scholarly journals Annexin II expressed by osteoblasts and endothelial cells regulates stem cell adhesion, homing, and engraftment following transplantation

Blood ◽  
2007 ◽  
Vol 110 (1) ◽  
pp. 82-90 ◽  
Author(s):  
Younghun Jung ◽  
Jingcheng Wang ◽  
Junhui Song ◽  
Yusuke Shiozawa ◽  
Jianhua Wang ◽  
...  

Differentiation of hematopoietic stem cells (HSCs) after birth is largely restricted to the bone marrow cavity, where HSCs are associated closely with osteoblasts (OBs). How OBs localize HSCs to the endosteal niche remains unclear. To explore adhesive interactions between HSCs and OBs, a cell blot analysis was used that revealed 2 major bands that corresponded to monomers and multimers of annexin II (Anxa2). Immunohistochemistry revealed that OBs and marrow endothelial cells express Anxa2 at high levels. Function-blocking studies confirmed that Anxa2 mediates HSC adhesion mainly via the N-terminal portion of the Anxa2 peptide. Adhesion of HSCs to OBs derived from Anxa2-deficient animals (Anxa2−/−) was significantly impaired compared with OBs obtained from wild-type animals (Anxa2+/+). Moreover, fewer HSCs were found in the marrow of Anxa2−/− versus Anxa2+/+ animals. Short-term lodging, engraftment, and survival of irradiated mice with whole marrow cells were substantially inhibited by N-terminal peptide fragments of Anxa2 or anti-Anxa2 antibodies. Similar findings were noted in long-term competitive repopulation studies. Collectively, these findings reveal that Anxa2 regulates HSC homing and binding to the bone marrow microenvironment and suggest that Anxa2 is crucial for determining the bone marrow niche of HSCs.

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1489-1489
Author(s):  
Monika Dolinska ◽  
Pingnan Xiao ◽  
Anne-Sofie Johansson ◽  
Lakshmi Sandhow ◽  
Makoto Kondo ◽  
...  

Abstract Myelodysplastic Syndrome with Ring Sideroblast (MDS-RS), a clonal hematopoietic cell neoplasm, is low risk MDS, characterized by anemia, hyperplastic ineffective erythropoiesis and marrow ring sideroblasts. Mouse studies have shown that bone marrow niche, including endothelial cells, osteoblasts, adipocytes and mesenchymal stem cells (MSCs), contribute to progression of various hematological disorders. However, in vivo contribution of the different bone marrow stromal cells to the progression of MDS-RS in patients remains largely unknown. To investigate this, we have phenotypically, molecularly and functionally characterized the BM native stromal cell subsets including MSCs freshly isolated by multi-color fluorescence activated cell sorting (FACS) from bone marrow of MDS-RS patients and age-matched healthy donors. We found: 1) the MDS-RS MSCs, estimated by colony forming unit-fibroblast (CFU-F), shared similar immunophenotype with normal MSCs (CD45-CD235a-CD31-CD44-, most of which were CD271+CD146+CD106+); 2) the frequency of CFU-Fs was significantly increased in the phenotypically defined MSCs of MDS-RS bone marrow compared to that of age-matched healthy controls (p=0.005); 3) multi-lineage differentiation assay revealed impaired osteogenic differentiation potential, but enhanced adipogenic differentiation potential of MDS-RS MSCs; 4) FACS analysis showed increased frequency of the adhesion receptor integrin α4 (ITGA4) in the CD44- MSCs from MDS-RS bone marrow (p=0.013); 5) Correspondingly, RNA-sequencing of the freshly isolated bone marrow MSCs and endothelial cells revealed altered gene expression profile of these cells in MDS-RS patients. Among those, ITGA4, ITGA11, ITGAE and ITGB1 are upregulated in the MDS-RS MSCs, indicating potential abnormal adhesive interaction of the MSCs with hematopoietic stem cells in the patients. In addition, the cell cycling gene KI67 is upregulated whereas cell cycle negative regulators, like CDKN1A and CDKN1C are downregulated in the MDS-RS MSCs, which is consistent with their increased CFU-F activity. Interestingly, we detected abnormal expression of hematopoietic growth factors such as downregulation of ANGPTL4 in the MDS-RS MSCs and upregulation of ANGPT1 in the MDS-RS endothelial cells. The functional relationship between the stromal cell alterations and the abnormal hematopoiesis as well as the underlying molecular mechanisms are currently under investigation. Taken together, our data provide new evidence for phenotypic, functional and molecular alterations of bone marrow mesenchymal cells in MDS-RS patients. The molecular pathways mediating bone marrow niche alteration could be potential therapeutic targets for new treatment of MDS-RS. Disclosures No relevant conflicts of interest to declare.


