Novel Evidence That Hematopoietic Stem/Progenitor Cells (HSPCs) Are Mobilized During Hemolysis in an Erythrocyte Lysis-Derived, Sphingosine-1-Phosphate (S1P)-Dependent manner—the Crucial Involvement of Complement Cascade (CC) Activation and Attenuation of CXCR4 Retention Signaling

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3189-3189
Author(s):  
Kasia Mierzejewska ◽  
Magdalena Kucia ◽  
Janina Ratajczak ◽  
Mariusz Z Ratajczak

Abstract Abstract 3189 Background. Hemolytic syndromes, such as sickle cell anemia and paroxysmal nocturnal hemoglobinuria, are characterized by an increased number of hematopoietic stem/progenitor cells (HSPCs) circulating in peripheral blood (PB). However, the molecular mechanisms responsible for this effect are unclear. In our previous work we have demonstrated that sphingosine-1-phosphate (S1P) released from lysed erythrocytes and activated platelets is a strong chemottractant for bone marrow (BM)-residing HSPCs (Leukemia 2010;24:976–85). Hypothesis. We hypothesized that S1P released from lysed erythrocytes is a major factor responsible for egress of HSPCs from BM into PB in hemolytic syndromes. Experimental approach. To test this hypothesis, normal mice were injected with phenylhydrazine (PHZ), a compound known to induce hemolysis, and we evaluated the number of Sca-1+Kit+Lin– (SKL) HSPCs circulating in PB as well as the number of clonogenic CFU-GM progenitors mobilized into PB. In parallel, we evaluated the blood plasma levels of S1P and stromal derived factor-1 (SDF-1) by sensitive ELISA, the free hemoglobin (Hb) level, as well as complement cascade (CC) activation by measuring the C5b-C9 (membrane attack complex, MAC) level. In some of the experiments, we combined PZH treatment with injection of the CXCR4 antagonist AMD3100. To better assess the role of CC activation, we also performed mobilization in C5-deficient mice, which do not activate the distal part of the CC and thus do not generate C5b-C9/MAC. Results. We found that hemolysis increases the PB plasma level of S1P but does not affect the SDF-1 level. Furthermore, while PHZ-induced hemolysis mobilizes HSPCs into PB with a peak at 6 h after infusion, this mobilization effect is significantly potentiated by administration of AMD3100, which attenuates CXCR4–SDF-1-mediated retention of HSPCs in the BM microenvironment. Of note, PHZ-induced hemolysis together with AMD3100 mobilized twice as many HSPCs as AMD3100 alone. The degree of mobilization of HSPCs correlated with the free Hb level in plasma and activation of the CC (by an increase in MAC level), and, more importantly, mobilization was not seen in C5-deficient mice. Conclusions. We confirmed our previous observation that with the steady-state S1P level, the PB has already established a strong chemotactic gradient for BM-residing HSPCs (Leukemia 2010;24:976–85), which are actively retained in BM niches in a CXCR4–SDF-1-dependent manner. Hemolysis alone, even if it elevates the S1P level in PB significantly, requires two important events i) attenuation of CXCR4–SDF-1 axis-mediated retention in BM niches and ii) simultaneous activation of the CC, which is crucial for induction of permeabilization of the BM–PB barrier. Furthermore, our data also support the notion that the S1P but not the SDF-1 level in PB establishes the critical chemotactic gradient for HSPCs and is responsible for egress of these cells. Based on the observation that C5-deficient mice are poor mobilizers, inactivation of the distal part of the CC should be considered as a therapeutic approach, not only in paroxysmal nocturnal hemoglobinuria but also in other hemolytic syndromes. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2477-2477
Author(s):  
Kasia Mierzejewska ◽  
Ahmed Abdel-Latif ◽  
Gabriela Schneider ◽  
Janina Ratajczak ◽  
Magdalena Kucia ◽  
...  

