red cells (Palis, et al. Development126:5073, 1999). The second “definitive” wave consists of definitive erythroid (BFU-E) and multiple myeloid progenitors that arise later in the yolk sac and are subsequently found in the fetal liver and postnatal marrow. While megakaryocyte progenitors have been detected in the yolk sac, the ontogeny of the megakaryocyte lineage is poorly understood. Furthermore, it is not been determined when platelets first enter the bloodstream of the mouse embryo. The presence and size of platelets in the embryonic bloodstream were examined by microscopy after staining with anti-GPV antibodies. Rare platelets were identified in 2 of 4 litters of mice at E10.5. These platelets were very large with a diameter of 4.2 ± 0.4 (mean ± SEM) microns. Platelet size remained large (4.0-3.8 microns) at E11.5–E12.5, but decreased to 3.3-3.2 microns in diameter between E13.5 and E15.5 of gestation. At birth, the mean platelet diameter (2.8 ± 0.04 microns) was similar to that of adult mice (2.7 microns). These results indicate that large, embryonic platelets begin to circulate in the mouse embryo beginning at E10.5 and raise the possibility that embryonic, fetal, and adult waves of platelets are produced during mammalian embryogenesis. Using a collagen-based culture system, megakaryocyte progenitors (Meg-CFC) were first identified in late primitive streak embryos (E7.25), concomitant with primitive erythroid progenitors. Meg-CFC numbers subsequently expand in the yolk sac along with BFU-E before the development of the fetal liver. To examine the relationship of the megakaryocyte and primitive erythroid lineages in the yolk sac, we stained hematopoietic colonies grown in collagen for 5–10 days with anti-GP1bβ (megakaryocyte) and anti-βH1-globin (primitive erythroid) antibodies. As expected, the majority of the stained colonies were primitive erythroid (containing only βH1-globin-positive cells) and approximately 15% of the colonies were megakaryocyte (containing only GP1bβ-positive cells). However, 15% of the colonies contained both GP1bβ- and βH1-globin-positive cells consistent with an origin from bipotential primitive erythroid/megakaryocyte progenitors. Furthermore, proplatelet formation was evident in both unipotential and bipotential megakaryocyte colonies cultured for 10 days. Our studies support the concept that the megakaryocyte and primitive erythroid lineages originate in the yolk sac from a bipotential precursor. This parallels lineage relationships in the bone marrow which contains bipotential definitive erythroid/megakaryocyte progenitors. Finally, we hypothesize that yolk sac-derived “primitive” Meg-CFC give rise to the first embryonic platelets that enter the bloodstream soon after the onset of circulation as the fetal liver becomes a hematopoietic organ.


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 ◽  
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 ◽  
2008 ◽  
Vol 111 (7) ◽  
pp. 3435-3438 ◽  
Author(s):  
Christopher T. Lux ◽  
Momoko Yoshimoto ◽  
Kathleen McGrath ◽  
Simon J. Conway ◽  
James Palis ◽  
...  

Abstract The relative contribution of yolk sac (YS)–derived cells to the circulating definitive hematopoietic progenitor cell (HPC) pool that seeds the fetal liver remains controversial due to the presence of systemic circulation and the onset of hematopoiesis within the embryo proper (EP) before liver seeding. Ncx1−/− embryos fail to initiate a heartbeat on embryonic day (E) 8.25, but continue to develop through E10. We detected normal numbers of primitive erythroid progenitors in Ncx1−/− versus wild type (WT) YS, but primitive erythroblasts did not circulate in the Ncx1−/− EP. While there was no significant difference in the number of definitive HPCs in Ncx1−/− versus WT YS through E9.5, the Ncx1−/− EP was nearly devoid of HPCs. Thus, primitive erythroblasts and essentially all definitive HPCs destined to initially seed the fetal liver after E9.5 are generated in the YS between E7.0-E9.5 and are redistributed into the EP via the systemic circulation.


2021 ◽  
Vol 218 (4) ◽  
Author(s):  
Francisca Soares-da-Silva ◽  
Laina Freyer ◽  
Ramy Elsaid ◽  
Odile Burlen-Defranoux ◽  
Lorea Iturri ◽  
...  

