scholarly journals Modulation of Purinergic Signaling Prior to Transplantation Both in Hematopoietic Stem Progenitor Cells (HSPCs) or Recipient Bone Marrow (BM) Microenvironment - As Novel Strategies to Accelerate and Facilitate HSPCs Homing and Engraftment

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3809-3809
Author(s):  
Mateusz Adamiak ◽  
Arjun Thapa ◽  
Kamila Bujko ◽  
Katarzyna Brzeźniakiewicz-Janus ◽  
Janina Ratajczak ◽  
...  

Abstract Background. The success rate of hematopoietic stem cell transplantation strongly depends on the number of transplanted hematopoietic stem/progenitor cells (HSPCs) and their speed of engraftment after infusion to the myeloablated transplant recipient. Therefore, clinical outcomes will benefit from accelerating the speed of homing and engraftment rate of transplanted HSPCs. This is important when the number of available HSPCs is low, as seen after poor harvest from BM, poor mobilization efficiency of the donor, and a low number of HSPCs present in the available umbilical cord blood (UCB) unit for an adult recipient. Our recent research demonstrated that purinergic signaling involving extracellular adenosine triphosphate (eATP) and its extracellular metabolite adenosine (eAdo) play a significant opposite role in homing/engraftment of HSPCs - reviewed in Curr Opin Hematol 2021, 28:251-261. To explain this eATP released from the cells of conditioned for transplantation by myeloablation recipient's BM facilitates homing of HSPCs, and subsequently becomes metabolized by cell surface ectonucleotidases CD39 and CD73 to eAdo, that inhibits this process. Therefore, eATP and eAdo upregulated in PB and BM modulate homing/engraftment in i) infused to the recipient donor-derived HSPCs and ii) in recipient BM microenvironment - in an opposite way. We also reported that the beneficial effect of eATP on homing/engraftment of HSPCs depends on the promotion of membrane lipid raft formation on the surface of HSPCs that incorporate homing receptors for their optimal interaction with BM released homing chemoattractants. This process is promoted by eATP activated Nlrp3 inflammasome. On the other hand, Nlrp3 inflammasome and membrane lipid raft formation are inhibited by eAdo in heme oxygenase-1 (HO-1)-dependent manner (Leukemia 2020; 34:1512-1523). Hypothesis. We hypothesized that proper modulation of eATP - eAdo signaling both at the level of transplanted HSPCs and recipient BM microenvironment will speed up the seeding efficiency of transplanted cells to BM niches. Material and Methods. We exposed HSPCs before transplantation ex vivo to i) exogenous eATP or ii) small molecular CD39 and CD73 inhibitors. We also inhibited CD39 and CD73 in transplant recipients BM at the time of myeloablative conditioning. In addition, we also activated ex vivo Nlrp3 inflammasome in HSPCs to be transplanted by specific activator nigericin. In control experiments, eATP stimulated Nlrp3 inflammasome activity was inhibited by the HO-1 activator that is CoPP. Homing of HSPCs was evaluated by measuring a number of donor-derived fluorochrome-labeled cells and clonogenic progenitors in BM of myeloablated hosts at 24 hours after transplantation. Early engraftment was assessed by counting the number of CFU-S and clonogeneic progenitors 12 days after transplantation and by evaluating kinetics of recovery of PB hematopoietic cell counts. Finally, while activation of Nlrp3 inflammasome was assessed by immunofluorescence assay, membrane lipid raft formation was evaluated by confocal microscopy. Results. We noticed that homing and engraftment of HSPCs was significantly accelerated after i) short exposure before transplantation to eATP, ii) inhibition of eAdo formation by CD39, and CD73 inhibitors, and iii) activation of Nlrp3 inflammasome by nigericin. Similarly, inhibition of eAdo formation in recipient BM microenvironment of transplanted mice by CD39 and CD73 inhibitors also improved homing and engraftment efficiency. This correlated with activation in the eATP-dependent manner of Nlrp3 inflammasome in HSPCs followed by membrane lipid raft formation. In the BM microenvironment, upregulation of eATP and inhibition of eAdo also enhanced expression of homing chemoattractants. Conclusions. Since all purinergic signaling modifiers employed in our studies are non-toxic against HSPCs, our data obtained in the animal model indicates that modulation of purinergic signaling before transplantation in HSPCs as well as in BM of the myeloablated recipient would significantly accelerate hematopoietic recovery after hematopoietic transplantation. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 3-3
Author(s):  
Magdalena Kucia ◽  
Kamila Bujko ◽  
Arjun Thapa ◽  
Janina Ratajczak ◽  
Mariusz Z Ratajczak

