Bone Morphogenetic Protein 4 Regulates Hematopoietic Stem Cell Maintenance in Vivo.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1399-1399
Author(s):  
Devorah C. Goldman ◽  
Alexis S. Bailey ◽  
Dana L. Pfaffle ◽  
Jan L. Christian ◽  
William H. Fleming

Abstract The secreted signaling molecule Bone Morphogenetic Protein 4 (BMP4) is expressed by osteoblasts and other cell types that comprise the hematopoietic microenvironment. Therefore, we hypothesized that BMP4 may play an important regulatory role in the maintenance and function of hematopoietic stem cells (HSCs). As the deletion of BMP4 is lethal during early embryogenesis, we exploited a viable BMP4 hypomorph in which a point mutation in BMP4 reduces, but does not abolish the amount of active BMP4 ligand. In these mutants, peripheral blood cell lineages and bone marrow cellularity are normal during steady state conditions. However, consistent with our hypothesis, 40% fewer c-kit+, Sca-1+, lineage- (KSL) cells were present in the femurs of mutants (7.4 x103 ± 0.4 x103 SEM) than in age-and sex-matched wild-type (WT) controls (12.4 x103 ± 1.3 x103, p<0.005). Transplantation of mutant KSL cells produced levels of hematopoietic engraftment indistinguishable from transplanted WT KSL cells, consistent with a stem cell extrinsic effect of BMP4. To functionally assess the mutant hematopoietic microenvironment, unfractionated WT bone marrow cells (BM) were transplanted into lethally irradiated mutants or WT controls. Although WT cells could engraft mutant primary recipients to the same degree as WT recipients, serial transplantation of these WT cells into secondary WT hosts revealed a marked depletion of hematopoietic reconstitution activity. Specifically, nearly a 4-fold reduction of donor cells was found in the peripheral blood of secondary recipients that received BM from reconstituted mutant hosts (p<0.0005). To determine whether such defects in the mutant microenvironment exist in the absence of myeloablative conditioning and transplantation, a parabiotic mouse model was employed. CD45.1 WT mice were joined to CD45.2 BMP4 hypomorphs for 8 weeks and then separated. As early as one month following separation, a greater than 2- fold reduction in circulating WT donor cells was detected in mutant hosts relative to the number of mutant donor cells detected in the WT hosts (p<0.005). Analysis of BM at 24 weeks following separation revealed a striking 23- fold reduction in WT donor hematopoietic cells in the mutant host BM compared to the number of mutant hematopoietic cells detected in WT host BM. Together, our findings reveal a novel, critical role for BMP4 in maintaining HSCs in vivo.

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2476-2476
Author(s):  
Kasia Mierzejewska ◽  
Ewa Suszynska ◽  
Sylwia Borkowska ◽  
Malwina Suszynska ◽  
Maja Maj ◽  
...  

