scholarly journals In Situ Microscopy Studies of Infused Megakaryocytes: Implications in Thrombopoiesis

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 4287-4287
Author(s):  
Hyunjun Kim ◽  
Danuta Jadwiga Jarocha ◽  
Ian Johnston ◽  
Hyunsook Ahn ◽  
Deborah L French ◽  
...  

Abstract The questions of whether thrombopoiesis - the release of platelets from megakaryocytes - occurs both as megakaryocytes emerge from the intramedullar space or occurs as well in the pulmonary vascular bed remains unanswered. Studies by Lefrançais E, et al, (Nature, 2017) demonstrated by in situ microcopy that perhaps 50% of all platelet release in mice occurs from megakaryocytes released from the marrow and traveled to the lungs where they undergo thrombopoiesis over a 20- to 60-minute time-period. We examined whether CD34+-derived human megakaryocytes infused into immunocompromized NSG mice would also shed platelets in the lungs in a similar fashion. We differentiated CD34+-derived hematopoietic stem-progenitors for 12 days in culture using conditions previously described (Wang Y, et al., Blood 2015). We found that unlike platelet-like-particle (PLP) formation in in vitro cultures of CD34+ hematopoietic progenitor cell (HPC)-derived (CD34+) megakaryocytes, which undergo asynchronous shedding of the PLPs, that over 95% of infused CD34+ megakaryocytes shed their platelets within the first 40 minutes much as has been observed for endogenous murine megakaryocytes. The average number of cytoplasmic extensions per megakaryocytes was ~2.7, again very similar to what was seen with endogenous murine megakaryocytes. In contrast, CD34+ cells grown in culture into megakaryocytes for a shorter period of time of only 7 days, poorly shed any cytoplasmic fragments. We also studied human megakaryocytes grown from immortalized megakaryocyte progenitor cell lines (imMKCLs) from induced pluripotent stem cells (iPSCs) generated by the Eto laboratory and kindly provided by Dr. Koji Eto, Kyoto University). These cells were grown and differentiated into terminal megakaryocytes as described (Nakamura S, Cell Stem Cell, 2014) for 4 days in culture. These cells have been proposed to be useful for large-scale preparation of PLPs in vitro for clinical use in place of donor-derived platelets. The resultant infused human imMKCL-derived megakaryocytes also synchronously shed platelets, but only 50% of the infused cells shed their cytoplasm in contrast to >95% of CD34+ megakaryocytes. Moreover, cytoplasmic extensions were decreased to an average of ~1.1 per megakaryocyte. We had proposed that in vitro-generated megakaryocytes might be directly infused into patients in place of further manipulating the megakaryocytes to release functional platelets in vitro using a bioreactor. However, such megakaryocytes will likely be contaminated with a higher level of HPCs than anticipated from in vitro-prepared platelets, and concern exists that they may lead to unacceptable graft versus host complications. We, therefore, examined whether irradiating megakaryocytes as one strategy to eliminate this concern results in megakaryocytes that are still functional and found that megakaryocytes irradiated with up to 25 Gy retain platelet yield per infused megakaryocytes with the platelets having the same half-life. If irradiated and kept in culture, these megakaryocytes begin to shed platelets and undergo apoptosis notably by 24 hours. We also examined whether the pulmonary bed differs from other vascular beds, and infused CD34+ megakaryocytes both intravenously and intra-arterially in parallel studies and found that following intra-arterial infusion, megakaryocytes were mostly entrapped in various organs, but shed few platelets. Thus, our studies suggest that the pulmonary bed is unique for platelet shedding from entrapped megakaryocytes. Whether this is due to the structural organization of the pulmonary beds, its endothelial lining, its reverse exchange in oxygen, carbon dioxide and pH from other capillary beds or the mechanical forces of inhalation and exhalation that expand and contract the capillary cross-sectional area needs to be examined. Our studies show that infused human megakaryocytes synchronously release platelets over a 40-minute window and can do so even after being irradiated and that this occurs specifically in the lungs not only has potential clinical application, but also raises biological questions about what determines thrombopoiesis-readiness and what are the features of the pulmonary bed that allows this synchronous release. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2009 ◽  
Vol 114 (6) ◽  
pp. 1186-1195 ◽  
Author(s):  
Hyung Chan Suh ◽  
Ming Ji ◽  
John Gooya ◽  
Michael Lee ◽  
Kimberly D. Klarmann ◽  
...  

