Ezrin Regulates Hematopoietic Stem/Progenitor Cell Motility

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 ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 214-214
Author(s):  
Tomer Itkin ◽  
Aya Ludin ◽  
Shiri Gur-Cohen ◽  
Carolin Ludwig ◽  
Robert Brooks ◽  
...  

Abstract Abstract 214 MicroRNAs (miRNAs) are small non-coding RNAs involved in various physiological processes, including hematopoiesis. Although miRNAs are broadly studied with regards to normal and malignant leukocyte development, the role of miRNAs in hematopoietic stem and progenitor (HSPC) migration and mobilization is poorly understood. Currently, induction of HSPC mobilization from the bone marrow (BM) to the peripheral blood (PB) is the major mean to harvest HSPCs for clinical transplantation. Recently, several miRNAs were found to be upregulated in macaque G-CSF-mobilized CD34+ HSPCs, among them the oncogenic miRNA mir-155 (Donahue et al., Blood 2009). To study the involvement of mir-155 in HSPC regulation, we examined hematopoiesis in mir-155 knock out (KO) mice. Of interest, mir-155 KO mice had normal BM and PB levels of mature cells, but reduced levels of immature BM Lineage−/Sca-1+/c-Kit+ (LSK) and primitive BM CD34−LSK HSPCs. Profiling of mir-155 expression in murine hematopoietic BM populations, following G-CSF treatment, revealed differential expression patterns in wild type (WT) mice. G-CSF treatment upregulated mir-155 levels in immature LSK cells, in T-cells and in Mac-1+/Gr-1+ monocyte/macrophages. In contrast, G-CSF downregulated mir-155 levels in common lymphoid progenitors and in B-cells. Suggesting that mature hematopoietic cells may also participate in HSPC mobilization process. G-CSF administration to mir-155 KO mice resulted in reduced HSPC mobilization, as assessed by CFU-C and LSK cell counts in the PB. Surprisingly, G-CSF treatment increased BM LSK cell frequency in mir-155 KO mice to the same levels as in WT mice. On the contrary, G-CSF treatment reduced BM CD34−LSK cell frequency in WT mice and increased it in mir-155 KO mice showing an opposing effect on the more primitive HSPC population. Since mir-155 is involved also in mesenchymal development regulating osteoblast differentiation, we propose that BM HSPC pool reduction could be mediated by the stromal microenvironment. Additionally, osteoblasts and other BM residing cells undergo substantial changes in response to G-CSF that might be mediated by mir-155. To determine whether the mobilization defect is hematopoietic cell-autonomous or due to an abnormal microenvironment, we examined G-CSF-induced mobilization in chimeric mice. Mir-155 KO mice reconstituted with wild type (WT) BM cells had normal mobilization as WT mice reconstituted with WT BM cells. Of interest, WT mice reconstituted with mir-155 KO BM cells showed reduced mobilization as mir-155 KO mice reconstituted with mir-155 KO BM cells. These results indicate that the mobilization defect in mir-155 KO mice is also due to a defect in HSPC motility. Since the CXCL12/CXCR4 axis plays a major role in HSPC mobilization, we examined the ability of mir-155 KO cells to perform CXCL12-induced migration and found reduced migration capacity of HSPCs in vitro. Although having reduced migration potential, mir-155 KO LSK cells had normal CXCR4 expression levels, suggesting that an aberrant intracellular response to SDF-1 is responsible for the observed defect. In support, AMD3100 treatment to mir-155 KO mice resulted in reduced HSPC rapid mobilization. Since SHIP-1 phosphatase mRNA is targeted by mir-155 in hematopoietic cells (Costinean et al., Blood 2009) and SHIP-1 KO hematopoietic cells exhibit increased migration towards CXCL12 (Kim et al., JCI 1999), we examined intracellular SHIP-1 expression during HSPC mobilization. SHIP-1 levels were downregulated in WT BM LSK cells in response to G-CSF or AMD3100 mobilizing treatments. In contrast, mir-155 KO BM LSK cells upregulated SHIP-1 levels in response to the mobilizing treatments. These results suggest that mir-155 may promote HSPC mobilization and increased motility via SHIP-1 downregulation. In summary, our data indicates that mir-155 directly promotes HSPC motility and mobilization by SHIP-1-mediated regulation of intracellular response to CXCL12 signaling. We also propose the mechanism of indirect regulation of BM HSPC pool size during steady state and following G-CSF treatment by mir-155, via stromal BM microenvironment, which is currently under investigation. Deciphering the mechanisms of HSPC migration and maintenance in general and by mir-155 in particular, may potentially improve clinical mobilization protocols and contribute to increased donor BM engraftment following transplantation. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2678-2678
Author(s):  
Florian E. Toegel ◽  
Jorge Isaac ◽  
Zhuma Hu ◽  
Kathy Weiss ◽  
Christof Westenfelder

