Human Embryonic Stem Cells Demonstrate That Mpl Expression Initially Appears on a Population of CD31+ Endothelial Cells with Hemogenic Potential.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3617-3617
Author(s):  
Tamihiro Kamata ◽  
Yuping Gong ◽  
Shaohui Wang ◽  
Andrew D. Leavitt

Abstract Mammalian hematopoiesis occurs adjacent to endothelial cells in the yolk sac and in the AGM region during fetal development, and in vivo cell labeling with Ac-LDL in fetal birds and mice indicates that hematopoietic cells can arise from endothelial cells. Blood forming potential has also been reported in endothelial cells isolated from human fetal liver and fetal bone marrow. However, the role of endothelial cells in the generation of hematopoietic cells remains poorly understood. While thrombopoietin (TPO) and its receptor, Mpl, are known for their critical role in megakaryocytopoiesis, Mpl−/ − mice and patients with congenital amegakaryocytic thrombocytopenia demonstrate that Mpl signaling is also important for the establishment and/or maintenance of hematopoietic stem cells. However, the nature of that role and the relevant Mpl-expressing cells remain unknown. To determine if Mpl signaling in early hematopoiesis involves hemogenic endothelium, we used human embryonic stem cells (hESCs) co-cultured with OP9 stromal cells, a robust model system for generating human hematopoietic cells. We found that CD34+ cells appeared by day 3–4 of co-culture, followed one day later by the appearance of CD31+ cells, nearly all of which appeared within the CD34+ population. The CD34+ cells increased to 20–40% of the total cell population by day 10, with nearly half co-expressing CD31+. Mpl+ cells first appeared on day 6–7, almost exclusively within the CD31+ population, increasing to 5–10% of total cells by day 10. The hESC:OP9 system generated a vascular-like network lined with von Willebrand Factor-expressing cells, which combined with FACS data showing the onset of Mpl expression on CD31+ cells, suggested that Mpl may first be expressed on vascular lining cells. Interestingly, Mpl expression precedes the detection of CD45+ cells, which are first observed on day 8–9. Moreover, round CD45+ cells appeared within the vascular networks, raising the possibility that vascular lining cells may give rise to hematopoietic cells. To begin to characterize the role of TPO/Mpl signaling in the CD31+ cell population, FACS-sorted CD31+ cells from day-9 and day-10 cultures were plated onto a fibronectin (FN)-coated surface, and non-adherent cells were removed after 1 hour. The adherent CD31+ cells uptake Ac-LDL and express vWF, characteristic of endothelial cells. When cultured in media supplemented with endothelial growth factors, including VEGF, EGF, bFGF, IGF-1, and heparin, the FN-adherent cells generated areas of cobblestone-like cell clusters and CD45+/CD34+ cells. When cultured in the same conditions plus TPO, the number of cobblestone-like clusters increased, and the number of CD45+/CD34+ hematopoietic cells generated increased 3–4 fold. Our data demonstrate that Mpl is expressed on a CD31+, vWF-expressing cell population with hemogenic potential, and that TPO/Mpl signaling increases the yield of hematopoietic cells generated from these cells. Ongoing sorting experiments, including the isolation and characterization of CD31+/Mpl+ cells, will further our understanding of where and how Mpl signaling affects early human hematopoietic development. HESCs provide a novel system for defining the role of TPO/Mpl signaling in early human hematopoietic development, which may lead to improved treatment of hematopoietic disorders.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2352-2352
Author(s):  
Ruben Hoya-Arias ◽  
Ling-Bo Shen ◽  
Ullas Mony ◽  
Elizabeth Peguero ◽  
Benet Pera ◽  
...  