Abstract Development of hematopoietic stem cells (HSCs) and their immediate progeny is maintained by the interaction with cells in the microenvironment. We found that hematopoiesis was dysregulated in Id1−/− mice. Although the frequency of HSCs in Id1−/− bone marrow was increased, their total numbers remained unchanged as the result of decreased bone marrow cellularity. In addition, the ability of Id1−/− HSCs to self-renew was normal, suggesting Id1 does not affect HSC function. Id1−/− progenitors showed increased cycling in vivo but not in vitro, suggesting cell nonautonomous mechanisms for the increased cycling. Id1−/− HSCs developed normally when transplanted into Id1+/+ mice, whereas the development of Id1+/+ HSCs was impaired in Id1−/− recipients undergoing transplantation and reproduced the hematologic features of Id1−/− mice, indicating that the Id1−/− microenvironment cannot support normal hematopoietic development. Id1−/− stromal cells showed altered production of cytokines in vitro, and cytokine levels were deregulated in vivo, which could account for the Id1−/− hematopoietic phenotypes. Thus, Id1 is required for regulating the hematopoietic progenitor cell niche but is dispensable for maintaining HSCs.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1282-1282
Author(s):  
Yi Zeng ◽  
Karl Staser ◽  
Keshav Mohan Menon ◽  
Su-jung Park ◽  
Muithi Mwanthi ◽  
...  

Abstract Abstract 1282 Ezrin is a member of the ERM (ezrin, moesin and radixin) protein family that links plasma membrane proteins to the actin cytoskeleton. Ezrin in other in vitro cell systems has been hypothesized to participate in cell-cell contact and could have a role in stem/ progenitor cell mobilization and adhesion. To test this hypothesis, we crossed ezrinflox/flox mice with Mx1 cre transgenic mice to generate an inducible ezrin knock out mouse model. Inducible disruption of the ezrin gene in hematopoietic cells was achieved by the administration of polyIC. Ezrin knock out HSPCs exhibited a 30–40% decrease in baseline and chemokine stromal cell-derived factor-1 (SDF-1) stimulated motility in transwell migration assays in vitro. In addition, loss of ezrin led to a 60% decrease in the homing capacity of HSPCs in lethally irradiated recipient mice following transplantation. There was a 40–55% decrease in colony forming cells in peripheral blood and spleen of the mice following ezrin knock out, suggesting that ezrin knock out HSPCs may be deficient in egressing out of the bone marrow. To further understand the cause of the impaired motility of ezrin knock out HSPCs, we examined F-actin level of HSPCs at baseline and in response to SDF-1. Ezrin knock out HSPCs displayed 1.5 to 2 fold higher level of F-actin at baseline when compared with wild type cells. Following stimulation with SDF-1, wild type HSPCs that migrated to the bottom compartment of the transwell demonstrated a 2 time greater decrease in F-actin level when compared with ezrin knock out cells, suggesting that ezrin may participate in the regulation of F-actin depolymerization in HSPCs. In summary, we demonstrate that ezrin modulates HSPC migration and homing likely through its regulation on F-actin organization. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2370-2370
Author(s):  
Marie-Lorraine Chretien ◽  
Romain Aucagne ◽  
Nathalie Droin ◽  
Arlette Hammann ◽  
Eric Solary ◽  
...  