Abstract The chemokine stromal cll-derived factor-1 (SDF-1) and its receptor CXCR4, expressed by hematopoietic stem cells (HSC) and other cells, represents a major mediator-system for G-CSF-induced hematopoietic stem cell (HSC) mobilization and stem cell (SC) homing to the bone marrow (BM) after BM transplantation. HSC have been shown to contribute to repair processes in kidneys damaged by ischemia and HSC as well as endothelial progenitor cells (EPC) are mobilized in response to kidney damage. The signals mediating this process are currently unknown. We tested the hypothesis that SDF-1 mediates both mobilization and homing of SCs into the kidney with acute renal failure (ARF) and may thus be crucial to SC-mediated organ repair. SDF-1 is expressed in normal kidneys and upregulated in ischemically injured (60 min bi- or unilateral renal pedicle clamp) mouse kidneys (FVB strain) as evaluated by immunohistochemistry, in-situ hybridization, quantitative RT-PCR and ELISA. Post ARF SDF-1 was upregulated in sublethally injured proximal tubular cells (PTC), peripheral blood levels of SDF-1 rose, and circulating CD34+ and Colony-forming Units of Cell numbers as well as EPCs increased significantly. In vitro chemotaxis of SC from the top of transwell inserts towards the bottom well, containing cultured PTC that exhibited augmented SDF-1 expression post partial ATP depletion, increased significantly, a response that was abolished when SC cells were preincubated with CXCR4 antibody. Injected, CFDA-labeled, BM cells were only recruited to ischemic kidneys (bi- or unilateral), a response that was also blocked after SC pre-incubation with CXCR4 antibody. Our data show that there is increased renal SDF-1 expression after ARF and secretion into the circulation, which, in turn, stimulates HSC and EPC-mobilization into the circulation and directs homing into the post-ischemic kidney. Therefore, both circulating and administered SC and other CXCR4-expressing cells can be recruited to the injured kidney, where they, as we showed previously, are able to greatly support effective tissue repair, functional recovery and animal survival following ARF.


Blood ◽  
2011 ◽  
Vol 118 (15) ◽  
pp. 4093-4101 ◽  
Author(s):  
Yiyue Zhang ◽  
Hao Jin ◽  
Li Li ◽  
F. Xiao-Feng Qin ◽  
Zilong Wen

Abstract The establishment of the HSC pool in vertebrates depends not only on the formation and the propagation of these stem cells but also on their proper trafficking among the defined hematopoietic organs. However, the physiologic mechanisms that regulate HSC mobilization remain elusive. Through analysis of the zebrafish cmyb mutant cmybhkz3, we show that the suppression of cMyb function abrogates larval and adult hematopoiesis, with concomitant accumulation of hematopoietic stem/progenitor cells (HSPCs) in their birthplace, the ventral wall of the dorsal aorta (VDA). Cell tracking and time-lapse recording reveal that the accumulation of HSPCs in cmybhkz3 mutants is caused by the impairment of HSPC egression from the VDA. Further analysis demonstrates that the HSPC migratory defects in cmybhkz3 mutants are at least partly because of adversely elevated levels of chemokine stromal cell-derived factor 1a (Sdf1a). Our study reveals that cMyb plays a hitherto unidentified role in dictating physiologic HSPC migration by modulating Sdf1a signaling.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1994-1994
Author(s):  
Mercedes Dessy-Rodriguez ◽  
Sara Fañanas-Baquero ◽  
Veronica Venturi ◽  
Salvador Payan ◽  
Cristian Tornador ◽  
...  