Abstract Abstract 2352 Blood is one of the most highly regenerative tissues and is generated by a rare population of hematopoietic stem cells (HSCs). Generation of HSCs seems to be restricted during early embryonic development to particular fetal tissues, such as yolk sac, aorta gonad mesonephros (AGM) region and placenta. The competence of HSCs to self-renew and differentiate towards multiple lineages sustains the generation of blood cells for an entire lifespan. Human HSCs are defined by their self-renewal and multilineage differentiation in functional xenotransplantation assays. Differentiation of human embryonic stem cells (hESCs) into transplantable HSCs continues to be a major challenge in the field. We believe that generation of functional multipotent HSCs from hESC requires an approach that recapitulates early human development both temporally and spatially. Thus, we have developed an innovative in vitro system that mimics the early events of human embryonic development prior to hematopoietic generation; where 1) induction of a single ESC monolayers to mesodermal fate; 2) single cell mesoderm progenitors are placed into a fetal AGM stromal environment under conditions that favor endothelial differentiation; and 3) hematopoietic inductors are added generating single free-floating hematopoietic progenitors in a temporal fashion. We have identified both hemogenic endothelium (adherent) and hematopoietic (non-adherent) cells in the AGM/mesoderm coculture. Hemogenic endothelium defined by expression of VE-Cadherin cocultured on AFT024 fetal liver stromal cells is able to generate hematopoietic precursors. Non-adherent cells start budding up by day 18 in the AGM/mesoderm coculture. Flow cytometry analysis shows that CD31 is the prevalent marker in this floating population. Non-adherent VE-Caherin-CD31+CD34+ cells arising between day 24 till 33 have shown the highest hematopoietic activity as measure by colony forming units (CFU) cells, cobblestone area-forming cell assays (week 4–5 CAFC) and their potential to generate both B and T-cells. Molecular characterization of hemogenic endothelium the non-adherent VE-Caherin-CD31+CD34+ cells by qRT-PCR confirm that also a temporal gene expression is taken place as cells transit from endothelium to hematopoietic cells. VE-Caherin-CD31+CD34+ cells also express CRCX4, suggesting their potential for engrafting in mice models. This novel approach provides a unique opportunity to study the early events and to identify molecular targets in the human hematopoietic commitment, lineage specification, and maturation; these hematopoietic cells may represent a promise for clinical therapies in a diverse range of blood diseases. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1999 ◽  
Vol 93 (4) ◽  
pp. 1168-1177 ◽  
Author(s):  
Minetaro Ogawa ◽  
Masami Kizumoto ◽  
Satomi Nishikawa ◽  
Tetsuhiro Fujimoto ◽  
Hiroaki Kodama ◽  
...  

Abstract Embryonic stem cells can differentiate in vitro into hematopoietic cells through two intermediate stages; the first being FLK1+ E-cadherin− proximal lateral mesoderm and the second being CD45− VE-cadherin+endothelial cells. To further dissect the CD45−VE-cadherin+ cells, we have examined distribution of 4-integrin on this cell population, because 4-integrin is the molecule expressed on hematopoietic stem cells. During culture of FLK1+ E-cadherin− cells, CD45− VE-cadherin+4-integrin− cells differentiate first, followed by 4-integrin+ cells appearing in both CD45− VE-cadherin+ and CD45−VE-cadherin− cell populations. In the CD45−VE-cadherin+ cell population, 4-integrin+ subset but not 4-integrin− subset had the potential to differentiate to hematopoietic lineage cells, whereas endothelial cell progenitors were present in both subsets. The CD45−VE-cadherin− 4-integrin+ cells also showed hematopoietic potential. Reverse transcription-polymerase chain reaction analyses showed that differential expression of the Gata2 and Myb genes correlated with the potential of the 4-integrin+ cells to give rise to hematopoietic cell differentiation. Hematopoietic CD45−VE-cadherin+ 4-integrin+ cells were also present in the yolk sac and embryonic body proper of 9.5 day postcoitum mouse embryos. Our results suggest that the expression of 4-integrin is a marker of the earliest precursor of hematopoietic cell lineage that was diverged from endothelial progenitors.


Blood ◽  
2008 ◽  
Vol 111 (1) ◽  
pp. 122-131 ◽  
Author(s):  
Petter S. Woll ◽  
Julie K. Morris ◽  
Matt S. Painschab ◽  
Rebecca K. Marcus ◽  
Aimee D. Kohn ◽  
...  