Background . It is known that prostaglandin E2 (PGE2) increases the homing and engraftment of hematopoietic stem/progenitor cells (HSPCs). However, aside from its role in upregulation of CXCR4 receptor expression on the surface of these cells, the exact mechanism has not been proposed. We have demonstrated in the past that an important step enabling the migration of HSPCs is the incorporation of CXCR4 into membrane lipid rafts on the leading surface (leading edge, in two dimensions) of migrating cells, which facilitates its interaction with cell migration signaling pathways (Wysoczynski M et al. Incorporation of CXCR4 into membrane lipid rafts primes homing-related responses of hematopoietic stem/progenitor cells to an SDF-1 gradient. Blood. 2005;105(1):40-48). Recently, we reported that Nlrp3 inflammasome-deficient HSPCs show a defect in lipid raft formation that results in defective migration of these cells in response to an SDF-1 gradient and their defective homing and engraftment after transplantation (Adamiak, M et al. Nlrp3 Inflammasome Signaling Regulates the Homing and Engraftment of Hematopoietic Stem Cells (HSPCs) by Enhancing Incorporation of CXCR4 Receptor into Membrane Lipid Rafts. Stem Cell Rev and Rep (2020). https://doi.org/10.1007/s12015-020-10005-w). An important activator of Nlrp3 inflammasomes is reactive oxygen species (ROS). Importantly, the enzyme that generates ROS, known as NADPH oxidase 2 (NOX2), is also associated with cell membrane lipid rafts. Hypothesis. Given the known roles of PGE2, membrane lipid rafts, and the Nlrp3 inflammasome in migration, homing, and engraftment of HSPCs, we hypothesized that PGE2 signaling promotes Nlrp3 inflammasome activation in a Nox2-ROS-dependent manner that results in incorporation of CXCR4 into membrane lipid rafts, which better explains the role of PGE2 in these phenomena.Materials and Methods. To test this hypothesis, murine SKL and human CD34+ cells enriched for HSPCs were stimulated with PGE2 to evaluate activation of genes of the Nlrp3 inflammasome complex at the mRNA and protein levels. Next, HSPCs from Nox2-KO mice were tested for membrane lipid raft formation in functional chemotaxis assays in response to SDF-1 gradients under conditions promoting membrane lipid raft formation. Formation of membrane lipid rafts in Nox2-KO cells was also evaluated by confocal analysis in the presence or absence of PGE2. Finally, the effect of the PGE2-Nox2-Nlrp3 inflammasome axis on the formation of membrane lipid rafts was evaluated in the presence of the ROS scavenger N-acethyl-cysteine (NAC). Results. We provide for the first time evidence that PGE2 activates Nlrp3 inflammasomes in HSPCs in a Nox2-ROS-dependent manner. This Nlrp3 inflammasome activation increases at the leading surface of migrating HSPCs with incorporation of the CXCR4 receptor into membrane lipid rafts. Formation of membrane lipid rafts was absent in Nox2-KO and Nlrp3-KO mouse HSPCs and in normal wild type cells after their exposure to NAC. Moreover, we also observed that Nox2-KO and Nlrp3-KO mice had a lower basal level of CXCR4 expression. Conclusions. Our results for the first time explain the role of PGE2 in promoting homing and migration of HSPCs, which occurs in response to PGE2 by activation of the Nox2-ROS-Nlrp3 inflammasome axis and thereby promotes incorporation of the CXCR4 receptor into membrane lipid rafts. Moreover, basal expression of the CXCR4 receptor was at a low level on the surface of HSPCs from Nlrp3-KO mice. Thus, our results provide evidence for the importance of the Nox2-ROS-Nlrp3 inflammasome axis in PGE2-mediated homing and engraftment of HSPCs and the role of PGE2-mediated lipid raft formation for optimal responsiveness of CXCR4 to SDF-1 in the BM microenvironment. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 11 ◽  
Author(s):  
Mariusz Z. Ratajczak ◽  
Magda Kucia