Abstract Congenital dyserythropoietic anemias (CDAs) are a group of inherited anemias that affect the development of the erythroid lineage. CDA type II is the most common one: it accounts for around 60% of all cases, and more than 600 cases have been reported so far. CDA II is caused by biallelic mutations in the SEC23B gene and is characterized by ineffective erythropoiesis with morphologic abnormalities of erythroblasts, hemolysis, and secondary iron overload, which is the most frequent complication. Patients usually suffer from variable degrees of jaundice, splenomegaly, and absolute reticulocyte count inadequate depending on the degree of anemia. Hydrops fetalis, aplastic crisis and gallstones are other associated clinical signs. CDA II bone marrow is characterized by the presence of more than 10% mature binucleated erythroblasts. Another distinctive feature of CDA II erythrocytes is hypoglycosylation of membrane proteins. The management of CDA II is generally limited to blood transfusion and iron chelation. Splenectomy has proved to reduce the number of transfusions in CDA II patients. However, allogenic hematopoietic stem cell transplant (HSCT) represents the only curative option for this disease. Autologous HSCT of genetically corrected cells will mean a definitive treatment for CDA II, overcoming the limitations of allogeneic HSCT, such as limited availability of HLA-matched donors, infections linked to immunosuppression or development of graft versus host disease. This strategy has been used to treat many inherited hematological diseases, including red blood cell diseases such as β-thalassemia, sickle cell disease or pyruvate kinase deficiency. Therefore, we have addressed a similar strategy to be applied to CDAII patients. Two different lentiviral vectors carrying either wild type or codon optimized versions of SEC23B cDNA (wtSEC23B LV or coSEC23B LV, respectively) under the control of human phosphoglycerate kinase promoter (PGK) have been developed. Taking advantage of a CDA II model, in which SEC23B knock-out was done in human hematopoietic progenitors through gene editing, we have determined the most effective SEC23B LV version and the most suitable multiplicity of infection (MOI) to compensate protein deficiency. SEC23B knock out human hematopoietic progenitors (CD34 + cells; 80% frame shift mutations; SEC23BKO) showed a sharp reduction in SEC23B protein level. Those SEC23BKO hematopoietic progenitors were transduced with both lentiviral vectors at MOIs ranged from 3 to 25. We observed that SEC23B protein reached physiological or even supraphysiological levels. In addition, the reduction in the number of erythroid colony forming units (CFUs) identified in SEC23BKO CD34 + cells, was partially restored in the LV transduced SEC23BKO progenitors. Significantly, we observed a clear correlation between the used MOI and the vector copy number (VCN) in the CFUs derived from transduced SEC23BKO CD34 + cells. Furthermore, SEC23BKO hematopoietic progenitors were subjected to an in vitro erythroid differentiation protocol. A sharp decrease in the cell growth throughout erythroid differentiation was observed in SEC23BKO condition. However, the transduction with any of SEC23B LVs at MOIs above 10 was able to recover cell expansion to values equal to wild type cells. Interestingly, total level of protein glycosylation during erythroid differentiation was enhanced after SEC23B LV transduction. Glycosylation level in wtSEC23B LV transduced SEC23BKO cells was most similar to the level in wild type cells. Then, we transduced peripheral blood-derived hematopoietic progenitors (PB-CD34 + cells) from a CDA II patient with wtSEC23B LV at MOI 25 and differentiated in vitro to erythroid cells. A complete restauration of SEC23B protein expression and a cell growth increase of wtSEC23B transduced CDAII was observed with vector copy numbers of 0.3 after 14 days under erythroid conditions. More importantly, we could find a decrease in the percentage of bi-/multinucleated erythroid cells generated in vitro after wtSEC23B LV transduction. In summary, SEC23B LV compensate the SEC23B deficiency in SEC23BKO and in CDAII hematopoietic progenitor cells, paving the way for gene therapy of autologous hematopoietic stem and progenitor cell as an alternative and feasible treatment for CDA II. Disclosures Bianchi: Agios pharmaceutics: Consultancy, Membership on an entity's Board of Directors or advisory committees. Sanchez: Bloodgenetics: Other: Co-Founder and promoter; UIC: Current Employment. Ramirez: VIVEBiotech: Current Employment. Segovia: Rocket Pharmaceuticals, Inc.: Consultancy, Research Funding. Quintana Bustamante: Rocket Pharmaceuticals, Inc.: Current equity holder in publicly-traded company.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1269-1269
Author(s):  
Lucia Fernandez ◽  
Africa Gonzalez ◽  
Carmen Sanchez-Valdepeñas ◽  
Luis Madero ◽  
Roland M Schmid ◽  
...  