Human embryonic stem cells (hESCs) provide an important means to effectively study soluble and cell-bound mediators that regulate development of early blood and endothelial cells in a human model system. Here, several complementary methods are used to demonstrate canonical Wnt signaling is important for development of hESC-derived cells with both hematopoietic and endothelial potential. Analyses using both standard flow cy-tometry, as well the more detailed high-throughput image scanning flow cytometry, characterizes sequential development of distinct early developing CD34brightCD31+Flk1+ cells and a later population of CD34dimCD45+ cells. While the CD34brightCD31+Flk1+ have a more complex morphology and can develop into both endothelial cells and hematopoietic cells, the CD34dimCD45+ cells have a simpler morphology and give rise to only hematopoietic cells. Treatment with dickkopf1 to inhibit Wnt signaling results in a dramatic decrease in development of cells with hematoendothelial potential. In addition, activation of the canonical Wnt signaling pathway in hESCs by coculture with stromal cells that express Wnt1, but not use of noncanonical Wnt5-expressing stromal cells, results in an accelerated differentiation and higher percentage of CD34brightCD31+Flk1+ cells at earlier stages of differentiation. These studies effectively demonstrate the importance of canonical Wnt signaling to mediate development of early hematoendothelial progenitors during human development.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2789-2789
Author(s):  
Igor I. Slukvin ◽  
Maxim A. Vodyanik ◽  
Jack A. Bork ◽  
James A. Thomson

Abstract hES cells provide an unique opportunity to study the earliest stages of hematopoietic commitment which are not easily accessible in the human embryo. To model early hematopoietic development, we cultured H1 and H9 hES cell lines on OP9 stromal cells without the addition of cytokines. On day 2 of co-culture, hES cells up-regulated brachyury expression and began to form mesodermal-like colonies. A transient population of blast colony-forming cells (CFCs) with the potential to differentiate into blood and endothelial cells was detected on days 3–6 of co-culture. CD34+ cells first appeared on day 3–4 of co-culture, which was coincident with induction of the transcription factors GATA-1, GATA-2 and SCL. CD43+ and CD41a+ cells along with CFCs emerged 2 days later within CD34+ population; 3–4 days before the appearance of CD45+ cells. We were able to obtain up to 20% of CD34+ cells from hES/OP9 co-culture and isolate up to 107 CD34+ cells with more than 95% purity from a similar number of initially plated hES cells after 8–9 days of culture. The hES cell-derived CD34+ cells were highly enriched in CFCs, displayed CD90+CD117+CD164+CD38- phenotype of primitive hematopoietic progenitors, and contained ALDHhigh cells as well cells with verapamil-sensitive ability to efflux rhodamine 123. Isolated CD34+ cells differentiated into lymphoid (NK cells) as well as myeloid (neutrophils and macrophages) lineages when cultured on MS-5 stromal cells in the presence of SCF, Flt3-L, IL7 and IL3. These data indicate that hES cell/OP9 co-culture reproduces the major events that are observed during embryonal hematopoietic development, including the formation of lympho-myeloid progenitors. We employed OP9 system for identification of the phenotype of early hematopoietic progenitors in humans and to directly differentiate hES cells into different blood lineages.


Blood ◽  
1999 ◽  
Vol 93 (4) ◽  
pp. 1168-1177 ◽  
Author(s):  
Minetaro Ogawa ◽  
Masami Kizumoto ◽  
Satomi Nishikawa ◽  
Tetsuhiro Fujimoto ◽  
Hiroaki Kodama ◽  
...  

Embryonic stem cells can differentiate in vitro into hematopoietic cells through two intermediate stages; the first being FLK1+ E-cadherin− proximal lateral mesoderm and the second being CD45− VE-cadherin+endothelial cells. To further dissect the CD45−VE-cadherin+ cells, we have examined distribution of 4-integrin on this cell population, because 4-integrin is the molecule expressed on hematopoietic stem cells. During culture of FLK1+ E-cadherin− cells, CD45− VE-cadherin+4-integrin− cells differentiate first, followed by 4-integrin+ cells appearing in both CD45− VE-cadherin+ and CD45−VE-cadherin− cell populations. In the CD45−VE-cadherin+ cell population, 4-integrin+ subset but not 4-integrin− subset had the potential to differentiate to hematopoietic lineage cells, whereas endothelial cell progenitors were present in both subsets. The CD45−VE-cadherin− 4-integrin+ cells also showed hematopoietic potential. Reverse transcription-polymerase chain reaction analyses showed that differential expression of the Gata2 and Myb genes correlated with the potential of the 4-integrin+ cells to give rise to hematopoietic cell differentiation. Hematopoietic CD45−VE-cadherin+ 4-integrin+ cells were also present in the yolk sac and embryonic body proper of 9.5 day postcoitum mouse embryos. Our results suggest that the expression of 4-integrin is a marker of the earliest precursor of hematopoietic cell lineage that was diverged from endothelial progenitors.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4308-4308
Author(s):  
Nico Lachmann ◽  
Mania Ackermann ◽  
Eileen Frenzel ◽  
Christine Happle ◽  
Olga Klimenkova ◽  
...  