Abstract Background Hematopoietic stem/progenitor cells (HSPCs) are exposed in vivo to several growth factors, cytokines, chemokines, and bioactive lipids in bone marrow (BM) in addition to various sex hormones circulating in peripheral blood (PB). It is known that androgen hormones (e.g., danazol) is employed in the clinic to treat aplastic anemia patients. However, the exact mechanism of action of sex hormones secreted by the pituitary gland or gonads is not well understood. Therefore, we performed a complex series of experiments to address the influence of pregnant mare serum gonadotropin (PMSG), luteinizing hormone (LH), follicle-stimulating hormone (FSH), androgen (danazol) and prolactin (PRL) on murine hematopoiesis. In particular, from a mechanistic view we were interested in whether this effect depends on stimulation of BM-residing stem cells or is mediated through the BM microenvironment. Materials and Methods To address this issue, normal 2-month-old C57Bl6 mice were exposed or not to daily injections of PMSG (10 IU/mice/10 days), LH (5 IU/mice/10 days), FSH (5 IU/mice/10 days), danazol (4 mg/kg/10 days) and PRL (1 mg/day/5days). Subsequently, we evaluated changes in the BM number of Sca-1+Lin–CD45– that are precursors of long term repopulating hematopoietic stem cells (LT-HSCs) (Leukemia 2011;25:1278–1285) and bone forming mesenchymal stem cells (Stem Cell & Dev. 2013;22:622-30) and Sca-1+Lin–CD45+ hematopoietic stem/progenitor cells (HSPC) cells by FACS, the number of clonogenic progenitors from all hematopoietic lineages, and changes in peripheral blood (PB) counts. In some of the experiments, mice were exposed to bromodeoxyuridine (BrdU) to evaluate whether sex hormones affect stem cell cycling. By employing RT-PCR, we also evaluated the expression of cell-surface and intracellular receptors for hormones in purified populations of murine BM stem cells. In parallel, we studied whether stimulation by sex hormones activates major signaling pathways (MAPKp42/44 and AKT) in HSPCs and evaluated the effect of sex hormones on the clonogenic potential of murine CFU-Mix, BFU-E, CFU-GM, and CFU-Meg in vitro. We also sublethally irradiated mice and studied whether administration of sex hormones accelerates recovery of peripheral blood parameters. Finally, we determined the influence of sex hormones on the motility of stem cells in direct chemotaxis assays as well as in direct in vivo stem cell mobilization studies. Results We found that 10-day administration of each of the sex hormones evaluated in this study directly stimulated expansion of HSPCs in BM, as measured by an increase in the number of these cells in BM (∼2–3x), and enhanced BrdU incorporation (the percentage of quiescent BrdU+Sca-1+Lin–CD45– cells increased from ∼2% to ∼15–35% and the percentage of BrdU+Sca-1+Lin–CD45+ cells increased from 24% to 43–58%, Figure 1). These increases paralleled an increase in the number of clonogenic progenitors in BM (∼2–3x). We also observed that murine Sca-1+Lin–CD45– and Sca-1+Lin–CD45+ cells express sex hormone receptors and respond by phosphorylation of MAPKp42/44 and AKT in response to exposure to PSMG, LH, FSH, danazol and PRL. We also observed that administration of sex hormones accelerated the recovery of PB cell counts in sublethally irradiated mice and slightly mobilized HSPCs into PB. Finally, in direct in vitro clonogenic experiments on purified murine SKL cells, we observed a stimulatory effect of sex hormones on clonogenic potential in the order: CFU-Mix > BFU-E > CFU-Meg > CFU-GM. Conclusions Our data indicate for the first time that not only danazol but also several pituitary-secreted sex hormones directly stimulate the expansion of stem cells in BM. This effect seems to be direct, as precursors of LT-HSCs and HSPCs express all the receptors for these hormones and respond to stimulation by phosphorylation of intracellular pathways involved in cell proliferation. These hormones also directly stimulated in vitro proliferation of purified HSPCs. In conclusion, our studies support the possibility that not only danazol but also several other upstream pituitary sex hormones could be employed to treat aplastic disorders and irradiation syndromes. Further dose- and time-optimizing mouse studies and studies with human cells are in progress in our laboratories. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 20-20
Author(s):  
James Bartram ◽  
Baobao (Annie) Song ◽  
Juying Xu ◽  
Nathan Salomonis ◽  
H. Leighton Grimes ◽  
...  