Abstract Abstract 1269 Hematopoietic stem cells (HSCs) maintain the production of all blood cells through the lifespan of an organism, and regenerate the whole hematopoietic system after stressful episodes such as high dose chemotherapy or upon transplantation. The functions of HSCs in these 2 situations, steady-state and under stress, are controlled by a variety of molecules, which may provide different contribution to each process. We investigated whether the NF-kB alternative pathway might have a role in HSCs functions, using mice deficient for two components of this pathway: NF-kB-inducing kinase (NIK) or p52. The activation of NIK is generally known as the alternative (or non-canonical) NF-kB pathway, and drives the post-translational processing of p100 to mature p52, which results in the translocation to the nucleus of p52-containing complexes such as p52/RelB. Apart from the already reported defects in B-cell maturation, both NIK- and p52-deficient mice did not present major disturbances in blood cells numbers. The absolute numbers of marrow cells were not different among the knocked-out and the wild-type mice. We first studied the compartment of marrow cells known to be enriched for HSCs, either lineage-depleted Sca1-positive ckit-positive cells (LSK), or CD150 positive CD48 negative cells. The proportions of marrow cells with the immunophenotype of HSCs in either NIK-deficient or p52-deficient mice were similar to those in control mice. The amount of clonogeneic progenitor cells in the marrow was assessed in standard CFU-GM cultures, and gave no differences in output in any of the mice studied. We set up in vitro liquid cultures with murine stem cell factor and human interleukin-11, and determined the cellular production weekly. Cultures started with NIK-deficient marrow cells produced significantly less numbers of cells and CFU-GM, compared with those started with wild type marrow. This deficit in hematopoietic capacity was further confirmed in a more stringent assay of HSC function, the in vivo competitive repopulation assay. Equal numbers of lineage-depleted (Lin-) CD45.2 marrow cells of either NIK-deficient, p52-deficient or wild-type mice, were mixed with Lin- CD45.1 marrow cells of syngeneic mice, and transplanted into lethally irradiated CD45.1 recipients. Four months after transplant, the chimeric status and the hematopoietic lineage repopulation of CD45.2 cells was assessed in peripheral blood (PB). NIK- or p52-deficient HSCs repopulated the B-, T- and myeloid-lineages but at significantly lower levels when compared to wild type HSCs. Total donor CD45.2 cells and total CD45.2 LSK cells were also significantly lower in the marrows of mice transplanted with NIK- or p52-deficient HSCs versus those of controls. We used the marrows of the repopulated mice for secondary transplants, and confirmed the defect in the repopulating capacity of NIK- and p52-deficient HSCs. Our results suggest that the NF-kB alternative pathway plays a role in the function of HSCs, and this role may be important under stress conditions. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2811-2811
Author(s):  
Xiaomei Yan ◽  
Yue Zhang ◽  
Goro Sashida ◽  
Aili Chen ◽  
Xinghui Zhao ◽  
...  