Abstract Hematopoietic in-vitro-differentiation of pluripotent stem cells (PSCs) such as embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs) holds great promise for disease modeling, drug testing, as well as cell- and gene-therapy strategies. Although hematopoietic differentiation of PSC has been shown to be feasible, the majority of current protocols apply a large number of different cytokines to direct differentiation. In this line, priming the differentiation process by a multitude of cytokines may alter the endogenous hematopoietic differentiation program of PSCs, thus hampering the usefulness of such protocols to gain insight into physiologic human hematopoietic development. To overcome this problem we have investigated the hematopoietic differentiation potential of human PSC, based on minimal cytokine application. Given the emerging role of IL3 as a critical factor in adult hematopoiesis and the pivotal role of M-CSF and G-CSF for terminal myeloid differentiation, we here employed IL3 in combination with either M-CSF or G-CSF on hematopoietic development. To prove our concept, human CD34+ cell-derived iPSC clones were subjected to an embryoid body (EB)-based myeloid differentiation protocol employing cytokines from day 5 onwards and yielding so-called “myeloid cell forming complexes” (MCFCs) within 7-10 days. Analysis of MCFC within 10 days of differentiation revealed expression of MIXL1, KDR1, GATA2, and RUNX1, as well as an early CD34+/CD45- population undergoing transition to a CD34+/CD45+ and thereafter CD34-/CD45+ phenotype. The hypothesis of a primitive hematopoietic cell arising from a population with dual (hematopoietic and vascular epithelial) potential was supported by co-staining of these populations with VE-cadherin (CD144). Here primarily the CD34+/CD45+/CD144- cells were capable of colony formation in vitro. Differentiation of PSC for more than 15-days resulted in the continuous shedding of hematopoietic cells from MCFCs and further differentiation along the IL3/M-CSF let to the generation of >99% pure monocytes/ macrophages (iPSC-MΦ), while IL3/G-CSF promoted granulopoiesis (iPSC-gra, purity >95%). Of note, hardly any CD34+ cells were detected among MCFC-shedded cells for the IL3/M-CSF as well as the IL3/G-CSF combination. In contrast, differentiation in IL3 only resulted in 10% MCFC-derived CD34+ cells, an observation further confirmed by a 10-times increased clonogenicity for cells shedded from MCFC exposed to IL3 only when compared to IL3/G-CSF or IL3/M-CSF cultures. Furthermore, cells cultured in IL3 maintained the capacity of subsequent M-CSF-driven terminal differentiation, whereas no suspension cells were observed following differentiation of PSC with G-CSF alone. Most strikingly, IL3/M-CSF or IL3/G-CSF cultures generated iPSC-MΦ or iPSC-gra from day 14-15 onwards over a period of 3-5 months at a quantity of 0.4-2.0 x 106 cells/week (cumulative 0.8-4.0 x 107 cells) per 3.5 cm well. For IL3/M-CSF cultures detailed characterization of mature myeloid cells demonstrated a typical MΦ-morphology of iPSC-MΦ by cytospins and a surface-marker profile of CD45, CD11b, CD14, CD163, and CD68. In addition, iPSC-MΦ had the ability to phagocytose latex-coated beads similar to peripheral blood (PB)-MΦ polarized to M2 and upon LPS stimulation secreted MCP1, IL6, IL8, and IL10, whereas IFNy, IL1b, IL4, IL5, and IL12 were absent. iPSC-gra showed surface expression of CD45, CD11b, CD16, CD15, CD66b and a differential count containing pro-myelocyte (3%), myelocyte (5%), meta-myelocyte (30%), bands (22%), eosinophils (2%), basophils (1%), and segmented-neutrophils (37%) . Moreover, iPSC-gra were able to migrate towards an IL8 or fMLP gradient, formed neutrophil extracellular traps, and up-regulated NADPH activity and ROS production upon PMA stimulation to a similar degree as PB granulocytes. In summary, we here present an in vitro differentiation protocols for human iPSC requiring minimal cytokine stimulation, which appears highly suited to model human hematopoietic development or generate cells for gene and cell-replacement strategies. We further provide evidence that IL3 constitutes a key cytokine driving the early hematopoietic specification of human PSC, whereas M-CSF and G-CSF function primarily as downstream “supporter” cytokines regulating the terminal differentiation towards macrophages and granulocytes, respectively. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 69-69
Author(s):  
Xinghui Tian ◽  
Melinda K. Hexum ◽  
Vesselin R. Penchev ◽  
Russell J. Taylor ◽  
Leonard Shultz ◽  
...  