2017 ◽  
Vol 214 (7) ◽  
pp. 2005-2021 ◽  
Author(s):  
Adam J. Mead ◽  
Wen Hao Neo ◽  
Nikolaos Barkas ◽  
Sahoko Matsuoka ◽  
Alice Giustacchini ◽  
...  

Although previous studies suggested that the expression of FMS-like tyrosine kinase 3 (Flt3) initiates downstream of mouse hematopoietic stem cells (HSCs), FLT3 internal tandem duplications (FLT3 ITDs) have recently been suggested to intrinsically suppress HSCs. Herein, single-cell interrogation found Flt3 mRNA expression to be absent in the large majority of phenotypic HSCs, with a strong negative correlation between Flt3 and HSC-associated gene expression. Flt3-ITD knock-in mice showed reduced numbers of phenotypic HSCs, with an even more severe loss of long-term repopulating HSCs, likely reflecting the presence of non-HSCs within the phenotypic HSC compartment. Competitive transplantation experiments established that Flt3-ITD compromises HSCs through an extrinsically mediated mechanism of disrupting HSC-supporting bone marrow stromal cells, with reduced numbers of endothelial and mesenchymal stromal cells showing increased inflammation-associated gene expression. Tumor necrosis factor (TNF), a cell-extrinsic potent negative regulator of HSCs, was overexpressed in bone marrow niche cells from FLT3-ITD mice, and anti-TNF treatment partially rescued the HSC phenotype. These findings, which establish that Flt3-ITD–driven myeloproliferation results in cell-extrinsic suppression of the normal HSC reservoir, are of relevance for several aspects of acute myeloid leukemia biology.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 15-16
Author(s):  
Alexandra A Soukup ◽  
Daniel R Matson ◽  
Kirby D Johnson ◽  
Emery H Bresnick