Nlrp3 inflammasome plays a pleiotropic role in hematopoietic cells. On the one hand, physiological activation of this intracellular protein complex is crucial to maintaining normal hematopoiesis and the trafficking of hematopoietic stem progenitor cells (HSPCs). On the other hand, its hyperactivation may lead to cell death by pyroptosis, and prolonged activity is associated with sterile inflammation of the BM and, as a consequence, with the HSPCs aging and origination of myelodysplasia and leukemia. Thus, we need to understand better this protein complex’s actions to define the boundaries of its safety window and study the transition from being beneficial to being detrimental. As demonstrated, the Nlrp3 inflammasome is expressed and active both in HSPCs and in the non-hematopoietic cells that are constituents of the bone marrow (BM) microenvironment. Importantly, the Nlrp3 inflammasome responds to mediators of purinergic signaling, and while extracellular adenosine triphosphate (eATP) activates this protein complex, its metabolite extracellular adenosine (eAdo) has the opposite effect. In this review, we will discuss and focus on the physiological consequences of the balance between eATP and eAdo in regulating the trafficking of HSPCs in an Nlrp3 inflammasome-dependent manner, as seen during pharmacological mobilization from BM into peripheral blood (PB) and in the reverse mechanism of homing from PB to BM and engraftment. We propose that both mediators of purinergic signaling and the Nlrp3 inflammasome itself may become important therapeutic targets in optimizing the trafficking of HSPCs in clinical settings.


Cells ◽  
2021 ◽  
Vol 10 (8) ◽  
pp. 2149
Author(s):  
Zhi Feng ◽  
Chenyu Lin ◽  
Limei Tu ◽  
Ming Su ◽  
Chunyu Song ◽  
...  

Hematopoietic stem cells (HSCs) are a specialized subset of cells with self-renewal and multilineage differentiation potency, which are essential for their function in bone marrow or umbilical cord blood transplantation to treat blood disorders. Expanding the hematopoietic stem and progenitor cells (HSPCs) ex vivo is essential to understand the HSPCs-based therapies potency. Here, we established a screening system in zebrafish by adopting an FDA-approved drug library to identify candidates that could facilitate HSPC expansion. To date, we have screened 171 drugs of 7 categories, including antibacterial, antineoplastic, glucocorticoid, NSAIDS, vitamins, antidepressant, and antipsychotic drugs. We found 21 drugs that contributed to HSPCs expansion, 32 drugs’ administration caused HSPCs diminishment and 118 drugs’ treatment elicited no effect on HSPCs amplification. Among these drugs, we further investigated the vitamin drugs ergocalciferol and panthenol, taking advantage of their acceptability, limited side-effects, and easy delivery. These two drugs, in particular, efficiently expanded the HSPCs pool in a dose-dependent manner. Their application even mitigated the compromised hematopoiesis in an ikzf1−/− mutant. Taken together, our study implied that the larval zebrafish is a suitable model for drug repurposing of effective molecules (especially those already approved for clinical use) that can facilitate HSPCs expansion.


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.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4528-4528
Author(s):  
Mateusz Adamiak ◽  
Kamila Bujko ◽  
Monika Plonka ◽  
Magdalena Kucia ◽  
Janina Ratajczak ◽  
...  