Abstract Background We have recently reported that hematopoietic stem/progenitor cells (HSPCs) that harbor mutations of the PIG-A gene are preferentially mobilized into peripheral blood (PB) during hemolytic events in paroxysmal nocturnal hemoglobinuria (PNH) patients (Leukemia 2012;26:1722). This effect has been explained by i) an increase in the plasma level of sphingosine-1-phosphate (S1P), which at physiological doses is a major chemoattractant for HSPCs and is released from lysed erythrocytes (Leukemia 2010;24:976), and ii) the fact that PNH-cloned HSPCs, in contrast to normal HSPCs, show defective SDF-1–CXCR4-mediated retention in BM niches. It is known that under steady-state conditions the concentration of S1P in PB is already 25x higher than its concentration in the BM microenvironment and increases additionally during hemolysis. Aim of the study Since erythrocytes are a major source of plasma S1P, we asked whether massive hemolysis of erythrocytes leading to an additional increase in plasma S1P level would trigger mobilization of HSPCs. Furthermore, to shed more mechanistic light on the mobilization of HSPCs in PNH patients and to distinguish the effect of an increase in the S1P chemotactic gradient in PB plasma from the effect of defective retention of HSPCs in the BM microenvironment, we performed mobilization studies in mice exposed to the hemolysis-inducing agent phenhlhydrazine (PHZ) ± blockade of the BM-retaining SDF-1–CXCR4 axis by AMD3100. Experimental approach Normal C57Bl6 mice were injected with i) PHZ to induce hemolysis and S1P release from erythrocytes, ii) AMD3100 to perturb the SDF-1–CXCR4-mediated retention of HSPCs in BM niches, or iii) both PHZ and AMD3100. Subsequently, we evaluated the number of circulating Sca-1+Kit+Lin– (SKL) HSPCs as well as the number of clonogenic CFU-GM progenitors in PB. In parallel, we evaluated the S1P blood plasma levels by liquid chromatography electrospray ionization tandem mass spectrometry (HPLC ESI MS/MS) and the SDF-1 level by ELISA. In addition, we measured complement cascade (CC) activation by measuring the C5b-C9 (membrane attack complex, MAC) levels. Results We found that hemolysis doubles the PB plasma level of S1P (from 1 to 2 mM). To assess the effect of plasma S1P versus plasma SDF-1 as chemoattractants mediating egress of HSPCs from BM, we employed plasma derived from control and PHZ-treated mice, W146, a receptor-specific inhibitor for the S1P receptor type 1 (S1P1), and the CXCR4 antagonist AMD3100 in Transwell migration assays. We observed that chemotaxis of BM-purified HSPCs was inhibited by blocking the S1P–S1P1 but not the SDF-1–CXCR4 axes, which demonstrates that the S1P level in plasma is a crucial chemoattractant for HSPCs present under normal steady-state conditions and in PHZ-treated mouse plasma. This observation also clearly shows that the S1P gradient, even under steady-state conditions, is already high enough to promote egress of HSPCs from BM into PB and supports our previous observations that, while the SDF-1–CXCR4 axis plays an important role in retention of HSPCs in BM niches, the SDF-1 plasma level is too low to induce egress of HSPCs (Leukemia 2010;24:976). In our in vivo mobilization studies, we observed that, in contrast to AMD3100 administration, PHZ-induced hemolysis alone had a negligible effect on mobilization of HSPCs, with a peak at 6 h after infusion of this hemolysis-inducing agent (Figure 1). However, when we combined PHZ with AMD3100, mice mobilized twice as many HSPCs as with administration of AMD3100 alone. The degree of mobilization of HSPCs correlated with the free Hb level in plasma and with activation of the complement cascade. Conclusions At the steady-state conditions S1P level in PB is already a strong chemotactic factros for BM-residing HSPCs. More importantly, to explain the differential mobilization of PNH-affected HSPCs versus normal HSPCs (Leukemia 2012;26:1722), we show here for the first time that hemolysis alone, even if it doubles the S1P level in PB, requires attenuation of CXCR4–SDF-1-mediated retention in BM niches. Thus, PNH-affected HSPCs, due to defective lipid raft formation, have impaired CXCR4-mediated retention in BM niches and are preferentially mobilized into PB. Finally, our data explain why, compared with PNH, HSPCs are mobilized to a much lesser degree in other hemolytic syndromes (e.g., sickle cell anemia). Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3472-3472
Author(s):  
Ali Jalili ◽  
Neeta Shirvaikar ◽  
Chris Korol ◽  
Anna Janowska-Wieczorek