Abstract Abstract 2811 MLL partial tandem duplication (MLL-PTD) is found in 5–8% of human MDS, secondary acute myeloid leukemia (s-AML) and de novo AML. The molecular and clinical features of MLL-PTD+ AML are different from MLL-fusion+ AML, although they share similar worse outcomes. Mouse knock-in model of Mll-PTD has been generated to understand its underlining mechanism (Dorrance et al. JCI. 2006). Using this model, we've recently reported hematopoietic stem/progenitor cell (HSPC) phenotypes of MllPTD/WT mice. Their HSPCs showed increased apoptosis and reduced cell number, but they have a proliferative advantage over wild-type HSPCs. Furthermore, the MllPTD/WT–derived phenotypic ST-HSCs/MPPs and even GMPs have self-renewal capabilities. However, MllPTD/WT HSPCs never develop MDS or s-AML in primary or transplanted recipient mice, suggesting that additional genetic and/or epigenetic defects are necessary for transformation (Zhang et al. Blood. 2012). Recently, high frequent co-existences of both MLL-PTD and RUNX1 mutations have been reported in several MDS, s-AML and de novo AML clinical cohorts, which strongly suggest a potential cooperation for transformation between these two mutations. Our previous study has shown that MLL interacts with and stabilizes RUNX1 (Huang et al. Blood. 2011). Thus, we hypothesize that reducing RUNX1 dosage may facilitate the MLL-PTD mediated transformation toward MDS and/or s-AML. We first generated the mice containing one allele of Mll-PTD in a Runx1+/− background and assessed HSPCs of MllPTD/wt/Runx1+/− double heterozygous (DH) mice. The DH newborns are runty; they frequently die in early postnatal stage and barely survive to adulthood, compared to the normal life span of wild type (WT) or single heterozygous (Mllwt/wt/Runx1+/− and MllPTD/wt/Runx1+/+) mice. We studied DH embryos fetal liver hematopoiesis and found reduced LSK and LSK/SLAM+ cells, partly because of increased apoptosis. Enhanced proliferation was found in DH fetal liver cells (FLCs) in vitro CFU replating assays over WT and MllPTD/wt/Runx1+/+ controls. DH FLCs also showed dominant expansion in both serial competitive and serial non-competitive BMT assays compared to WT controls. The DH derived phenotypic ST-HSCs/MPPs and GMPs also have enhanced self-renewal capabilities, rescuing hematopoiesis by giving rise to long-term repopulating cells in recipient mice better than cells derived from MllPTD/wt/Runx1+/+ mice. However, DH HSPCs didn't develop MDS or s-AML in primary or in serial BMT recipient mice. We further generated MllPTD/wt/Runx1Δ/Δ mice using Mx1-Cre mediated deletion. These mice showed thrombocytopenia 1 month after pI-pC injection, and developed pancytopenia 2–4 months later. All these MllPTD/wt/Runx1Δ/Δ mice died of MDS induced complications within 7–8 months, and tri-lineages dysplasias (TLD) were found in bone marrow aspirate. However, there are no spontaneous s-AML found in MllPTD/wt/Runx1Δ/Δ mice, which suggests that RUNX1 mutants found in MLL-PTD+ patients may not be simply loss-of-function mutations and present gain-of-function activities which cooperate with MLL-PTD in human diseases onsets. In conclusion, our study demonstrates that: 1) RUNX1 gene dosage reverse-correlates with HSPCs self-renewal activity; 2) Runx1 complete deletion causes MDS in Mll-PTD background. Future studies are needed to fully understand the collaboration between MLL-PTD and RUNX1 mutations for MDS development and leukemic transformation, which should facilitate improved therapies and patient outcomes. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1269-1269
Author(s):  
Colleen E. Annesley ◽  
Rachel E. Rau ◽  
Daniel Magoon ◽  
David Loeb ◽  
Patrick Brown