Abstract Hematopoietic stem cells are endowed with high regenerative potential but their actual self-renewal capacity is limited. Studies using the H2B-retention labeling system show HSC functional decline at each round of division (Qiu, Stem Cell Reports 2014). We have shown that mitochondria drive HSC functional decline with division history after transplantation (Cell Stem Cell 2020). Here we examined the link between mitochondrial metabolism, in vivo division at steady state, and HSC functions using the GFP label-Histone 2B (GFP-H2B) mouse model driven by a doxycycline-inducible promoter. Five months after doxycycline removal, mitochondrial membrane potential (MMP) was examined using TMRE in HSC with varying GFP intensity. HSC were separated into an H2B-labeled retention population and an H2B-labeled population. Interestingly, within the H2B-labeled retention population, HSC could be further subdivided into GFP high, medium, and low. MMP increased in a stepwise fashion with GFP dilution in HSC. We noted the presence of 2 TMRE peaks within each GFP high and medium populations leading to 5 populations: GFP-high;MMP-low (G1), GFP-high;MMP-high (G2), GFP-medium;MMP-low (G3), GFP-medium;MMP-high (G4), GFP-low;MMP-high (G5). We examined the repopulation activity of each population in a serial competitive transplant assay. G1 and G2 maintained higher peripheral blood chimerism up to 24 weeks post-transplant than G3 and G4. G5 did not engraft at all. However, only G1 reconstituted high frequency of HSC in primary recipients. In secondary recipients, G1, G2, G3 but not G4 gave rise to positive engraftment. Interestingly, G1 and G2 grafts showed myeloid/lymphoid balanced engraftment whereas the G3 graft was myeloid-bias, suggesting that myeloid skewing can be acquired upon HSC division. We further examined lineage fate maps of bone marrow cells derived from G1 or G3 population in vivo, using single cell RNA sequencing, 10X genomics. Surprisingly, G3-derived bone marrow cells displayed a distinct myeloid cell trajectory from G1-derived bone marrow cells, in which G3 gave rise to increased immature neutrophils but fewer myeloid precursors. Remarkably, each lineage population derived from G3 donor cells had different gene expression signatures than those derived from G1 donor cells. Therefore, HSC that have divided in vivo in the same bone marrow microenvironment are intrinsically and molecularly different such that not only do they exhibit lineage potential differences but they also produce progeny that are transcriptionally different. These findings imply that cellular division rewires HSC and that this rewiring is passed down to their fully differentiated progeny. When G1 and G3 single HSC were cultured in-vitro, G1 had a slower entry into cell-cycle which has been associated with increased stemness. Additionally, when single HSC from G1 and G3 were assessed for their multipotency in a lineage differentiation assay, G1 HSC had a higher propensity to produce all four myeloid lineages (megakaryocytes, neutrophils, macrophages, and erythroid), further supporting increased stemness in G1 compared to G3 HSC. Finally, HSC from G1, G2, G3 and G4 populations carried mitochondria that were morphologically different, and express distinct levels of Sca-1, CD34 and EPCR, with Sca-1 high, CD34-, EPCR+ cells more enriched in G1. In summary, this study suggests that HSC transition into distinct metabolic and functional states with division history that may contribute to HSC diversity and functional heterogeneity. It also suggests the existence of a cell-autonomous mechanism that confers HSC divisional memory to actively drive HSC functional heterogeneity and aging. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1993 ◽  
Vol 82 (2) ◽  
pp. 445-455 ◽  
Author(s):  
DM Bodine ◽  
NE Seidel ◽  
KM Zsebo ◽  
D Orlic

We have examined the effects of administration of stem cell-factor (SCF) on the number and distribution of pluripotent hematopoietic stem cells (PHSC) in normal mice. Using the competitive repopulation assay we found that in vivo administration of SCF increases the absolute number of PHSC per mouse threefold. The increased numbers of PHSC are found in the peripheral blood and spleen of the SCF-treated animals. The spleen and peripheral blood stem cells completely repopulated the erythroid, myeloid, and lymphoid lineages of irradiated or W/Wv hosts, similar to bone marrow PHSC. PHSC from the peripheral blood of SCF- treated mice have a lineage marker-negative, c-kit-positive phenotype that is indistinguishable from that of bone marrow PHSC. The increase in the absolute number of spleen PHSC is associated with efficient gene transfer to these cells without prior treatment with 5-fluorouracil. This is a US government work. There are no restrictions on its use.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 496-496
Author(s):  
Stefan P. Tarnawsky ◽  
Mervin C. Yoder ◽  
Rebecca J. Chan