Abstract The complement system, a vital component of the immune system, has been shown to play a role in hematopoietic stem/progenitor cell (HSPC) trafficking. C3a is known to be important in the retention of HSPC in the bone marrow (BM) as C3a-deficient mice are good mobilizers, and C5a is important in the mobilization of HSPC because C5a-deficient mice are poor mobilizers (Stem Cells2007; 25: 3093). Further, granulocyte-colony stimulating factor (G-CSF) activates the complement system via the classical pathway, down-regulates stromal-derived factor (SDF)-1 in BM stromal cells and decreases expression of its receptor, CXCR4, in myeloid cells. In this work investigated the mechanism of C5a involvement in HSPC mobilization. Using RT-PCR and FACS we examined the expression of the C5a receptor (CD88) on mobilized peripheral blood (PB) and steady-state PB HSPC and mature white blood cells, and in vitro-expanded myeloid, erythroid, and megakaryocytic progenitor cells. We found that CD88, like the G-CSF receptor, is not expressed on BM, PB or cord blood HSPC (CD34+ cells); during CD34+ cell differentiation the expression of CD88 increases in myelocytic and megakaryocytic progenitors; and the percentage of monocytes and polymorphonuclear (PMN) cells expressing CD88 is significantly higher in mobilized than in steady-state PB. Examing the function of C5a (using flow cytometry) we found that, unlike C3a, C5a decreases CXCR4 expression in a dose-dependent manner in monocytes and PMN, but not in lymphocytes; and this effect was not seen when anti-C5a antibody was added. Interestingly, we found that G-CSF down-regulation of CXCR4 expression on PMN was partially restored by anti-C5a antibody, suggesting that the mobilizing effects of G-CSF are at least in part due to the action of C5a. Moreover, chemotaxis of PMN towards SDF-1 (chemotaxis assay) increased when these cells were stimulated with C3a but decreased with both C5a and G-CSF, reflecting the CXCR4 expression status of these cells after stimulation. We also examined the effect of C5a on matrix metalloproteinase (MMP) secretion in BM leukocytes and found that, like G-CSF, C5a increased MMP-9 and MMP-2 secretion into media (zymography). Since the SDF-1/CXCR4 axis plays an integral role in the retention of HSPC in the BM, we conclude that C5a promotes mobilization by disrupting this axis, as well as increasing MMP-9 and MMP-2 secretion by BM leukocytes, thereby allowing egress of HSPC into the PB.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 367-367
Author(s):  
Marcin Wysoczynski ◽  
HakMo Lee ◽  
Rui Liu ◽  
Wan Wu ◽  
Janina Ratajczak, ◽  
...  

Abstract Abstract 367 We reported that complement cascade (CC) becomes activated in bone marrow (BM) during mobilization of hematopoietic stem/progenitor cells (HSPCs) by immunoglobulin (Ig)-dependent pathway and/or by alternative Ig-independent pathway as seen during G-CSF- or Zymosan mobilization, respectively. As a result, several potent bioactive CC anaphylatoxins (C3 and C5 cleavage fragments) are released that regulate egress of HSPCs (Blood 2003;101,3784; Blood 2004;103,2071; Blood 2005;105,40, Leukemia 2009; in press.). This explains why: i) NOD/SCID and RAG-/- animals that do not activate the Ig-dependent CC classical pathway; ii) C2fB-/- and C3-/- mice that do not activate the classical and alternative CC pathways; and iii) C5-/- mice that do not activate the distal pathway of CC are all poor G-CSF- and/or Zymosan mobilizers. In this study, we evaluated the role of CC in mobilization induced by CXCR4 antagonist AMD3100. We noticed that all CC activation-deficient mice mentioned above, except C5-/- mice, mobilize normally in response to AMD3100 administration. Accordingly, the number of mobilized CD34- SKL cells, leucocytes, and CFU-GM clonogeneic progenitors in mutant mice was similar to wt littermates. More important we observed that AMD3100 mobilization of HSPCs was preceded by a massive egress of leucocytes from BM and that AMD3100 was able to stimulate in these cells i) phosphorylation of MAPKp42/44 and ii) secretion of MMP-9. At the same time, ELISA data to detect CC activation revealed that serum levels of CC cleavage fragments, which were low in the initial phase of AMD3100 mobilization during granulocyte egress, become elevated later during HSPC egress. Thus, our data show that despite a fact that G-CSF and AMD3100 mobilize HSPCs by involving different mechanisms, activation of CC is a common phenomenon occurring during mobilization induced by both compounds. This further supports a pivotal role of CC activation in the egress of HSPCs from BM; however, both compounds activate CC differently. While G-CSF administration initiates CC activation at its proximal C1q-C3 level, AMD3100 induces CC activation at the distal C5 level, pointing to a crucial role of C5 cleavage in executing mobilization. To support this, all mice employed in our studies that display defects in activation of proximal stages of CC (NOD/SCID, RAG, C2fB-/-, and C3-/-) are normal AMD3100 mobilizers. However, C5 is cleavage required for mobilization occurs in the plasma of these animals latter on - directly by proteases released from AMD3100-stimulated granulocytes that egress from the BM as a first wave of mobilized cells. This compensatory mechanism cannot occur from obvious reasons in C5-/- mice. We conclude that AMD3100-directed mobilization similarly as G-CSF-induced one depends on activation of CC; however, AMD3100 in contrast to G-CSF activates CC at distal stages – directly by proteases released from mobilized/activated granulocytes. Cleavage of C5 and release of C5a and desArgC5a create a sinusoid-permissive environment in BM for HSPCs egress. This suggests involvement of both C5 cleavage fragments as well as a potential role of downstream elements of CC activation - membrane attack complex - MAC (C5b-C9) in stem cell mobilization. Therefore, some poor AMD3100 patient responders could possess a defect in activation of the distal steps of CC. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1497-1497 ◽  
Author(s):  
Chun Shik Park ◽  
Takeshi Yamada ◽  
H. Daniel Lacorazza