In the embryo, the first hematopoietic cells derive from the yolk sac and are thought to be rapidly replaced by the progeny of hematopoietic stem cells. We used three lineage-tracing mouse models to show that, contrary to what was previously assumed, hematopoietic stem cells do not contribute significantly to erythrocyte production up until birth. Lineage tracing of yolk sac erythromyeloid progenitors, which generate tissue resident macrophages, identified highly proliferative erythroid progenitors that rapidly differentiate after intra-embryonic injection, persisting as the major contributors to the embryonic erythroid compartment. We show that erythrocyte progenitors of yolk sac origin require 10-fold lower concentrations of erythropoietin than their hematopoietic stem cell–derived counterparts for efficient erythrocyte production. We propose that, in a low erythropoietin environment in the fetal liver, yolk sac–derived erythrocyte progenitors efficiently outcompete hematopoietic stem cell progeny, which fails to generate megakaryocyte and erythrocyte progenitors.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2539-2539
Author(s):  
Kathleen E. McGrath ◽  
Jenna M Frame ◽  
George Fromm ◽  
Anne D Koniski ◽  
Paul D Kingsley ◽  
...  

Abstract Abstract 2539 Poster Board II-516 A transient wave of primitive erythropoiesis begins at embryonic day 7.5 (E7.5) in the mouse as yolk sac-derived primitive erythroid progenitors (EryP-CFC) generate precursors that mature in the circulation and expand in numbers until E12.5. A second wave of erythroid progenitors (BFU-E) originates in the yolk sac beginning at E8.25 that generate definitive erythroid cells in vitro. These BFU-E colonize the newly forming liver beginning at E10.5, prior to the initial appearance there of adult-repopulating hematopoietic stem cells (HSCs) between E11.5-12.5. This wave of definitive erythroid yolk sac progenitors is proposed to be the source of new blood cells required by the growing embryo after the expansion of primitive erythroid cells has ceased and before HSC-derived hematopoiesis can fulfill the erythropoietic needs of the embryo. We utilized multispectral imaging flow cytometry both to distinguish erythroid lineages and to define specific stages of erythroid precursor maturation in the mouse embryo. Consistent with this model, we found that small numbers of definitive erythrocytes first enter the embryonic circulation beginning at E11.5. All maturational stages of erythroid precursors were observed in the E11.5 liver, consistent with these first definitive erythrocytes having rapidly completed their maturation in the liver. The expression of βH1 and εy-beta globin genes is thought to be limited to primitive erythroid cells. Surprisingly, examination of globin gene expression by in situ hybridization revealed high levels of βH1-, but not εy-globin, transcripts in the parenchyma of E11.5-12.5 livers. RT-PCR analysis of globin mRNAs confirmed the expression of βH1- and adult β1-, but not εy-globin, in E11.5 liver-derived definitive (ckit+, Ter119lo) proerythroblasts sorted by flow cytometry to remove contaminating primitive (ckit-, Ter119+) erythroid cells. A similar pattern of globin gene expression was found in individual definitive erythroid colonies derived from E9.5 yolk sac and from early fetal liver. In vitro differentiation of definitive erythroid progenitors from E9.5 yolk sac revealed a maturational “switch” from βH1- and β1-globins to predominantly β1-globin. βH1-globin transcripts were not observed in proerythroblasts from bone marrow or E16.5 liver or in erythroid colonies from later fetal liver. ChIP analysis revealed that hyperacetylated domains encompass all beta globin genes in primitive erythroid cells but only the adult β1- and β2-globin genes in E16.5 liver proerythroblasts. Consistent with their unique gene expression, E11.5 liver proerythroblasts have hyperacetylated domains encompassing the βh1-, β1- and β2-, but not εy-globin genes. We also examined human globin transgene expression in mice carrying a single copy of the human beta globin locus. Because of the overlapping presence and changing proportion of primitive and definitive erythroid cells during development, we analyzed sorted cell populations whose identities were confirmed by murine globin gene expression. We confirmed that primitive erythroid cells express higher levels of γ- than ε-globin and little β-globin. E11.5 proerythroblasts and cultured E9.5 progenitors express γ- and β-, but not ε-globin. E16.5 liver proerythroblasts express β- and low levels of γ-globin, while adult marrow proerythroblasts express only β-globin transcripts. In summary, two forms of definitive erythropoiesis emerge in the murine embryo, each with distinct globin expression patterns and chromatin modifications of the β-globin locus. While both lineages predominantly express adult globins, the first, yolk sac-derived lineage uniquely expresses low levels of the embryonic βH1-globin gene as well as the human γ-globin transgene. The second definitive erythroid lineage, found in the later fetal liver and postnatal marrow, expresses only adult murine globins as well as low levels of the human γ-globin transgene only in the fetus. Our studies reveal a surprising complexity to the ontogeny of erythropoiesis. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2305-2305
Author(s):  
Kathleen E McGrath ◽  
Paul D Kingsley ◽  
James R Bowen ◽  
Jennifer L McLaughlin ◽  
James Palis