Abstract Selection for CD34+ cells from human bone marrow, umbilical cord blood and mobilized peripheral blood cells has been used to enrich for putative hematopoietic stem cells that are typically characterized by the ability to provide long-term multi-lineage engraftment after transplantation into NOD/SCID mice. In vitro studies have shown that CD34+ cells derived from human embryonic stem cells (hESC) have the similar capacity to develop into diverse mature blood cell lineages under defined conditions, though they may represent an even more primitive progenitor population with bi-potent ability to developed into both hematopoietic and endothelial cells. Previously, we and others have demonstrated in vivo hematopoietic engraftment of an unsorted heterogeneous population of differentiated hESC-derived cells injected into NOD/SCID mice. Here, to determine whether sorted CD34+ cells isolated from differentiated hESC have the equivalent SCID-repopulating potential, we directly delivered the CD34+ cells into the liver of neonatal NOD/SCID/gamma c (null) mice. Stable expression of firefly luciferase (luc) in the hESC derived-CD34+ cells allowed us to non-invasively track the engraftment patterns by in vivo bioluminescent imaging. Luc+ CD34+ cells were produced by co-culture of hESC with M2-10B4 mouse bone marrow-derived stromal cells. Each mouse (n=18) was injected with 2.5 – 9.4 × 10^5 CD34+ cells isolated at an early stage of differentiation (day 6–9 of co-culture). Successful injections were routinely confirmed by the bioluminescent signal detected in the liver at 1 week post-transplantation. There was typically a decrease of luc signal observed between 2–5 weeks post-transplantation, followed by the stabilizing and increasing of luc-expressing cells at later time points. Dramatic increase of bioluminescent signal in the livers was routinely demonstrated 8–12 weeks post-transplantation. The bioluminescence foci were initially maintained in the liver. Migration of luc+ cells from liver to bone marrow, spleen, kidney and mediastinum/thymus was also detected at later time points (10+ weeks post-transplantation). Flow cytometric analysis demonstrated a significant population of human HLA-class I+ cells (0.1–5.35%) in the engrafted livers (11 out of 15 mice), with the majority of the human CD34+CD31+ endothelial cells (0.03–4.78%). In most of cases, these endothelial cells also co-expressed CD73. No human CD45+ cells were found in any of the tissues evaluated. This is in contrast to CD34+ cells obtained from human cord blood utilized as a positive control cell populations that routinely demonstrated high levels of CD45+ cells (86.92±10.5%) of diverse hematopoietic lineages in the liver when transplanted in this model. Immunohistochemical staining showed that the hESC-derived CD34+CD31+ cells localized in vessels of the engrafted liver. Remarkably, the hESC-derived luc+CD34+CD31+ cells could be isolated several months post-transplant and re-cultured to clearly demonstrate typical endothelial cell characteristics including expression of vonWillebrand factor and VE-Cadherin, taking up of ac-LDL, and formation of capillary-like tubes when cultured in Matrigel. Additionally, ex vivo imaging of the exercised spleens and their surrounding connective tissues demonstrated luc+ cells located in the surrounding connective/mesenchymal tissues. Similar luc+ mesenchymal tissues were also found around the kidneys. Importantly, no visible teratomas were found in any of the transplanted mice. Together, these results demonstrate hESC-derived CD34+ cells preferentially develop into endothelial cells after transplantation into the fetal liver environment, with some additional development of mesenchymal tissues, but minimal hematopoietic cells. These studies demonstrate hESC-derived CD34+ cells are suitable for in vivo models to correct of vascular disease and further studies are underway to define signals required for improved hematopoietic engraftment of purified hESC-derived CD34+ cells.