Abstract Abstract 2370 TIF1gamma (or TRIM33) is an ubiquitous nuclear protein that belongs to the transcriptional intermediary factor 1 family. Human and mouse TIF1gamma are closely related to zebrafish moonshine (mon), a gene whose mutations disrupt embryonic and adult hematopoiesis with severe red blood cell aplasia. Targeted deletion of Tif1gamma is embryonic lethal in mice. In zebrafish and human CD34+ cells, TIF1gamma functionally links positive elongation factors such as p-TEFb and FACT to blood specific transcription complexes (e.g. the SCL/TAL1 complex) to regulate elongation of genes by antagonizing Pol II pausing. TIF1gamma also affects the human hematopoietic progenitor cell response to the cytokines of the transforming growth factor-beta superfamily through various mechanisms. Recently, we showed that the loss of Tif1gamma in mouse hematopoietic stem cells (cFES-Cre-Tif1gamma) favors the expansion of the granulo-monocytic progenitor compartment. The gene deletion induces the age-dependent appearance of a cell-autonomous myeloproliferative disorder with myelodysplastic features, monocytosis, and hepatosplenomegaly that recapitulates essential features of human chronic myelomonocytic leukemia (CMML). Interestingly, TIF1gamma is almost undetectable in leukemic cells of 35% of patients with CMML. This down-regulation is related to the hyper-methylation of CpG sequences in the gene promoter. Our results demonstrated that TIF1gamma is an epigenetically regulated tumor suppressor gene in hematopoietic cells. In addition, an altered production of peritoneal macrophages was observed in our mouse model. These macrophages did not adhere to the plastic and were morphologically abnormal in vitro. In bone marrow and in Lin- progenitor cells, Tif1gamma deletion leads to a significant decrease of cfms (Csf-1r) expression, required for the differentiation, proliferation, and survival of monocytic phagocytes. We also identified in CMML patients the association between low levels of TIF1gamma and cFMS (Aucagne et al., J. Clin. Invest., 121, 2361–2370, 2011). To gain insight into the possible mechanism accounting for diminished accumulation of macrophages, we examined the expression of c-Fms. We show that level of its expression is reduced significantly in blood monocytes isolated from Tif1gamma-deleted mice. When Tif1gamma-deleted sorted myeloid cells were induced to differentiate into macrophages in presence of CSF-1, a delayed production of few abnormal large macrophages was observed. Apoptosis was associated with this alteration of differentiation. This phenomenon was also characterized in young mice not developing the disease yet. The morphological abnormalities were correlated with very important alterations of specific macrophage differentiation markers. Expression of specific transcription factors involved in macrophage differentiation was deeply deregulated. Moreover, macrophage function such as migration, cytokine or chemokine secretion in response to LPS was altered. Likewise, in vitro differentiation of monocytes into dendritic cells was also abnormal. Altogether, our results suggest that monocyte plasticity is at least partially orchestrated by Tif1gamma. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2357-2357
Author(s):  
Hong Wang ◽  
Adam M Sorkin ◽  
Ramasamy Sakthivel

Abstract Abstract 2357 Infection by Plasmodium Vivax (P. Vivax) is the most common cause of Sleeping Malaria. P. Vivax and other plasmodia have grown increasingly resistant to antimalarial drugs. Introduced by mosquito bite, P. vivax sporozoites enter circulation and preferentially penetrate reticulocytes by attaching to the Fya and Fyb Duffy antigen/chemokine receptor (DARC) via PvRBP-1 and PvRBP-2 proteins located at their apical poles. Once in a reticulocyte, the parasite begins to reproduce asexually, releasing of thousands of merozoites into circulation. At this point, merozoites can also enter the liver and triggering relapses months or years later. The emergence of drug-resistant strains of p. vivax has stimulated development of new vaccines and treatments, but progress has been slowed by the dearth of reliable screening platforms. Many vaccine candidates have been developed to act upon vivax merozoites by preventing binding of PvRBP-1 and 2 to DARC, thereby arresting reproduction. However, there is a distinct lack of in vitro models to evaluate candidates that employ this mechanism. We are addressing this issue with a novel ex vivo expansion and differentiation technology for large-scale production of DARC expressing reticulocytes for in vitro P. vivax infection studies. This technology comprises an expansion system that can produce high yields of hematopoietic precursors (CD133+/CD34+ cells) from a variety of sources (marrow, peripheral blood, and cord blood), and a differentiation