Juvenile Myelomonocytic Leukemia (JMML) is a rare childhood myelodysplastic / myeloproliferative overlap disorder. JMML exhibits myeloid populations with mutations in Ras-Erk signaling genes, most commonly PTPN11, which confer growth hypersensitivity to GM-CSF. While allogeneic hematopoietic stem cell transplant (HSCT) is the treatment of choice for children with JMML, 50% of children succumb to leukemia relapse; however, the mechanism leading to this high relapse rate is unknown. We hypothesized that the hyperinflammatory nature of JMML may damage the bone marrow microenvironment, leading to poor engraftment of normal donor cells following transplant, permitting residual leukemia cells to outcompete the normal graft, and thus promoting leukemia relapse. Using Vav1 promoter-directed Cre, we generated a mouse model of JMML that conditionally expresses gain-of-function PTPN11D61Yin utero during development. While PTPN11D61Y/+; VavCre+embryos did not demonstrate in utero lethality, we observed a modest reduction of PTPN11D61Y/+; VavCre+ mice at the time of weaning compared to predicted Mendelian frequencies. Further, surviving PTPN11D61Y/+; VavCre+ mice developed elevated peripheral blood leukocytosis and monocytosis as early as 4 weeks of age compared to PTPN11+/+; VavCre+ controls. To address the hypothesis that an aberrant bone marrow microenvironment in the PTPN11D61Y/+ mice leads to poor engraftment of wild-type donor cells following transplant, we examined engraftment of wild-type hematopoietic stem and progenitor cells (HSPCs) in the PTPN11D61Y/+; VavCre+ mice and monitored animals for disease relapse. 16-24 week-old diseased PTPN11D61Y/+; VavCre+ and control PTPN11+/+; VavCre+ mice were lethally irradiated (11 Gy split dose) and transplanted with 5 x 105 CD45.1+ wild-type bone marrow low density mononuclear cells (LDMNCs), which simulates a limiting stem cell dose commonly available in a human HSCT setting. 6 weeks post-HSCT, PTPN11D61Y/+; VavCre+recipients demonstrated an unexpected elevated CD45.1+ donor cell contribution in peripheral blood compared to the control PTPN11+/+; VavCre+ recipients. However, despite superior engraftment in the PTPN11D61Y/+; VavCre+ recipients, these mice had a significantly shorter median survival post-HSCT due to a resurgence of recipient CD45.2-derived leukemic cells. We repeated the experiment using a high dose of CD45.1+ LDMNCs (10 x 106 cells) to determine if providing a saturating dose wild-type cells could prevent the relapse of recipient-derived leukemogenesis and normalize the survival of the PTPN11D61Y/+; VavCre+recipients. While this saturating dose of wild-type cells resulted in high peripheral blood chimerism in both the PTPN11D61Y/+; VavCre+ and PTPN11+/+; VavCre+ recipients, the PTPN11D61Y/+; VavCre+ animals nevertheless demonstrated significantly reduced overall survival. When we examined the cause of mortality in the HSCT-treated PTPN11D61Y/+; VavCre+mice, we found enlarged spleens, hypercellular bone marrow, and enlarged thymuses. Flow cytometry revealed that the majority of cells in the peripheral blood, bone marrow, and spleen were recipient-derived CD45.2+ CD4+ CD8+ T cells. To verify that the disease was neoplastic in origin, secondary transplants into CD45.1/.2 recipients were performed from two independent primary PTPN11D61Y/+; VavCre+and two independent primary PTPN11+/+; VavCre+ controls. Secondary recipients of bone marrow from PTPN11D61Y/+; VavCre+ animals rapidly succumbed to a CD45.2-derived T-cell acute lymphoid leukemia (T-ALL). Previous studies demonstrated that wild-type PTPN11 is needed to protect the integrity of the genome by regulating Polo-like kinase 1 (Plk1) during the mitosis of the cell cycle (Liu et al., PNAS, 2016). We now demonstrate that even when PTPN11 mutant animals are provided with saturating doses of wild-type HSCs, dysregulated residual recipient cells are able to produce relapsed disease. Collectively, these studies highlight the propensity of residual mutant PTPN11 cells to transform after being subjected to mutagenic agents that are commonly used for conditioning regimens prior to allogeneic HSCT. These findings suggest that modified pre-HSCT conditioning regimens bearing reduced mutagenicity while maintaining adequate cytoreductive efficacy may yield lower post-HSCT leukemia relapse in children with PTPN11mutations. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2410-2410
Author(s):  
Karolina Komorowska ◽  
Hanna K.A Mikkola ◽  
Jonas Larsson ◽  
Mattias Magnusson