Biomolecules ◽  
2020 ◽  
Vol 10 (12) ◽  
pp. 1614
Author(s):  
Paulina Podkalicka ◽  
Jacek Stępniewski ◽  
Olga Mucha ◽  
Neli Kachamakova-Trojanowska ◽  
Józef Dulak ◽  
...  

Inadequate supply of oxygen (O2) is a hallmark of many diseases, in particular those related to the cardiovascular system. On the other hand, tissue hypoxia is an important factor regulating (normal) embryogenesis and differentiation of stem cells at the early stages of embryonic development. In culture, hypoxic conditions may facilitate the derivation of embryonic stem cells (ESCs) and the generation of induced pluripotent stem cells (iPSCs), which may serve as a valuable tool for disease modeling. Endothelial cells (ECs), multifunctional components of vascular structures, may be obtained from iPSCs and subsequently used in various (hypoxia-related) disease models to investigate vascular dysfunctions. Although iPSC-ECs demonstrated functionality in vitro and in vivo, ongoing studies are conducted to increase the efficiency of differentiation and to establish the most productive protocols for the application of patient-derived cells in clinics. In this review, we highlight recent discoveries on the role of hypoxia in the derivation of ESCs and the generation of iPSCs. We also summarize the existing protocols of hypoxia-driven differentiation of iPSCs toward ECs and discuss their possible applications in disease modeling and treatment of hypoxia-related disorders.


Stem Cells ◽  
2012 ◽  
Vol 30 (4) ◽  
pp. 643-654 ◽  
Author(s):  
Nicole M. Kane ◽  
Lynsey Howard ◽  
Betty Descamps ◽  
Marco Meloni ◽  
John McClure ◽  
...  

Haematologica ◽  
2021 ◽  
Author(s):  
Vikas Madan ◽  
Zeya Cao ◽  
Weoi Woon Teoh ◽  
Pushkar Dakle ◽  
Lin Han ◽  
...  

Recurrent loss-of-function mutations of spliceosome gene, ZRSR2, occur in myelodysplastic syndromes (MDS). Mutation/loss of ZRSR2 in human myeloid cells primarily causes impaired splicing of the U12-type introns. To investigate further the role of this splice factor in splicing and hematopoietic development, we generated mice lacking ZRSR2. Unexpectedly, Zrsr2-deficient mice developed normal hematopoiesis with no abnormalities in myeloid differentiation evident in either young or ≥1-year old knockout mice. Repopulation ability of Zrsr2-deficient hematopoietic stem cells was also unaffected in both competitive and non-competitive reconstitution assays. Myeloid progenitors lacking ZRSR2 exhibited mis-splicing of U12-type introns, however, this phenotype was moderate compared to the ZRSR2- deficient human cells. Our investigations revealed that a closely related homolog, Zrsr1, expressed in the murine hematopoietic cells, but not human, contributes to splicing of U12-type introns. Depletion of Zrsr1 in Zrsr2 KO myeloid cells exacerbated retention of the U12-type introns, thus highlighting a collective role of ZRSR1 and ZRSR2 in murine U12-spliceosome. We also demonstrate that aberrant retention of U12-type introns of MAPK9 and MAPK14 leads to their reduced protein expression. Overall, our findings highlight that both ZRSR1 and ZRSR2 are functional components of the murine U12-spliceosome, and depletion of both proteins is required to model accurately ZRSR2-mutant MDS in mice.


Sign in / Sign up

Export Citation Format

Share Document