The transcription factor GATA2 is essential for the generation and function of hematopoietic stem and progenitor cells (HSPCs), erythroid precursors, and endothelial cells. A conserved intronic GATA2 enhancer, 9.8 kb downstream of the transcriptional start site (+9.5 in the mouse), is mutated in patients with GATA2 deficiency syndrome. Patient mutations within this region include a c.1017+512del28 deletion, removing E-box and GATA motifs, c.1017+532T>A that disrupts the E-box, and, most frequently, C>T in a 3' Ets motif (c.1017+572C>T) (Soukup and Bresnick, 2020). Homozygous mutation of the Ets motif in mice allows normal developmental and steady-state hematopoiesis but impairs hematopoietic regeneration (Soukup et al., 2019). In addition to HSPCs, GATA2 is expressed in non-hematopoietic cells in the bone marrow niche, e.g. endothelial cells and neurons (Katsumura et al., 2017). As the +9.5(Ets) mutation is not hematopoietic cell-specific, we asked whether regenerative defects of +9.5(Ets)-/- mice reflect disruption of cell-intrinsic or -extrinsic activities. In a competitive transplant assay, +9.5(Ets)-/- HSPCs were 3-fold less effective at long-term reconstitution than WT, and mechanistic studies indicated that the motif functions in hematopoietic cells to promote regeneration (Soukup et al., 2019). We conducted a reciprocal transplant of WT HSPCs into irradiated WT or +9.5(Ets)-/- recipients and quantified reconstitution by peripheral blood counts 4, 8, 12, and 16 weeks post-transplant. This analysis revealed no significant differences between WT and mutant recipients. At week 16, donor-derived leukocytes were 92% (+9.5(Ets)-/- recipients) and 96% (WT recipients) of total; the contribution did not differ significantly at any time. After 16 weeks, animals were sacrificed and HSPCs analyzed, confirming no significant alterations in mutant recipients. These results rigorously establish the mutant microenvironment as competent to support WT HSPC functions, emphasizing the critical hematopoietic cell-intrinsic activity of the +9.5 Ets motif. As the +9.5 Ets motif promotes regenerative hematopoiesis, and the +9.5 E-box;GATA is essential for developmental hematopoiesis, we devised a strategy to leverage these activities to innovate new models for GATA2 function in adult HSPCs. We generated compound heterozygous (CH) mice containing a mutant E-box;GATA sequence on one allele and a mutant Ets motif on the other allele. CH mice survived past weaning, with adults exhibiting significant steady-state defects, including a 4.4-fold decrease in GATA2hi megakaryocytes (p < 0.0001) and 20% decrease (p = 0.02) in platelets. To test whether the CH mutations compromise regeneration, we quantified HSPC populations in bone marrow from mice treated with vehicle or 5-fluorouracil (FU) 9- and 10- days post treatment. Steady-state HSC (Lin−Sca1+Kit+CD48-CD150+) levels were unaltered in CH animals. Days 9 and 10 post-FU treatment, WT HSC levels increased 17- (p = 0.0006) and 18-fold (p = 0.0007) relative to vehicle-treated animals. CH HSCs did not expand and were <10% of the steady-state level. 7 days post-FU treatment, Gata2 expression increased 1.9-fold in WT HSCs (p = 0.029); this response was abrogated in CHs. We asked if CH HSCs were capable of reconstitution in a competitive transplant assay. Four weeks post-transplant, CH progeny were 40-fold lower than WT (p < 0.0001). At 8-, 12-, and 16-weeks post-transplant, CH contribution was reduced 90-, 266-, and 280-fold, respectively. Defects persisted upon secondary transplantation, demonstrating that the defects cannot be restored by passage through a WT microenvironment. Thus, CH and +9.5(Ets)-/- mice share phenotypes, but CH mutations more severely impair regeneration and long-term reconstituting activity. This supports a paradigm in which the Ets motif and additional +9.5 sequences are critical for regeneration. This study revealed molecular determinants for steady-state and regenerative enhancer functions to enable discovery of +9.5-like enhancers with common operating mechanisms. We predict that such enhancers reside at a GATA2-regulated gene cohort, including genes that will reveal new mechanisms in hematopoiesis. As CH mice are poised for hematopoietic collapse, but can be propagated as relatively normal adults, studies are underway with this unique model to identify triggers of bone marrow failure and leukemogenesis. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 4 (1) ◽  
Author(s):  
José Gabriel Barcia Durán ◽  
Tyler Lu ◽  
Sean Houghton ◽  
Fuqiang Geng ◽  
Ryan Schreiner ◽  
...  

AbstractJak3 is the only non-promiscuous member of the Jak family of secondary messengers. Studies to date have focused on understanding and targeting the cell-autonomous role of Jak3 in immunity, while functional Jak3 expression outside the hematopoietic system remains largely unreported. We show that Jak3 is expressed in endothelial cells across hematopoietic and non-hematopoietic organs, with heightened expression in the bone marrow. The bone marrow niche is understood as a network of different cell types that regulate hematopoietic function. We show that the Jak3–/– bone marrow niche is deleterious for the maintenance of long-term repopulating hematopoietic stem cells (LT-HSCs) and that JAK3-overexpressing endothelial cells have increased potential to expand LT-HSCs in vitro. This work may serve to identify a novel function for a highly specific tyrosine kinase in the bone marrow vascular niche and to further characterize the LT-HSC function of sinusoidal endothelium.


Blood ◽  
2020 ◽  
Author(s):  
Yue Sheng ◽  
Rui Ma ◽  
Chunjie Yu ◽  
Qiong Wu ◽  
Steven Zhang ◽  
...  