Abstract The transcription factor Hepatic Leukemia Factor (HLF) was originally identified in a chromosomal translocation with the gene E2A causing a subset of childhood B-lineage acute lymphoid leukemia. Moreover, HLF has been described as a regulator of circadian rhythm and recent findings have implicated HLF as a candidate “stemness” gene in both normal and malignant stem cells. Accordingly, overexpression of HLF in human hematopoietic stem cells (HSC) results in an enhanced reconstitution capability in NOD-SCID mice. However, little is known about HLF’s physiological role in hematopoiesis and HSC regulation. Using quantitative PCR, we found that HLF is highly expressed in mouse (C57Bl/6) HSC and is downregulated upon differentiation (HSC 3.2 (±0.95) fold (p<0.001), LSK 1.9 (±0.47) fold (p<0.05), CMP, GMP MEP all less then 0.1 fold, all values are compared to HPRT). This encouraged us to further investigate HSC function in the absence of HLF. The conventional HLF knockout (KO) mice (C57bl/6 background) were viable, born at normal Mendelian ratios and showed normal hematopoietic parameters (bone marrow cellularity: WT 2.7x107 (±5.4 x106), KO 3.3x107 (±6.4 x106), p>0.2 n=9). In addition, the HLF KO mice demonstrated normal lineage distribution of both mature cells in the peripheral blood and bone marrow as well as the frequency of immunophenotypic HSC (Lin-Sca1+ckit+CD34-Flt3-: WT 0.0005 (±0.5x10-4)%, KO 0.0005 (±0.1x10-3)%; n>10). However, in a serial competitive transplantation assay using whole bone marrow (200 000 cells 1:1 ratio), HLF KO cells demonstrated a significant reduction in reconstitution capacity in primary recipients (WT 56 (±15)%, KO 40.2 (±16)%, p=0.028, n>10), which was further increased in the secondary recipients (WT 87.2 (±26)%, KO 8.7 (±5.8)%, p<0.001, n>10). Almost no engraftment was detected from the HLF KO cells in tertiary recipients. To further evaluate stem cell activity in the absence of HLF, we next enumerated the number of competitive repopulating units (CRU) by limiting dilution assay, which revealed a 2.6 fold reduction, of CRU in the HLF KO mice compared to WT controls (WT 1.6 (±0.4)/105 bone marrow cells, KO 0.6 (±0.2)/105 bone marrow cells). Similarly, transplantation of sorted HSC (Lin-Sca1+ckit+CD34-Flt3-) also showed a 2.4 fold (WT 47.3 (±24)%, KO 19.4 (±25)%, p=0.16, n=9) reduced engraftment of total cells but with enhanced T cell frequency in peripheral blood (WT 19.5 (±6.2)%, KO 40.8 (±7.4)%, p=0.01, n=9). Since we also found that HLF was highly expressed in fetal liver derived HSC, we transplanted fetal liver HLF KO cells from E14.5 in a competitive repopulation setting. In line with the phenotype seen in the adult HLF KO mice, the fetal liver HLF KO cells demonstrated impaired reconstitution ability (WT 52.8 (±16)%, KO 0.9 (±1.4)%, n>10). Intriguingly, the phenotype was stronger than in the adult HLF KO HSC, indicating that HLF is particularly important during the expansion phase of HSC in embryonic development. The underlying mechanism of the reduced HSC activity is still unclear, but preliminary findings show that HLF KO HSC have enhanced ROS levels (WT 337 (±33), KO 510 (±55), p<0.05, n=3) and increased cycling HSC (G0: WT 66.5 (±6.4)%, KO 58.5 (±4.7)%; G1/S/G2/M: WT 33.6 (±6.6)%, KO 41.7 (±4.9)%, n=3). We are currently performing global gene expression analysis to further understand the mechanism of HLF in HSC regulation. Interestingly, we also found that HLF appears to regulate the identity of HSC by modulating the expression of the SLAM code on the cell surface of the HLF KO HSC. In contrast to the normal frequency of LSK Flt3-CD34- cells, the HLF KO mice displayed a 3.5 fold reduction in the frequency of LSK CD150+CD48- cells (WT 1.94x10-4 (±4.4x10-5)%, KO 0.56x10-4 (±1.5x10-5)%, p<0.001 n>10). Strikingly, transplantation of as many as 150 LSK CD150+CD48-HLF KO cells showed a complete lack of repopulating capacity in vivo. This did not correlate to the number of functional HSC seen when transplanting whole bone marrow and indicates that HLF affects the identity of HSC by modulating the expression of the SLAM markers. Taken together, we show here for the first time that HLF has a fundamental role in HSC biology during both fetal and adult hematopoiesis by regulating HSC activity and identity. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1248-1248
Author(s):  
Esther Heideveld ◽  
Maartje van Den Biggelaar ◽  
Floris P. van Alphen ◽  
Marieke Von Lindern ◽  
Emile van den Akker