Abstract Background The WT1 gene encodes for a zinc finger-containing transcription factor involved in differentiation, cell cycle regulation and apoptosis. WT1 expression is developmentally regulated and tissue-specific, with expression maintained in the kidney and in CD34+ hematopoietic progenitor cells. Inactivating mutations of this tumor suppressor gene are well-described in sporadic Wilms tumor and as germline mutations in Wilms tumor predisposition syndromes. WT1 mutations have been reported in approximately 10% of both adult and pediatric patients with cytogenetically-normal acute myeloid leukemia (CN-AML), and have been associated with treatment failure and a poor prognosis. These reported mutations consist of insertions, deletions or point mutations. Many are frameshift mutations in exon 7, can occur as biallelic double mutations, and result in truncated proteins which may alter DNA-binding ability. Missense mutations in exon 9 have also been identified, and reports suggest that these may act in a dominant-negative manner, resulting in a loss of function. Despite these observations, the functional contribution of WT1 mutations to leukemogenesis is still largely undetermined. Methods/Results We obtained a novel knock-in WT1 mutant mouse model, which is heterozygous for the missense mutation R394W in exon 9, and homologous to exon 9 mutations seen in human AML. We hypothesized that WT1 mutations may have an aberrant effect on hematopoiesis, and specifically, could alter progenitor cell differentiation or proliferation. To investigate this, we collected lineage-negative bone marrow (lin- BM) cells from two-month old WT1 mutant (WT1mut) and wild-type (wt) mice. We performed methylcellulose colony-forming assays, serially replating cells every 10-12 days. Strikingly, WT1mut progenitor cells showed higher in vitro colony-forming capacity and an increased ability to serially replate, suggesting aberrantly enhanced self-renewal capability. Furthermore, WT1mut colonies from secondary and tertiary passages were larger and more cohesive than wild-type colonies, demonstrating increased proliferation and morphology consistent with blast colony-forming units (CFU-blast). Flow cytometric analysis of these WT1mut cells at tertiary replating revealed an immature, largely c-Kit+ population. Next, in order to study the effects of WT1mut on HSCs in vivo, we performed serial competitive transplantation of HSC-enriched, lineage-depleted BM into lethally irradiated mice. At 14 weeks post-transplant, the donor bone marrow cells were harvested and analyzed by flow cytometry. We observed a significant expansion of the LT-HSC compartment in the WT1mut mice compared to wild-type mice. These data provide new insight into the biology and functional role of WT1 mutations in the aberrant regulation of hematopoietic stem and progenitor cell expansion. Conclusion Oncogenic WT1 mutations confer enhanced proliferation and renewal of myeloid progenitor cells in vitro and expansion of LT-HSCs in vivo. Our findings suggest that WT1 mutations enhance stem cell self-renewal, potentially priming these cells for leukemic transformation upon acquisition of cooperative events. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 659-659 ◽  
Author(s):  
Darja Karpova ◽  
Michael P. Rettig ◽  
Linda Eissenberg ◽  
Julie Ritchey ◽  
Matthew Holt ◽  
...  