Abstract Abstract 2305 In the adult, all platelets are derived from hematopoietic stem cells (HSCs). However, we previously determined that the megakaryocyte (meg) lineage is specified several days before HSC emergence in the murine embryo and that circulating platelets exist in myb-null embryos, which lack HSCs. Pre-HSC meg progenitors arise in the yolk sac beginning at embryonic day 7.5 (E7.5) and have lower proliferative potential than adult meg progenitors. The fetal liver is colonized by over 1,000 meg progenitors by E10.5, before HSCs are found there. By E12.5, there are over one million circulating embryonic platelets that are larger than adullt platelets with smaller α-granules. There are also indications in humans of intrinsic differences between embryonic/fetal and adult thrombopoiesis, including the natural history of several congenital platelet disorders, as well as the small size, rapid maturation, and reduced platelet output of fetal/neonatal meg progenitors. We compared embryonic versus adult megakaryopoiesis and platelet function in the mouse. E12.5 fetal livers contain predominantly small megs with low ploidy (8% >4N versus 33% >4N in the adult marrow). Fetal megs have higher cell surface levels of CD41 and GP1bß and are larger than similar ploidy adult megs. Further evidence of rapid maturation of fetal megs was seen in the punctate localization pattern of endostatin and the presence of α-granules and a forming demarcation membrane system in small E12.5 liver megs. Like their primary counterparts, in vitro-generated embryonic megs from E9.5 yolk sac progenitors have lower ploidy than megs differentiated from bone marrow progenitors and show similar evidence of rapid cytoplasmic maturation. Initial analysis of megs generated from ES cells demonstrated low ploidy and rapid maturation similar to yolk sac-derived megs. These data support the concept that embryonic megakaryopoiesis is characterized by a rapid maturation and low ploidy phenotype that is cell intrinsic and is similar to that observed during ES cell megakaryopoiesis. An analysis of primary fetal versus adult platelets reveals similar patterns of VEGF and endostatin distribution in α-granules. However, there are differences in the expression of several other factors associated with platelet activation and function, including higher expression of the thrombin receptor PAR1 and lower expression of the ADP receptor P2Y12 and P-selectin in embryonic platelets. While primary adult and embryonic platelets have altered side scatter characteristics and binding of the activation-specific Jon/A antibody after thrombin treatment, embryonic platelets fail to express P-selectin on their surface. Taken together, our findings indicate that embryonic/fetal megakaryopoiesis is characterized by low ploidy and rapid maturation, and leads to the generation of platelets with marked differences in size, structure and function compared with adult platelets. A better understanding of hematopoietic ontogeny is particularly relevant to the generation of blood cells from embryonic stem (ES) cells and induced pluripotent stem (iPS) cells, whose differentiation recapitulate early embryonic development. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3236-3236
Author(s):  
Stefan Pasichnyk Tarnawsky ◽  
Momoko Yoshimoto ◽  
Gordon Chan ◽  
Benjamin Neel ◽  
Rebecca J. Chan ◽  
...  