Abstract Abstract 1497 Poster Board I-520 KLF4 is a tumor suppressor in the gastrointestinal tract known to induce cell cycle arrest in a cell context dependent manner. We recently reported that KLF4 maintains quiescence of T lymphocytes downstream of T-cell receptor signaling (Yamada et al., Nature Immunology, 2009). The role of KLF4 in reprogramming adult somatic cells into pluripotent stem cells along with Oct3/4, c-Myc and Sox2 suggests that KLF4 restricts proliferation of undifferentiated cells. In spite of a redundant role of KLF4 in fetal liver hematopoietic stem cells (HSC), its role in the maintenance of adult bone marrow HSCs has not been studied yet. To study the role of KLF4 in the hematopoietic system we used gain- and loss-of-function mouse models. Retroviral transfer of KLF4 into wild type bone marrow (BM) cells led to significant reduction of colony forming units (CFU) in methylcellulose cultures due to increased apoptosis and lower proliferation. Then, Mx1-Cre was used to induce deletion of Klf4-floxed mice by polyI:C administration. Analysis of peripheral blood cells up to 6-9 months post polyI:C administration showed significant reduction of monocytes, as previously reported, and expansion of CD8+CD44+ T cells due to their increased proliferative potential. BM cells from Klf4-deficient mice exhibited increased number of myeloid progenitor cells measured by flow cytometry (Lin-Sca-1-c-kit+FcRII/III+CD34+ cells), CFU and CFU-S8. Cytoablation with 5-fluorouracil (5-FU) showed lower nadir of peripheral white blood cells in Klf4-deficient mice compared to control mice. In spite of normal multilineage reconstitution in BM transplants experiments, competitive reconstitution with Klf4-deficient and normal BM cells resulted in reduced contribution of Klf4-deficient cells to peripheral blood, likely due to homing and proliferative differences. Collectively, our data shows that KLF4 has an important role in function of hematopoietic stem and progenitor cells. Disclosures: No relevant conflicts of interest to declare.


Leukemia ◽  
2021 ◽  
Author(s):  
Arjun Thapa ◽  
Mateusz Adamiak ◽  
Kamila Bujko ◽  
Janina Ratajczak ◽  
Ahmed K. Abdel-Latif ◽  
...  

AbstractLike their homing after transplantation to bone marrow (BM), the mobilization of hematopoietic stem/progenitor cells (HSPCs) is still not fully understood, and several overlapping pathways are involved. Several years ago our group proposed that sterile inflammation in the BM microenvironment induced by pro-mobilizing agents is a driving force in this process. In favor of our proposal, both complement cascade (ComC)-deficient and Nlrp3 inflammasome-deficient mice are poor G-CSF and AMD3100 mobilizers. It is also known that the Nlrp3 inflammasome mediates its effects by activating caspase-1, which is responsible for proteolytic activation of interleukin-1β (IL-1β) and interleukin-18 (IL-18) and their release from cells along with several danger-associated molecular pattern molecules (DAMPs). We observed in the past that IL-1β and IL-18 independently promote mobilization of HSPCs. In the current work we demonstrated that caspase-1-KO mice are poor mobilizers, and, to our surprise, administration of IL-1β or IL-18, as in the case of Nlrp3-KO animals, does not correct this defect. Moreover, neither Caspase-1-KO nor Nlrp3-KO mice properly activated the ComC to execute the mobilization process. Interestingly, mobilization in these animals and activation of the ComC were both restored after injection of the DAMP cocktail eATP+HGMB1+S100A9, the components of which are normally released from cells in an Nlrp3 inflammasome–caspase-1-dependent manner. In addition, we report that caspase-1-deficient HSPCs show a decrease in migration in response to BM homing factors and engraft more poorly after transplantation. These results for the first time identify caspase-1 as an orchestrator of HSPC trafficking.