Here we report a dosage effect of c-Myc on hematopoiesis and its distinct role in mediating the Wnt/b-catenin pathway in hematopoietic stem cell (HSC) and bone marrow niche cells. We showed that c-Myc haploinsufficiency led to ineffective hematopoiesis by inhibiting HSC self-renewal and quiescence, and promoting its apoptosis. We have identified Nr4a1, Nr4a2 and Jmjd3, which are critical for the maintenance of HSC functions, as previously unrecognized downstream targets of c-Myc in HSCs. c-Myc directly binds to the promoter regions of Nr4a1, Nr4a2 and Jmjd3 and regulates their expression. Our results revealed that Nr4a1 and Nr4a2 mediated the function of c-Myc in regulating HSC quiescence while all three genes contribute to the function of c-Myc in the maintenance of HSC survival. Adenomatous polyposis coli (Apc) is a negative regulator of the Wnt/b-catenin pathway. We have provided the first evidence that Apc haploinsufficiency induced a blockage of erythroid lineage differentiation through promoting expression and secretion of IL6 in bone marrow endothelial cells. We found that c-Myc haploinsufficiency failed to rescue defective function of Apc-deficient HSCs in vivo but it was sufficient to prevent the development of severe anemia in Apc heterozygous mice, and to significantly prolong survival of such mice. Furthermore, we showed that c-Myc mediated Apc loss-induced IL6 secretion in endothelial cells, and c-Myc haploinsufficiency reversed the negative effect of Apc deficient endothelial cells on erythroid cell differentiation. Our studies indicate that c-Myc has a context-dependent role in mediating the function of Apc in hematopoiesis.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 874-874
Author(s):  
Katie A Matatall ◽  
Hamilton Wang ◽  
Roman Jaksik ◽  
Marek Kimmel ◽  
Dongsu Park ◽  
...  

Abstract Prolonged exposure to proinflammatory conditions, such as during chronic infection, can lead to the development of bone marrow failure syndromes including aplastic anemia. Indeed, we have previously shown that chronic infection activates hematopoietic stem cells (HSCs), ultimately leading to bone marrow suppression due to an IFNgamma (IFNγ)-dependent depletion of HSCs. Defining the mechanisms behind inflammation mediated HSC loss may provide important insight into how inflammation affects hematopoiesis in many clinical conditions including infection, ageing, and myelodysplastic syndrome. HSC quiescence is maintained in large part through interactions with the surrounding cells of the bone marrow niche. In particular, HSCs closely colocalize with perivascular stromal cells that secrete high levels of the chemokine Cxcl12 and other hematopoietic maintenance factors such as angiopoietin, IL7, and stem cell factor. We hypothesized that IFNγ-induced activation may affect the positioning of HSCs relative to supporting cells within the bone marrow microenvironment. We used 3-D intravital imaging to assess the localization of HSCs relative to Cxcl12-abundant reticulocytes (CAR cells). When CMTMR-stained HSCs were transplanted into Cxcl12-GFP mice we found that IFNγ treatment led to a distancing of HSCs away from quiescence-enforcing CAR cells. In contrast, TNF-alpha treatment, which does not induce HSC proliferation at the concentration used, resulted in no such relocalization. Furthermore, no displacement occurred in response to IFNγ treatment when Ifngr1-deficient HSCs were transplanted, suggesting a cell-autonomous mechanism of relocalization. RNA expression analysis and chemotaxis assays showed that IFNγ-treated HSCs are not impaired in their ability to respond to Cxcl12 signaling, indicating that a change in Cxcl12-Cxcr4 interactions does not account for the movement of HSCs away from CAR cells. To further assess HSC-specific changes that may account for IFNγ-dependent relocalization, we performed gene expression analysis of HSCs from control or IFNγ-treated mice. We saw no change in the expression of common HSCs receptors that are thought to play a role in maintaining HSC quiescence, such as cKit, Cdh2, Mpl, Itgb1, Itbg2, Itga4, and Itga1. However, the surface protein, Bst2, also known as tetherin, was significantly upregulated in HSCs upon IFNγ stimulation. This was confirmed at both the RNA and protein level. A prior report identified Bst2 as a noncanonical E-selectin ligand. This finding is particularly interesting given that HSCs found in close proximity to E-selectin-expressing endothelial cells exist in an activated, proliferative state. Furthermore, the interaction between HSCs and E-selectin+ endothelial cells was previously noted to be independent of canonical E-selectin ligands such as Psgl1, CD44, or the Lewis family antigens. Thus we hypothesized that IFNγ-induced upregulation of surface Bst2 may facilitate proliferation by promoting HSC binding to E-selectin. Indeed, in vitro E-selectin binding assays showed that IFNγ treatment increased binding of progenitor cells to E-selectin, but not to the closely related adhesin, P-selectin. Inflammation plays an important role in the development of many cancers including acute myeloid leukemia (AML), leading us to investigate if Bst2 upregulation is an important factor in AML biology. Indeed, we found that increased levels of Bst2 are associated with poor survival in AML patients. Further, murine studies have shown that leukemic blasts have greatly increased E-selectin binding capacity following transformation. Using two independent AML cell lines, we found that IFNγ treatment upregulates Bst2 expression and increases E-selectin binding capacity. These findings suggest a mechanism by which inflammation could drive cell proliferation in AML. In summary, we have uncovered a potential new mechanism for IFNγ-induced HSC activation, whereby HSCs are relocalized in a Bst2-dependent manner from the quiescence-enforcing CAR niche to an active E-selectin-positive niche. Furthermore, we demonstrate that IFNγ-mediated Bst2 expression also occurs in AML, suggesting a mechanistic link between inflammation and AML progression. This study may open up new potential therapeutic avenues for the treatment of patients with chronic infection, inflammatory diseases, and cancer. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 174-174
Author(s):  
Cindy L Hochstetler ◽  
Yuxin Feng ◽  
Yi Zheng