system to produce a relatively pure population of enucleated erythrocytes. In this study, we have refined the polyethersulfone (PES) nanofiber-based culturing system containing growth factors and cytokines in a serum-free media, to expand hematopoietic stem and progenitor cells (HSPC) ex vivo. This expansion technology allows rapid 200-fold ex vivo proliferation within 7 days of umbilical cord blood derived CD133+/CD34+ HSPCs from a DARC+ donor. Following expansion, over 50% of these cells retained HSPC phenotype (expression of CD34+). We have subsequently demonstrated that feeder layer free three-step differentiation of nanofiber-expanded cells using cytokines results in a population containing predominately enucleated reticulocyte-like cells. At 21 days of differentiation, cells had expanded 50-fold. Around 41% of cells were enucleated reticulocytes. These cells expressed glycophorin-A, a major sialoglycoprotein present on the human erythrocyte membrane. ∼28% of cells were CD36+, and ∼70% were CD71+ indicating an erythroid lineage. These results suggest that this technology can produce a population of DARC+ reticulocytes that is ∼5,000-fold greater than the starting population of HSPCs. We are partnering with leading malaria vaccine researchers to demonstrate that these reticulocytes can be parasitized by p. vivax. We believe that this will provide a unique platform to jumpstart research of malaria parasites and enable rapid development of effective vaccines. Further development of this technology may also have significant implications for large-scale ex vivo production of erythrocytes for general use. Reticulocyte-like cells and expelled nuclei during differentiation of nanofiber-expanded HSPC. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. SCI-39-SCI-39
Author(s):  
Luc Douay

Abstract Abstract SCI-39 The generation of red blood cells (RBCs) in vitro using biotechnologies could represent an interesting alternative to classical transfusion products, in that it would combine adequate supplies with the specific production of blood products of a particular phenotype and the reduction of infection risks. This presentation will review how it is now possible to obtain in vitro complete maturation of the erythroid line to the stage of enucleation, starting from hematopoietic stem cells (HSCs) from peripheral blood, bone marrow or umbilical cord blood, or embryonic stem cells or adult pluripotent stem cells (induced pluripotent stem cells, iPSCs). This presentation will discuss how the functionality of cultured human RBCs (cRBCs) is settled in terms of deformability, hemoglobin maturation, oxygen carrying capacity, enzyme content, and terminal maturation from the reticulocyte stage to mature RBC after infusion into the NOD/SCID mouse model. The clinical feasibility of this concept has recently been demonstrated by reporting that cRBCs generated in vitro from peripheral HSCs under GMP conditions encounter in vivo the conditions required for their maturation and that they persist in the circulation for several weeks in humans. These data have established the proof of principle for transfusion of in vitro-generated RBCs and the pathway toward new developments in transfusion medicine. The most proliferative source of stem cells for generating cRBCs is cord blood, but it is limited in terms of HSCs and is dependent on donations. Pluripotent stem cell technology represents a potentially unlimited source of RBCs and opens the door to the development of a new generation of allogeneic transfusion products. Because iPSCs can be selected for a phenotype of interest, they are obviously the best candidate for organizing complementary sources of RBCs for transfusion. It is established that only three human iPSC clones would have been sufficient to match more than 99 percent of the patients in need of RBC transfusions. As a whole, a very limited number of RBC clones would provide for the needs of most alloimmunized patients and those with a rare blood group. Generating cRBCs from iPSCs has been done but needs to be optimized to lead to a clinical application in blood transfusion. Several crucial points remain to be resolved, notably, the choice of the initial cell type, the method of reprogramming (i.e., to ensure the safety of the iPSCs and to ensure their clinical grade), the optimization of the erythrocyte differentiation, and the definition of GMP conditions for industrial production. Assuming that in vitro large-scale cultured RBC production efficiently operates in the near future, this presentation will highlight the potential applications for alloimmunized patients and those with a rare blood group. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 296-296 ◽  
Author(s):  
Carine Domenech ◽  
Alix Rousseau ◽  
Laurence Petit ◽  
Sandra Sanfilippo ◽  
Jean Soulier ◽  
...  