Abstract Erythropoiesis occurs in erythroblastic islands, specific structures in the bone marrow comprising a central macrophage surrounded by erythroid precursors at different stages of terminal differentiation. The central macrophage of the erythroblastic island supports proliferation and differentiation of erythroblasts, as well as phagocytosis of the extruded erythroblast nuclei, the pyrenocytes. Its identity, however, has been poorly characterized. We previously showed that macrophages also enhance in vitro erythropoiesis because they support hematopoietic stem cell (HSC) survival [Heideveld et al. 2015]. Thus, bone marrow macrophages affect all stages of erythropoiesis. The aim of our study is to characterize the relevant human bone marrow macrophages and unravel the mechanism by which they support erythropoiesis with the ultimate goal (i) to optimize erythroblast culture systems that produce erythrocytes for transfusion purposes, and (ii) to target macrophages in vivo to improve erythropoiesis in anemic patients. Macrophages are a heterogeneous population, that can be divided into pro-inflammatory M1 and anti-inflammatory M2 macrophages. Macrophages that we showed to support stem cell survival, and subsequently enhance the yield of erythroid cell cultures, were characterized as a subclass: M2c-like macrophages. These macrophages were derived from CD14+ cells isolated from human peripheral blood mononuclear cells that were cultured in serum-free media supplemented with stem cell factor, erythropoietin and dexamethasone. Within three days these macrophages expressed CD163high, CD169, mannose receptor (MR), CXCR4 and HLA-DR and harbored characteristics of bone marrow resident macrophages. This differentiation process was dependent on glucocorticoid receptor activation. Mass spectrometry of monocytes cultured in presence and absence of dexamethasone showed that expression of CD163 and MR was strictly Dex-dependent, underscoring the role of glucocorticoids in the phenotype of M2c macrophages. Protein ontology analysis revealed dexamethasone-mediated enrichment of lysosome, endocytosis and endothelial development (e.g. STAB1, IL13RA1, CD81, SLC1A3 and FKBP5). We wondered whether these macrophages with increased endosomal and lysosomal capacity not only support stem cell survival and enable erythroid commitment, but also support erythroblastic islands. In mice, it has been shown that clearance of the pyrenocytes by central macrophages occurs presumably via TAM-receptors on the macrophages. Indeed, mRNA expression of cultured M2c-like macrophages showed increased levels of TAM family members MerTK and AXL. Functionally, these macrophages have the capacity to phagocytose zymosan and to bind nuclei. Furthermore, co-culture of the M2c-like macrophages with erythroblasts yielded GPA+(erythroid marker)/CD14+ cell aggregates that suggested the formation of erythroblastic islands. Interestingly, M2c-like macrophages expressing CD163high, MR and CD169 were also observed in human bone marrow aspirates and human fetal livers resembling macrophages induced in in vitro cultures in presence of dexamethasone. Currently, we investigate the mechanism by which glucocorticoids induce monocytes to differentiate into macrophages that may be used to model erythroblastic island-mediated erythropoiesis. Knowledge on the function of such a erythroblastic island is lacking by the absence of an in vitro model. Furthermore, targeting this mechanism in vivo may enhance the recovery of erythropoiesis following bone marrow transplantation. CD14+ cells from peripheral blood positively regulate hematopoietic stem and progenitor cell survival resulting in increased erythroid yield. (2015) Heideveld E, Masiello F, Marra M, Esteghamat F, Yağcı N, von Lindern M, Migliaccio AR, van den Akker E. Haematologica. 100(11):1396-1406 Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2593-2593
Author(s):  
Valentina Goncharova ◽  
Shinji Iizuka ◽  
Yu Yamaguchi ◽  
Sophia Khaldoyanidi