Abstract Juvenile Myelomonocytic Leukemia is the most common pediatric myeloproliferative neoplasm (MPN). JMML is characterized by myeloid populations with mutually-exclusive mutations in Ras-Erk signaling genes, most commonly PTPN11, which confer growth hypersensitivity to GM-CSF. JMML is notable among pediatric MPNs as being refractory to chemotherapy and having a 50% relapse rate following allogeneic hematopoietic stem cell (HSC) transplantation. As such, there is an urgent need for novel JMML therapies. The recent discovery of yolk sac myeloid lineages that persist into adulthood independently of bone marrow HSC contributions suggests a mechanism for JMML relapse following HSC transplantation. In this study, we sought to determine whether yolk sac HSC-independent myeloid progenitors bear hallmarks of MPN in a mouse model of JMML. Using the Vav1 promoter-directed Cre recombinase, we generated a mouse model of JMML that expresses the PTPN11D61Y gain of function mutation in all waves of embryonic and adult hematopoiesis, including yolk sac myeloid progenitors that emerge prior to and independently from HSCs. PTPN11D61Y/+; VavCre+ mice are viable, born at expected Mendelian ratios, and develop peripheral blood monocytosis as early as 4 weeks of age. Given this early onset, we hypothesized MPN may develop in these mice during embryonic development. E14.5 fetal liver progenitors from PTPN11D61Y/+; VavCre+ embryos displayed marked GM-CSF hypersensitivity in methylcellulose colony forming assays (Figure-1A), possessed hyperactive Ras-Erk pathway signaling (Figure-2), and had a skewed progenitor distribution with a greater proportion of megakaryocyte-erythroid progenitors (63.5% vs. 50.1%, p<0.01) and fewer common myeloid progenitors (9.2% vs. 19.3%, p<0.01) than littermate controls. Since the E14.5 fetal liver contains both HSC-dependent and HSC-independent myeloid progenitors, we repeated these experiments using E9.5 yolk sac samples to restrict our analysis to HSC-independent hematopoiesis. Compared to littermate controls, PTPN11D61Y/+; VavCre+ E9.5 embryos had no difference in overall number of yolk sac myeloid progenitors (Ter119-; cKit+, CD41Dim), and in the number and distribution of CFU-E/GM/GEMM colonies in methylcellulose assay. However, PTPN11D61Y/+; VavCre+ yolk sac progenitors demonstrated marked GM-CSF hypersensitivity in colony forming assay (Figure-1B) as well as hyperactive Ras-Erk signalling by western blot analysis (Figure-2) and intracellular flow cytometry with antibodies against pSTAT5 and pERK (Figure-3). We have demonstrated that HSC-independent myeloid lineages from the murine yolk sac possess GM-CSF hypersensitivity and Ras-Erk pathway hyperactivation in a mouse model of JMML. These findings suggest that HSC-independent hematopoietic populations may be involved in the development of JMML. Our study highlights the need to further assess the role of bone marrow-independent myeloid lineages in pediatric MPN, and to identify innovative therapies that can specifically target HSC-independent hematopoietic lineages. Figure 1. Embryonic myeloid progenitors from PTPN11D61Y/+; VavCre+ embryos demonstrate GM-CSF growth hypersensitivity. A) E14.5 fetal liver mononuclear cells (n=7) or B) E9.5 yolk sac cells (n=8) were plated in methylcellulose colony forming assays and colonies were counted 7 days later. * p<0.05; ** p<0.001 by two-tailed Student’s t-test. Figure 1. Embryonic myeloid progenitors from PTPN11D61Y/+; VavCre+ embryos demonstrate GM-CSF growth hypersensitivity. A) E14.5 fetal liver mononuclear cells (n=7) or B) E9.5 yolk sac cells (n=8) were plated in methylcellulose colony forming assays and colonies were counted 7 days later. * p<0.05; ** p<0.001 by two-tailed Student’s t-test. Figure 2. Western blot analysis demonstrates hyperactive Ras-Erk signaling in E14.5 fetal liver and E9.5 yolk sac progenitors from PTPN11D61Y/+; VavCre+ embryos. Cultured progenitors were starved overnight and stimulated with GM-CSF for 60min prior to protein extraction. p, phosphorylated protein; t, total protein; WT, wild type. Figure 2. Western blot analysis demonstrates hyperactive Ras-Erk signaling in E14.5 fetal liver and E9.5 yolk sac progenitors from PTPN11D61Y/+; VavCre+ embryos. Cultured progenitors were starved overnight and stimulated with GM-CSF for 60min prior to protein extraction. p, phosphorylated protein; t, total protein; WT, wild type. Figure 3. Yolk Sac Myeloid Progenitors from PTPN11D61Y/+; VavCre+ embryos demonstrate Ras-Erk pathway hypersensitivity at baseline and following GM-CSF stimulation. Cultured yolk sac progenitors were stimulated for 30min with 5ng/ml of GM-CSF, processed for intracellular flow cytometry, and stained with the indicated fluorescent antibody. Histograms display representative median fluorescence intensity among CD45+ cells in each sample (n=3). * p<0.05 by two-tailed Student’s t-test. ADDIN EN.REFLIST Figure 3. Yolk Sac Myeloid Progenitors from PTPN11D61Y/+; VavCre+ embryos demonstrate Ras-Erk pathway hypersensitivity at baseline and following GM-CSF stimulation. Cultured yolk sac progenitors were stimulated for 30min with 5ng/ml of GM-CSF, processed for intracellular flow cytometry, and stained with the indicated fluorescent antibody. Histograms display representative median fluorescence intensity among CD45+ cells in each sample (n=3). * p<0.05 by two-tailed Student’s t-test. ADDIN EN.REFLIST Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document