Abstract Fanconi anemia (FA) is a genetic disorder due to mutations in one of the sixteen FANC genes involved in DNA repair. Many FA patients develop bone marrow failure (BMF) during childhood, and FA strongly predisposes to myelodysplasia syndrome and/or acute myeloid leukaemia. The pathogenesis of the BMF remains uncompletely understood. Low hematopoietic progenitor cell (HPCs) counts observed early in life and preceeding the onset of blood cytopenia in patients led we, and other, to hypothesize that the hematopoietic development might be abnormal in the FA embryo. Indeed, unlike adult hematopoietic stem cells (HSCs) which are quiescent in the BM niche, during embryonic life HSCs are in active proliferation in sites of expansion such as fetal liver and placenta, where they get amplified and acquire properties of adult HSC .We hypothesized that in FA, the FA defect in response to the replicative stress could impair the expension of the HSC pool.In order to investigate this hypothesis, we carried out studies in Fancg-/- knock out mice and in human FA fetuses obtained with informed consent from medical abortion. In Fancg-/- mice, FACS analysis revealed a 1,5- to 3-fold deficiency in hematopoietic stem and progenitor cells (HSPC) very early during embryonic development (i.e 11.5 days of gestation - E11.5) in fetal liver (FL) and placenta (Pl) (p <0.001). In both organs, this defect persists during the whole period of amplification (until E14.5 for FL and E12.5 for Pl). In vitro clonogenic assays also demonstrated a 2- fold defect in granulocyte, erythrocyte and macrophage (GEM) progenitors both in Fancg-/- FL or Pl compared to WT (p <0.001), and 4 to 5- fold defect in more immature mixed GEM progenitors in FL (p <0.001). LTC-IC frequency of the HSC-enriched Lineage- Sca1+ AA4.1+ population (LSA) of E14.5 Fancg-/- FL comforted this later result, since it was 5-fold lower than for WT. In vivo long-term hematopoietic reconstitution (LTR) assays confirmed a deficit of the HSC enriched LSA population of E14.5 Fancg-/- FL. Indeed, although the percentage of mice reconstituted was as good as that obtained with the same number of WT LSA, the CD45 Ly5.2 chimerism was reduced (49±20% vs 84±4% for 1000 LSA injected, and 56±12% vs 87±2% for 5000 LSA). Interestingly, bone marrow analysis of mice reconstituted with Fancg-/- LSA 22 weeks after injection showed a level of CD45 Ly5.2 chimerism 3-fold lower than that found in blood, spleen and thymus, as well as a very low chimerism for myeloid GEM lineages, contrasting with a high chimerism for B and T lymphoid lineages. Moreover, we were able to demonstrate that this deficit is already present at E12.5, both in Fancg-/- FL and Pl. Indeed, no mice reconstituted with 3.105 total Fancg-/- fetal liver cells, while 100% injected with the same number of WT FL cells got reconstituted with a chimerism of 59,5±5%. For Pl, when 500 000 cells were injected, reconstitution was observed in only 1 out of 3 mice for Fancg-/- (29% chimerism), and in 3 out of 3 mice for WT (88±4% chimerism). In human FA FL of 14 weeks of gestation, we also observed a 4-fold defect of HSPC with a total lack of in vitro amplification compared to control, in agreement with the mice data. Taken together, these data demonstrate that a profound deficit of HSCs and progenitors cells is present since the earlier stages of embryonic development in FA. In addition, using organotypic cultures of E11 aortas, we could show that this defect of amplification is already present in HSCs emerging from Fancg-/- aorta, which showed a 2-fold lower rate of amplification compared to WT. More importantly, our results show for the first time exhaustion in myeloid lineage of FA, in agreement with what is observed in children with FA disease. Altogether, our work suggests a role of the FA pathway during the development of the hematopoietic system leading to a deficit of amplification of HSC. Comparison of FA HSC transcriptome with that of control HSC in FL and Pl is in progress. It should allow to identify the key pathways involved in the embryonic HSC amplification that are deregulated in FA, and hopefully getting more insights in the pathogenesis of the BMF and leukemogenesis in FA patients. Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Vol 9 (1) ◽  
Author(s):  
Stefan Radtke ◽  
André Görgens ◽  
Symone Vitoriano da Conceição Castro ◽  
Lambros Kordelas ◽  
Angela Köninger ◽  
...  