Abstract Abstract 2593 The bone marrow microenvironment regulates a variety of hematopoietic stem cell (HSC) functions, including their recruitment into the marrow following transplantation. However, an insufficient understanding of the biology of the hematopoietic microenvironment reflects gaps in our knowledge of fundamental stem cell biology and remains an obstacle to an optimal therapeutic approach. While the involvement of hyaluronan (HA), one of the major components of the bone marrow ECM, in normal cell and tumor biology is generally appreciated, little is known of how HA contributes to the regulation of the hematopoietic microenvironment. In this study we investigated whether HA contributes to the recruitment of circulating HSCs into marrow. Stimulation of bone marrow cells with HA induced production of chemokines including SDF-1, MIP-1a, MIP-1b, IL-8, eotaxin, CXCL16, RANTES, LIX and MCP-1. HA-induced conditioned media containing these chemokines enhanced SDF-1-mediated HSC transmigration in vitro, whereas HA alone had no effect, suggesting that endogenous HA participates in the recruitment of HSC indirectly, via regulation of the production of chemokines. We next tested the relevance of this finding in vivo. Since HA synthase 2 knockout mice are embryonically lethal, we developed an alternative in vivo model that allows deprivation of HA. Mice were lethally irradiated to eliminate HSCs and to degrade endogenous HA, and injected with 4-MU, an HA synthase inhibitor, to prevent de novo synthesis of HA. Pre-treated mice were transplanted with HSCs, and after 24 hours the bone marrow was assayed by competitive reconstitution. This assay demonstrated that the number of HSCs which migrated into the marrow of mice pretreated with 4MU was significantly lower as compared to control. To further understand the role of endogenous HA in bone marrow microenvironment we used compound mutant mice in which Has1 and Has3, two genes encoding hyaluronan synthase, were knocked out (Has1-/-;Has3-/-). The number of hematopoietic progenitors (HPs) circulating in peripheral blood was significantly increased in the double knockout (dKO) mice as compared to the wild type (WT) mice. This increase in the number of circulating HP was further enhanced in triple Has knockout mice (Prx1-Cre;Has2flox/flox;Has1-/-;Has3-/-). This correlated with the higher number of hematopoietic progenitors in spleens of Has1-/-;Has3-/- double KO mice and Prx1-Cre;Has2flox/flox;Has1-/-;Has3-/- triple KO mice as compared to the WT mice. In summary, our data indicate that low levels of HA correlate with re-distribution of HSCs and HPs from bone marrow into peripheral blood and spleen. This suggests that HA associated with the hematopoietic microenvironment is important for recruitment and retention of HSCs and HPs, confirming that HA plays a critical regulatory role in the hematopoietic microenvironment. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1993 ◽  
Vol 82 (2) ◽  
pp. 445-455 ◽  
Author(s):  
DM Bodine ◽  
NE Seidel ◽  
KM Zsebo ◽  
D Orlic

Abstract We have examined the effects of administration of stem cell-factor (SCF) on the number and distribution of pluripotent hematopoietic stem cells (PHSC) in normal mice. Using the competitive repopulation assay we found that in vivo administration of SCF increases the absolute number of PHSC per mouse threefold. The increased numbers of PHSC are found in the peripheral blood and spleen of the SCF-treated animals. The spleen and peripheral blood stem cells completely repopulated the erythroid, myeloid, and lymphoid lineages of irradiated or W/Wv hosts, similar to bone marrow PHSC. PHSC from the peripheral blood of SCF- treated mice have a lineage marker-negative, c-kit-positive phenotype that is indistinguishable from that of bone marrow PHSC. The increase in the absolute number of spleen PHSC is associated with efficient gene transfer to these cells without prior treatment with 5-fluorouracil. This is a US government work. There are no restrictions on its use.


2015 ◽  
Vol 2015 ◽  
pp. 1-11 ◽  
Author(s):  
Lindsay T. McDonald ◽  
Meenal Mehrotra ◽  
Amanda C. LaRue