Leukemia ◽  
2019 ◽  
Vol 33 (12) ◽  
pp. 2967-2970 ◽  
Author(s):  
Mateusz Adamiak ◽  
Anna M. Lenkiewicz ◽  
Monika Cymer ◽  
Magda Kucia ◽  
Janina Ratajczak ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 31-31 ◽  
Author(s):  
HakMo Lee ◽  
Marcin Wysoczynski ◽  
Wan Wu ◽  
Rui Liu ◽  
Magdalena Kucia ◽  
...  

Abstract Abstract 31 We reported that complement cascade (CC) is activated in bone marrow (BM) during mobilization of hematopoietic stem/progenitor cells (HSPCs) and that CC clevage fragments direct egress of HSPCs from BM into peripheral blood (PB) (Blood 2003;101,3784; Blood 2004;103,2071; Blood 2005;105,40). We also reported that C5 cleavage fragments play a crucial role in the mobilization process by: i) inducing proteolytic activity in the BM environment; ii) directing BM egress of granulocytes that “pave a road” for HSPCs; and iii) inducing secretion of cationic peptides from activated granulocytes that prime HSPC egress (Leukemia 2009; in press). In this study, we sought to determine which major chemottractant is present in PB that is responsible for egress of HSPCs and whether activation of CC plays some role in its level/expression. We noticed that plasma derived from normal and mobilized PB strongly chemoattracts murine and human HSPCs. This chemotactic effect was not dependent on plasma SDF-1 levels because: i) it occurs unaffectedly in the presence of CXCR4 antagonist AMD3100; ii) it was still robust to heat-inactivated sera; and iii) ELISA studies revealed negligible concentrations of SDF-1, which did not correlate with good or poor mobilizer status. However, to our surprise, we noticed that plasma isolated from G-CSF-mobilized mice and patients contains traces of free hemoglobin, which suggests some level of hemolysis occurs in mobilized PB. As such, we performed chemotactic assays in the presence of different concentrations of lysed erythrocytes and noticed that such diluted lysates are potent chemoattractants for HSPCs. The chemotactic activities of these lysates were resistant to heat inactivation similarly as patient sera. Based on this, we focused on S1P, a thermo-resistant lipid that, as reported, chemoattracts HSPCs and whose major reservoirs are erythrocytes (FASEB J 2007:21;1202). In fact we found by ELISA that S1P level increases during mobilization in PB and that SP1 is the most potent chemoattractant for BM-residing HSPCs, much stronger than SDF-1 - if both compounds are compared in physiologically relevant concentrations. Furthermore, activation of S1P receptors on BM-derived HSPCs augmented responsiveness to SDF-1 gradient up to 50%. However, these chemotactic effects of S1P were not visible for previously mobilized PB or umbilical cord blood HSPCs, which we explain by a fact that these mobilized cells are already desensitized to S1P gradient. Therefore, we propose the following scenario. First, a mobilizing agent (e.g., G-CSF) induces activation of CC in BM that subsequently contributes to the release of protelolytic enzymes from granulocytes that perturb SDF-1-CXCR4/VLA-4-VCAM1 interactions and stimulate egress of activated granulocytes from BM that “pave a road” for egress of HSPCs. Simultaneously, the final product of CC activation (C5b-C9), the membrane attack complex (MAC), induces in BM sinusoids the release of S1P from erythrocytes. S1P accumulating in BM sinusoids and cationic peptides released from activated granulocytes, but not changes in plasma SDF-1 levels, are crucial executors of HSPCs egress from BM into PB. Thus, our results provide novel evidence that CC activation/membrane attack complex (MAC)-induced elevated plasma S1P level is essential for egress/mobilization of HSPCs. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document