Abstract Endothelial and mesenchymal stromal cells (ECs/MSCs) are crucial components of hematopoietic bone marrow stem cell niches. Both cell types appear to be required to support the maintenance and expansion of multipotent hematopoietic cells, i.e. hematopoietic stem cells (HSCs) and multipotent progenitors (MPPs). With the aim to exploit niche cell properties for experimental and potential clinical applications, we analyzed the potential of primary ECs alone and in combination with MSCs to support the ex vivo expansion/maintenance of human hematopoietic stem and progenitor cells (HSPCs). Even though a massive expansion of total CD34+ HSPCs was observed, none of the tested culture conditions supported the expansion or maintenance of multipotent HSPCs. Instead, mainly lympho-myeloid primed progenitors (LMPPs) were expanded. Similarly, following transplantation into immunocompromised mice the percentage of multipotent HSPCs within the engrafted HSPC population was significantly decreased compared to the original graft. Consistent with the in vitro findings, a bias towards lympho-myeloid lineage potentials was observed. In our conditions, neither classical co-cultures of HSPCs with primary ECs or MSCs, even in combination, nor the xenograft environment in immunocompromised mice efficiently support the expansion of multipotent HSPCs. Instead, enhanced expansion and a consistent bias towards lympho-myeloid committed LMPPs were observed.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3450-3450
Author(s):  
Yuhuan Wang ◽  
Vincent M. Hayes ◽  
Lin Lu ◽  
Xiaoji Chen ◽  
Rudy Fuentes ◽  
...  

Abstract Abstract 3450 Our goal is to generate sufficient PLTs from ex vivo-generated MKs for clinical utility in PLT transfusions. A critical step in this process begins with ex vivo-generated hMKs and deriving clinically relevant PLTs. We reported that infused mature, high-ploidy murine (m) MKs derived from fetal liver (FL) cells increased PLT counts in recipient mice in a clinically relevant fashion, thus avoiding the need to generate ex vivo functional PLTs. We examined whether this strategy applies to hMKs derived from FL cells (gestational age, 17–21 weeks) and bone marrow (BM) cells in a xeno-transfusion model using immunodeficient NOD/SCID/IL-2Rγcnull (NSG) mice. Infused hPLTs isolated from blood had a half-life (T1/2) of 10 hours (hrs), compared to 24 hrs for infused murine PLTs. The hPLTs were functional in NSG mice as demonstrated by their incorporation into growing thrombi in situ. Both hFL hematopoietic mononuclear cells and hBM-CD34+ cells were cultured in serum-free media supplemented with optimized cytokine cocktails to generate hMKs. In contrast to the murine studies where the FL cell-derived mMKs were the most efficient source of derived mPLTs, FL cell-derived hMKs had low ploidy (0% ≥ 8N ploidy), gave rise to ∼16 PLTs/infused hMK, and had a short T1/2 (6 hrs). In contrast, 17% of hBM cell-derived MKs had a ploidy of ≥ 8N, and after infusion into NSG mice, resulted in a wave of MKs transiently entrapped in the pulmonary microvasculature and then over ∼0.5–3 hrs released PLTs with a T1/2 of 10 hrs, comparable to infused hPLTs. Maximally, we achieved a level of 5% of circulating total PLTs being derived from human cells with ∼32 PLTs/infused hMK. These hPLTs were normal in size, displayed normal levels of surface markers, were functional, and incorporated into growing thrombi. One strategy to increase hPLT yield is to expose developing MKs to drugs reported to increase MK maturation, thrombopoiesis, and/or facilitate hematopoietic progenitor cell expansion. Such drugs include dimethylfasudil (diMF) (an inhibitor of several kinases