Abstract Background . One of the problems with the mobilization of hematopoietic stem/progenitor cells (HSPCs) in the clinic is that a significant number of patients are poor mobilizers. In order to develop more efficient mobilization strategies, we have to better understand the mobilization process at the molecular and cellular levels. We have reported that HSPCs express heme oxygenase 1 (HO-1), which is a negative regulator of the complement cascade (ComC), which in turn is required for stem cell mobilization (Stem Cell Rev. 2015, 11:110-8, Leukemia. 2017, 31:446-458). Moreover, very recently we became interested in the novel role of extracellular nucleotides (EXNs) and nucleosides in this process (Leukemia 2018, in press, doi: 10.1038/s41375-018-0122-0). The most important EXN is adenosine triphosphate (ATP), which is involved in intracellular energy transfer but if released from activated cells into the extracellular space becomes an important signaling molecule involved in purinergic signaling. Secretion of ATP from activated cells occurs in a pannexin channel-dependent manner and is additionally augmented by a positive regulatory loop after ATP binding to the P2X7 purinergic receptor. We recently found that the ATP concentration during the mobilization process increases to micromolar concentrations in the BM microenvironment. Moreover, we found that in the extracellular BM space ATP is processed by the CD39 and CD73 ectonucleotidases to ADP, AMP, and finally to adenosine, which is also an important mediator of purinergic signaling. Hypothesis. We hypothesized that EXNs and purinergic signaling via extracellular ATP and its metabolite adenosine are involved in triggering the mobilization of HSPCs and that this effect is mediated by the expression of HO-1 in HSPCs. Materials and Methods. To address this question, 2-month-old P2X7-/-, CD39-/-, and CD73-/-mice as well as their normal wild type (WT) littermates were mobilized with G-CSF or AMD3100. Following mobilization, we measured i) the total number of white blood cells (WBCs) and ii) the number of circulating clonogenic colony-forming unit granulocyte/macrophage (CFU-GM) progenitors and Sca-1+c-kit+lineage- (SKL) cells circulating in PB. The secretion of ATP from BM cells was inhibited by employing the pannexin 1 blocking drug probenecid or a synthetic pannexin 1 blocking peptide. In parallel, we evaluated i) the expression of HO-1 in HSPCs by employing RQ-PCR and western blot analysis and ii) the activation of the ComC by C5a ELISA. Results. Here we provide evidence that ATP, as an EXN secreted in a pannexin-1-dependent manner from BM cells, triggers activation of the ComC and initiates mobilization of HSPCs. Inhibition of the pannexin 1 channel by probenecid or a pannexin 1 blocking peptide inhibited this process. Furthermore, mobilization of HSPCs was augmented in a P2X7 receptor-dependent manner, evidenced by the finding that P2X7-/- mice were poor mobilizers. Furthermore, ATP is processed to adenosine in the extracellular space by CD39 and CD73 ectonucleotidases, and to our surprise we observed that CD73-deficient mice mobilize more HSPCs than their control wild type (WT) littermates, which indicates a novel negative role for adenosine in the mobilization process. This finding was confirmed by injecting mice with adenosine during AMD3100 administration. Finally, we found that ATP and adenosine modulate the mobilization process by regulating in opposite ways HSPC expression of HO-1, which is a negative regulator of mobilization. Therefore, while ATP downregulates expression of HO-1 in HSPCs and enhances mobilization, adenosine upregulates its expression and negatively affects the egress of HSPCs from BM into PB. Conclusions. We demonstrate for the first time that purinergic signaling involving ATP and its metabolite adenosine regulate the mobilization of HSPCs in an HO-1-dependent manner. While ATP triggers and promotes this process, adenosine has an inhibitory effect. The opposite effects of ATP and adenosine on the mobilization process can be explained by their different effects on the regulation of HO-1 expression in HSPCs. Finally, administration of ATP as a signaling molecule (together with G-CSF or AMD3100), inhibition of CD73 or HO-1 by small-molecule antagonists may provide the basis for more efficient mobilization strategies. Disclosures No relevant conflicts of interest to declare.


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

Blood ◽  
2011 ◽  
Vol 117 (23) ◽  
pp. 6083-6090 ◽  
Author(s):  
Ann Dahlberg ◽  
Colleen Delaney ◽  
Irwin D. Bernstein

AbstractDespite progress in our understanding of the growth factors that support the progressive maturation of the various cell lineages of the hematopoietic system, less is known about factors that govern the self-renewal of hematopoietic stem and progenitor cells (HSPCs), and our ability to expand human HSPC numbers ex vivo remains limited. Interest in stem cell expansion has been heightened by the increasing importance of HSCs in the treatment of both malignant and nonmalignant diseases, as well as their use in gene therapy. To date, most attempts to ex vivo expand HSPCs have used hematopoietic growth factors but have not achieved clinically relevant effects. More recent approaches, including our studies in which activation of the Notch signaling pathway has enabled a clinically relevant ex vivo expansion of HSPCs, have led to renewed interest in this arena. Here we briefly review early attempts at ex vivo expansion by cytokine stimulation followed by an examination of our studies investigating the role of Notch signaling in HSPC self-renewal. We will also review other recently developed approaches for ex vivo expansion, primarily focused on the more extensively studied cord blood–derived stem cell. Finally, we discuss some of the challenges still facing this field.


Sign in / Sign up

Export Citation Format

Share Document