Abstract Introduction : Since the first description of hematopoietic stem and progenitor cell (HSPC) mobilization over forty years ago, it has become the standard of care for both autologous and allogeneic transplantation. A five-day course of G-CSF represents the most commonly used mobilization regimen today. The CXCR4 inhibitor, plerixafor, is a more rapid but weak mobilizer when used as a single agent, thus emphasizing the need for faster acting agents with more predictable mobilization responses and fewer side effects. Methods : Given the critical role of VLA4/VCAM1 signaling for migration and retention of HSPC, we were seeking to identify small molecule antagonists of VLA4 with improved potency and bioavailability. Relative to previously described comparators Bio5192 (▢4▢1-specific) and firategrast (▢4▢1 and ▢4▢7 dual-specific), our lead candidate, CWHM-823, exhibited increased aqueous solubility and ~10-100 fold better activity in blocking VLA4 and mobilizing HSPC in mice. CWHM-823 pharmacokinetics and mobilization were assessed in BALB/c and DBA/2 mice at different doses (3 to 15 mg/kg) and time points (15 to 240 min) when administered alone or in combination with the truncated isoform of the CXCR2 agonist Gro-beta (tGro-β, 2.5 mg/kg, generously provided by GlaxoSmithKline). HSPC mobilization was monitored using flow cytometry and clonogenic in vitro assays. "True" stem cells were measured in a serial competitive transplantation assay. The combination of tGro-β and VLA4 antagonist was further tested in diabetic mice in comparison to G-CSF (9 x 100μg/kg, q12h). RNA profiling of flow-sorted HSPC was performed via microarray analysis. Results : The combination of tGro-β with each VLA4 antagonist resulted in a dramatic synergistic increase in circulating HSPC numbers when compared to steady state (50-70-fold) or treatment with single agents (3-10 fold) including tGroβ. Mobilization with tGro-β + CWHM-823 was rapid, peaking at 15-30 minutes after injection. In a model of streptozotocin-induced diabetes, the mobilopathy (reduction in stem cell mobilization compared to wild type mice) was considerably less pronounced with the combination tGro-β + CWHM-823 (~1.5-fold lower CFU mobilization in diabetic mice) versus the 5-day course of G-CSF (~3-fold reduction). Despite the superior progenitor cell mobilization achieved with G-CSF (~2-fold more CFU and LSK/ml), the concentration of serially repopulating units (RU) was equally high in the tGro-β + CWHM-823 and G-CSF mobilized grafts suggesting a higher HSC frequency (1 RU out of 200 vs. 1 RU out of 400 LSK/CFU) in the tGro-β + CWHM-823 mobilized grafts (Figure 1). RNA profiling demonstrated close similarity between the expression profile of tGro-β + CWHM-823 mobilized, BM resident, and G-CSF mobilized LSK, with less than 0.5% of genes found to be significantly up- or downregulated. CXCR2 chemokine receptor stimulation was critical for the observed synergistic response, as pretreatment ("priming") or simultaneous treatment with tGro-β resulted in subsequent enhanced mobilization using VLA4 inhibitors, whereas reversed administration (VLA4 antagonist followed by tGro-β) had no effect on potency of either agent. Lack of surface CXCR2 expression on HSPC suggested that a rapidly acting effector molecule released from tGro-β-stimulated mature myeloid cells may subsequently influence VLA4-mediated HSPC adhesion/retention. Consistent with this theory, we observed increased protease MMP-9 in plasma within minutes after treatment with tGro-β + CWHM-823. Conclusions: We describe a novel strategy for rapid, reliable, and potent mobilization of HSPC in mice using a combination of VLA4 blockade (via novel and potent ▢4▢1 inhibitors) and CXCR2 activation (via tGro-β). The combination of tGro-β + VLA4 inhibitors or tGro-β followed by VLA4 inhibitors results in synergistic and rapid HSPC mobilization with quantity and quality of repopulating units similar to optimal mobilization with G-CSF. These data suggest further development of tGro-β + VLA4 inhibitor combinations for clinical testing is warranted. Figure 1. Mobilization of repopulating units (RU) (n=8-10 recipients per group, mean±SEM) Figure 1. Mobilization of repopulating units (RU) (n=8-10 recipients per group, mean±SEM) Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1998 ◽  
Vol 92 (3) ◽  
pp. 901-907 ◽  
Author(s):  
Takuya Matsunaga ◽  
Fumiya Hirayama ◽  
Yuji Yonemura ◽  
Richard Murray ◽  
Makio Ogawa

The receptors for interleukin-3 (IL-3), granulocyte-macrophage colony-stimulating factor (GM-CSF), and IL-5 share a common signaling subunit βc. However, in the mouse, there is an additional IL-3 signaling protein, βIL-3, which is specific for IL-3. We have previously reported that IL-3 abrogates the lymphoid potentials of murine lymphohematopoietic progenitors and the reconstituting ability of hematopoietic stem cells. We used bone marrow cells from βc- and βIL-3–knock-out mice to examine the relative contributions of the receptor proteins to the negative regulation by IL-3. First, we tested the effects of IL-3 on lymphohematopoietic progenitors by using lineage-negative (Lin−) marrow cells of 5-fluorouracil (5-FU)-treated mice in the two-step methylcellulose culture we reported previously. Addition of IL-3 to the combination of steel factor (SF, c-kit ligand) and IL-11 abrogated the B-lymphoid potential of the marrow cells of both types of knock-out mice as well as wild-type mice. Next, we investigated the effects of IL-3 on in vitro expansion of the hematopoietic stem cells. We cultured Lin−Sca-1–positive, c-kit–positive marrow cells from 5-FU–treated mice in suspension in the presence of SF and IL-11 with or without IL-3 for 7 days and tested the reconstituting ability of the cultured cells by transplanting the cells into lethally irradiated Ly-5 congenic mice together with “compromised” marrow cells. Presence of IL-3 in culture abrogated the reconstituting ability of the cells from both types of knock-out mice and the wild-type mice. In contrast, addition of GM-CSF to the suspension culture abrogated neither B-cell potential nor reconstituting abilities of the cultured cells of wild-type mice. These observations may have implications in the choice of cytokines for use in in vitro expansion of human hematopoietic stem cells and progenitors. © 1998 by The American Society of Hematology.


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.


Sign in / Sign up

Export Citation Format

Share Document