involved in polyploidization), UNC0638 (a G9a histone methyltransferase inhibitor), SR1 (an AhR antagonist), and nicotinamide (a sirtuin histone/protein deacetylases inhibitor). Although diMF promoted size and polyploidization of hMKs, diMF markedly worsened yield of PLTs/infused hMK and decreased PLTs T1/2 in vivo. UNC0638 led to significant cell expansion, but lowered hMKs ploidy and PLTs/infused hMK yield. Nicotinamide increased maturation, size and polyploidization of hMKs, but PLT release following MK infusion needs further study. Of note, SR1 that has been reported to promote the expansion of human HSC, not only increased size and ploidy of hMKs, but also hPLT release in vitro and in vivo. SR1-treated hMKs resulted in a 3-fold increased yield of normal size, T1/2 and functional PLTs/infused hMK compared to a DMSO-treated control. In summary, like mMKs, infused hMKs into mice release PLTs in the pulmonary vasculature though at a lower efficiency. Released hPLTs were functional and T1/2 was as expected. diMF enhanced MK ploidy, but worsened PLT yield and T1/2, while an AhR antagonist SR1 that also improved MK ploidy appears to markedly enhance yield of PLT/infused hMK, while maintaining T1/2. The ability of SR1 to enhance PLT release from induced pluripotent stem cells (iPSCs)-derived MKs remains to be tested, but this drug appears to be a strong candidate for a therapeutic strategy to take ex vivo-grown hMKs and generate PLTs in clinical relevant numbers. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1413-1413
Author(s):  
Abraham Avigdor ◽  
Chiara Medaglia ◽  
Alexander Kalinkovich ◽  
Tomer Itkin ◽  
Menachem Bitan ◽  
...  

Abstract Introduction Motility, proliferation, bone marrow (BM) retention and egress to the circulation of hematopoietic stem and progenitor cell (HSPCs) are key elements in normal hematopoiesis and also in the pathogenesis of acute myeloid leukemia (AML). HSPCs are tightly regulated in the BM niche by various molecules. Among them, CXCL12 and its major receptor CXCR4, expressed by both HSPCs and BM niche components, govern HSPC retention in the BM. Administration of CXCR4 antagonists lead to mobilization of both normal HSPCs and leukemia-inducing stem cells (LICs) from the BM to the circulation. We have previously found that normal human HSPCs functionally express b2-adrenergic receptors (b2-AR) that are up-regulated by myeloid cytokines such as G-CSF. The catecholaminergic neurotransmitters epinephrine and norepinephrine (NE) activate both Wnt and GSK3β signaling pathways via b2-AR, leading to enhanced HSPC proliferation, motility and BM repopulation (Spiegel et al, Nat Immunol 2007; Lapid et al, JCI 2013). These findings indicate HSPC regulation by dynamic interactions of the sympathetic nervous and hematopoietic systems. However, the role of catecholamines in regulation of AML remains elusive. Results We found that several human AML cell lines from different FAB subtypes express b2-AR. NE, a b2-AR activating ligand, increased b2-AR expression and the CXCL12-induced migration of AML primary patients’ cells and cell lines. In addition, NE treatment significantly enhanced CXCL12induced actin polymerization, which drives most of the cellular movements. Looking for a downstream effector of b2-AR, we focused on RGS16, a G-protein signaling regulator, which negatively regulates CXCL12/CXCR4 axis (Berthebaud et al., Blood 2005). Concurrently with the increase in cell migration, NE decreased RGS16 expression (both in protein and mRNA level) in monocytic AML cells. However, no effect on either cell migration or RGS16 expression was observed in non-monocytic AML cells. These data provided evidence for the