Abstract The bone marrow niche is an important milieu where hematopoietic stem and progenitor cells (HSPCs) are maintained to ensure their lifelong contribution to hematopoiesis. Recent evidence has highlighted the critical importance of the perivascular bone marrow (BM) niche as the key host and regulator of HSPCs. Bone marrow endothelial cells (BMECs) are major components of the vascular niche. While studies have shown that an alteration in a component of the niche can affect hematopoiesis and promote the development of myeloproliferative disorders/myelodysplastic syndromes, it remains unclear how altered BMECs can impact hematopoiesis. To this end, we have generated a Tamoxifen (TAM)-inducible Tie2-CreER/LSL-KRasG12D;tdTomato mouse model to introduce an oncogenic KRas mutation specifically in adult endothelial cells. The tdTomato reporter overlaps with the CD31 and vascular endothelial growth factor receptor 2 (VEGFR2) endothelial cell markers and shows no detectable leakage into the adult hematopoietic compartment. To evaluate changes in hematopoiesis, we performed complete blood counts at 12 weeks post TAM injection and found that the Tie2-CreER/LSL-KRasG12D mice (KRasG12D mice) had significantly more leukocytes (p=0.031) and neutrophils (p=0.002) than controls. Flow cytometry analysis confirmed that the KRasG12D mice had a significantly higher percentage of myeloid cells with concurrent decrease in lymphocyte percentage in the peripheral blood (p=0.016). At 16 weeks post TAM injection, a significant decrease in B cells could also be noted in the blood of KRasG12D mice (p=0.028). Compared to controls, the KRasG12D mice displayed splenomegaly (p=0.025) and their spleens had a higher percentage of myeloid cells (p=0.002). There was an increase in the common myeloid progenitor compartment in the spleen and a significant increase in the granulocyte macrophage progenitor compartment (p=0.014) of KRasG12D mice. These mice also had an increase in the short-term hematopoietic stem cell (ST-HSC) compartment both in the BM and spleen. Colony forming assays revealed that KRasG12D mice had a higher number of total colonies formed from BM (p=0.044), spleen (p=0.007) and blood cells (p=0.56). Genotyping PCR showed no KRasG12D activation in hematopoietic cells, confirming that the observed phenotypes were due to an effect in BMECs. To complement our native inducible mouse model, we transplanted BM cells from syngeneic BoyJ mice into lethally irradiated Tie2-CreER;KRasG12D or KRasWT recipients. The endothelial KRasG12D recipientsdied between 75-200 days post transplantation (p=0.0079) while the KRasWT recipients remained alive. The KRasG12D recipients also displayed splenomegaly (p=0.004). Competitive transplant studies with donor cells from KRasG12D or KRasWT mice with competitor cells from syngeneic mice (CD45.1) showed that BM cells from the KRasG12D mice (CD45.2) outcompeted cells from KRasWT mice with a significantly higher percentage of CD45.2 donor chimerism in all blood lineages examined. To uncover any molecular events underpinning these hematopoietic changes, we performed quantitative real-time polymerase chain reaction. Our preliminary experiments from total BM RNA of KRasG12D or KRasWT mice indicate that there is a significant increase in VEGFα and a decrease in transforming growth factor β in KRasG12D mice, accompanying the above noted increase in the ST-HSC population. Collectively, our data provide strong evidence that an abnormal vascular niche caused by oncogenic insults in BMECs can disrupt normal hematopoiesis and promote a myeloproliferative phenotype, thereby implicating abnormal BMECs as novel contributors to blood pathogenesis. Studies are underway to further assess the molecular contributions from the disrupted vascular niche and the resulting HSPCs. Uncovering the mechanism of how altered BMECs can remodel hematopoiesis holds the exciting promise of better therapeutic strategies. Disclosures No relevant conflicts of interest to declare.