Multiple origins, including the bone marrow, have been suggested to contribute to fibroblast populations in the lung. Using bone marrow reconstitution strategies, the present study tested the hypothesis that the bone marrow hematopoietic stem cell (HSC) gives rise to lung tissue fibroblastsin vivo. Data demonstrate that the nonadherent bone marrow fraction is enriched for CD45+HSC-derived cells and was able to reconstitute hematopoiesis in lethally irradiated animals. Analysis of peripheral blood and lung tissues from engrafted mice demonstrated the ability of this population to give rise to CD45+/Discoidin-Domain Receptor-2+(DDR2) circulating fibroblast precursors (CFPs) in blood and fibroblast populations in lung. An HSC origin for lung fibroblasts was confirmed using a novel clonal cell transplantation method in which the bone marrow is reconstituted by a clonal population derived from a single HSC. Together, these findings provide evidence for an HSC contribution to lung fibroblasts and demonstrate a circulating intermediate through the CD45+/DDR2+HSC-derived CFP.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 87-87 ◽  
Author(s):  
Matthew J. Christopher ◽  
Priya K. Gopalan ◽  
Daniel C. Link

Abstract Accumulating evidence suggests that osteoblast-lineage cells play a key role in supporting hematopoiesis, providing signals that maintain the normal function of hematopoietic stem cells (HSC). We and others previously showed that treatment with G-CSF reduces the number and activity of osteoblasts in the bone marrow. Here we confirm these findings using transgenic mice expressing GFP in osteoblast lineage cells under control of a 2.3 kb fragment of the col1a1 promoter. Analysis of these mice confirmed that G-CSF treatment reduced the number of osteoblasts 2.9-fold compared to control mice (n=4, p&lt;.01). Consistent with these findings, expression of several osteoblast genes was sharply reduced during G-CSF treatment; alkaline phosphatase, osteocalcin, stromal-derived factor 1, osteoprotegerin, and bone sialoprotein were all decreased (7.9, 19, 3.8, 14.2, and 4.1-fold decreased; n=5–12 each group, p&lt;.05). These findings are consistent with either a true loss of osteoblasts or induction of osteoblast quiescence. To distinguish between these possibilities, the fate of osteoblasts during G-CSF treatment was assessed by in vivo BrdU labeling. At baseline, 34.9 ± .8% of osteoblasts were labeled after 16 days of BrdU administration. In untreated mice, this percentage decreased to 14.8% ± 0.05% after 9 days. However, in mice harvested 4 days after a 5-day course of G-CSF, the percentage of BrdU positive osteoblasts decreased to 6.5% ± 0.9% (n=2 each group, p&lt;.05). These data suggest that G-CSF treatment increases the turnover of osteoblasts rather than inducing their quiescence. Since apoptosis is one potential mechanism of osteoblast turnover, we measured caspase 3 activation in G-CSF-treated mice. No increased apoptosis was observed [percent activated caspase 3 positive cells + SEM: 8.0% ± 1.5% (untreated),10.5% ± 0.5% (day 3), 5.8% ± 1.2% (day 4); n=2–4 each group, p=ns]. The sharp decrease in osteoblast number suggests that HSC activity might be altered by G-CSF treatment. To address this possibility, competitive repopulation assays were performed with HSC harvested from G-CSF or control mice. In mice competitively reconstituted with control HSC, the percentage of donor cells was 33.7% ± 4.7% at 4 months after transplantation. In contrast, in mice receiving G-CSF treated HSC, the percentage of donor cells was only 10.0% ± 2.6% (n=3–4 each group, p&lt;.05). As this loss in repopulation potential may result from decreased HSC number or function, we estimated HSC number by measuring Kit+ lineage− Sca+ CD34− cells. We found no significant difference in this fraction between treated and untreated mice [number KLS CD34− per femur + SEM: 966 ± 229 (untreated) versus 945 ± 183 (treated); n=6–7 each group, p=ns]. These results suggest that the diminished repopulation potential of G-CSF treated bone marrow results from impaired HSC function rather than reduced HSC number. Finally, we hypothesized that the loss of long-term repopulation potential in HSC from G-CSF treated mice may result from the loss of factors in the bone marrow niche that maintain HSC quiescence. We therefore measured BrdU uptake in the KLS fraction of bone marrow cells. Preliminary results show a trend toward increased BrdU uptake in KLS cells from G-CSF treated mice (48.5% ± 9.2%) compared with untreated mice (26.5% ± 1.4%; n=2 each group, p=.14). In summary, these results suggest a model in which G-CSF induces increased osteoblast turnover, disruption of the stem cell microenvironment, and loss of HSC quiescence.


Sign in / Sign up

Export Citation Format

Share Document