involvement of catecholamines in the regulation of AML cell migration and showed a correlation between AML FAB subtypes, cell motility and RGS16 expression. One of the regulators of RGS16 levels is miR126, which is highly expressed in AML and normal HSPCs and plays an important role in mobilization and proliferation of normal HSPCs. Indeed, in search for the mechanisms underlying the above observed differences, we found that the enhancing effect of NE on CXCL12-induced migration of monocytic AML cells was accompanied by up-regulation of miR126 expression concurrently with down-regulation of RGS16 expression, whereas in non-monocytic AML cells we observed the opposite effects, suggesting that NE differently regulates AML cells belonging to different FAB subtypes. Upon studying human normal HSPCs, we found that in steady state, normal cells express low levels of β2-AR and NE did not affect either RGS16 expression or CXCL12-induced migration of both mononuclear and CD34+ cells derived from human cord blood and BM. Conclusions Our results demonstrate that while normal and AML cells share common mechanisms that govern their motility, there are unrevealed yet mechanisms, apparently cell-type dependent, which uniquely lead to opposite effects in normal HSPCs, monocytic and non-monocytic AML cells. Altogether, these findings suggest that targeting of miR126 and RGS16 pathways by specific agonists and antagonists may serve as a new approach for selective eradication of LICs. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3674-3674
Author(s):  
Michihiro Kobayashi ◽  
Yuanshu Dong ◽  
Hao Yu ◽  
Yunpeng Bai ◽  
Sisi Chen ◽  
...  

Abstract The phosphatase of regenerating liver family of phosphatases, consisting of PRL1, PRL2 and PRL3, represents an intriguing group of proteins implicated in cell proliferation and tumorigenesis. However, the role of PRLs in normal and malignant hematopoiesis is largely unknown. While SCF/KIT signaling plays an important role in hematopoietic stem and progenitor cell (HSPC) maintenance, how SCF/KIT signaling is regulated in HSPCs remains poorly understood. We here report that PRL2 regulates HSPC maintenance through regulating SCF/KIT signaling. To define the role of PRL2 in hematopoiesis, we analyzed the hematopoietic stem cell (HSC) behavior in Prl2 deficient mice generated by our group. Prl2 deficiency results in ineffective hematopoiesis and impairs the long-term repopulating ability of HSCs. In addition, Prl2 null HSPCs are less proliferative and show decreased colony formation in response to SCF stimulation. Furthermore, Prl2 null HSPCs show reduced activation of the PI3K/AKT and ERK signaling in steady state and following SCF stimulation. Importantly, we found that PRL2 associates with KIT and the ability of PRL2 to enhance SCF signaling depends on its enzymatic activity, demonstrating that PRL2 mediates SCF/KIT signaling in HSPCs. Thus, PRL2 plays a critical role in hematopoietic stem and progenitor cell maintenance through regulating SCF/KIT signaling. Furthermore, loss of Prl2 decreased the ability of oncogenic KITD814V mutant in promoting hematopoietic progenitor cell proliferation and in activation of signaling pathways. We also checked the expression of PRL2 proteins in human AML cell lines and found increased level of PRL2 proteins in some acute myeloid leukemia (AML) cells compared with normal human bone marrow cells, indicating that PRL2 may play a pathological role in AML. Our results suggest that the PRL2 phosphatase may be a druggable target in myeloproliferative disease (MPD) and acute myeloid leukemia (AML) with oncogenic KIT mutations. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document