Cancers ◽  
2020 ◽  
Vol 13 (1) ◽  
pp. 68
Author(s):  
Fulvio Massaro ◽  
Florent Corrillon ◽  
Basile Stamatopoulos ◽  
Nathalie Meuleman ◽  
Laurence Lagneaux ◽  
...  

Aging of bone marrow is a complex process that is involved in the development of many diseases, including hematologic cancers. The results obtained in this field of research, year after year, underline the important role of cross-talk between hematopoietic stem cells and their close environment. In bone marrow, mesenchymal stromal cells (MSCs) are a major player in cell-to-cell communication, presenting a wide range of functionalities, sometimes opposite, depending on the environmental conditions. Although these cells are actively studied for their therapeutic properties, their role in tumor progression remains unclear. One of the reasons for this is that the aging of MSCs has a direct impact on their behavior and on hematopoiesis. In addition, tumor progression is accompanied by dynamic remodeling of the bone marrow niche that may interfere with MSC functions. The present review presents the main features of MSC senescence in bone marrow and their implications in hematologic cancer progression.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Marie-Theresa Weickert ◽  
Judith S. Hecker ◽  
Michèle C. Buck ◽  
Christina Schreck ◽  
Jennifer Rivière ◽  
...  

AbstractMyelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) are clonal hematopoietic stem cell disorders with a poor prognosis, especially for elderly patients. Increasing evidence suggests that alterations in the non-hematopoietic microenvironment (bone marrow niche) can contribute to or initiate malignant transformation and promote disease progression. One of the key components of the bone marrow (BM) niche are BM stromal cells (BMSC) that give rise to osteoblasts and adipocytes. It has been shown that the balance between these two cell types plays an important role in the regulation of hematopoiesis. However, data on the number of BMSC and the regulation of their differentiation balance in the context of hematopoietic malignancies is scarce. We established a stringent flow cytometric protocol for the prospective isolation of a CD73+ CD105+ CD271+ BMSC subpopulation from uncultivated cryopreserved BM of MDS and AML patients as well as age-matched healthy donors. BMSC from MDS and AML patients showed a strongly reduced frequency of CFU-F (colony forming unit-fibroblast). Moreover, we found an altered phenotype and reduced replating efficiency upon passaging of BMSC from MDS and AML samples. Expression analysis of genes involved in adipo- and osteogenic differentiation as well as Wnt- and Notch-signalling pathways showed significantly reduced levels of DLK1, an early adipogenic cell fate inhibitor in MDS and AML BMSC. Matching this observation, functional analysis showed significantly increased in vitro adipogenic differentiation potential in BMSC from MDS and AML patients. Overall, our data show BMSC with a reduced CFU-F capacity, and an altered molecular and functional profile from MDS and AML patients in culture, indicating an increased adipogenic lineage potential that is likely to provide a disease-promoting microenvironment.


Sign in / Sign up

Export Citation Format

Share Document