Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Nov 1.
Published in final edited form as: Prostaglandins Other Lipid Mediat. 2011 Jun 21;96(1-4):3–9. doi: 10.1016/j.prostaglandins.2011.06.004

Pleiotropic effects of Prostaglandin E2 in hematopoiesis; Prostaglandin E2 and other eicosanoids regulate hematopoietic stem and progenitor cell function

Louis M Pelus 1, Jonathan Hoggatt 1
PMCID: PMC3206178  NIHMSID: NIHMS311780  PMID: 21722751

Abstract

Eicosanoids have been implicated in the physiological regulation of hematopoiesis with pleiotropic effects on hematopoietic stem cells and various classes of lineage restricted progenitor cells. Herein we review the effects of eicosanoids on hematopoiesis, focusing on new findings implicating prostaglandin E2 in enhancing hematopoietic stem cell engraftment by enhancing stem cell homing, survival and self-renewal. We also describe a role for cannabinoids in hematopoiesis. Lastly, we discuss the yin and yang of various eicosanoids in modulating hematopoietic stem and progenitor cell functions and summarize potential strategies to take advantage of these effects for therapeutic benefit for hematopoietic stem cell transplantation.

Keywords: eicosanoids, hematopoietic stem cells, transplantation, hematopoietic stem cell homing, stem cell engraftment

Introduction

Higher organisms maintain adequate numbers of blood cells throughout their entire lifespan to meet the normal physiological requirements of blood cell turnover, as well as respond to increased demand such as injury or infection. In man, approximately 1 trillion blood cells are produced every day, including 200 billion erythrocytes (red blood cells (RBCs)) and 70 billion neutrophils. This life long process is termed hematopoiesis. Stochastic and instructive mechanisms play important active roles in maintaining steady hematopoiesis and response to hematopoietic stress.

The hematopoietic stem cell (HSC) is at the center of blood cell production, having the capacity to produce all mature circulating blood cells, i.e., erythrocytes, platelets, lymphocytes, monocytes/macrophages, and all types of granulocytes. HSCs are defined by two fundamental characteristics: the ability to self-renew; i.e., the ability to form new HSCs, and the ability to differentiate through multilineage and lineage restricted hematopoietic progenitor cells (HPC) into all mature blood lineages. Breakthrough studies in the 1960’s by Till and McCulloch and colleagues showed that single clonogenic cells existed within the bone marrow that could self-renew and restore hematopoiesis [15] postulating the in vivo existence of a hematopoietic stem cell. These assays enumerated macroscopic nodules, colony-forming units-spleen (CFU-S) that formed on the spleens in proportion to the number of bone marrow cells injected [1]. While the hypothesis that CFU-S were HSC has turned out not to be true, rather they are more differentiated multipotent progenitor cells, these studies laid the groundwork for clinical hematopoietic transplantation. What is now clear is that the only true measure of HSC function is the ability to fully repopulate a lethally irradiated host. Assays that assess long-term repopulating cells (LTRC), an HSC synonym, utilize a donor HSC graft admixed with a competing congenic graft and markers distinct for the donor and competitor graft to distinguish blood production from each source of cells and calculation of competing repopulating units, a measure of HSC [6,7]. When compared in limiting-dilution, the frequency of competitive repopulating units (CRU) contained within the test graft can be determined by Poisson statistics [810]. Recently, it has become clear that HSCs are a heterogeneous population and classes of HSC with short (up to 16 weeks), intermediate (up to 32 weeks) and long-term (>32 weeks) [11] engraftment capabilities have been characterized. In light of these various potentials for self-renewal, the most stringent test of HSC potential, specifically the long-term HSC, is serial transplantation from primary recipients into secondary recipients, or beyond.

Before the advent of transplantation assays for HSC function, in vitro assays for culturing hematopoietic cells allowed many of the developmental pathways involved in hematopoietic homeostasis to be identified and the regulatory hematopoietic factors directing this process to be identified and cloned. These colony-forming cell assays identify populations of hematopoietic progenitor cells with distinct lineage-restricted differentiation patterns characterized by the type of colonies formed in semi-solid media. These colonies were determined to be clonally derived [12] and functionally distinct, establishing the beginnings of a hierarchical model. Alongside transplantation and clonogenic colony assays, development of monoclonal antibodies that define phenotypic markers of various hematopoietic cells has enabled placement of the various hematopoietic populations along a differentiation hierarchy, or “hematopoietic tree” (Figure 1).

Figure 1.

Figure 1

Blood cell development is a hierarchical process with self-renewal and maturational divisions occurring as a continuum under the direction of single or multiple growth factors. Shown is a simplistic representation incorporating current understandings of the hematopoietic process. Specific progenitor cells include the common lymphoid progenitor (CLP), common myeloid progenitor (CMP), granulocyte-monocyte progenitor (GMP), megakaryocyte-erythrocyte progenitor (MEP), the megakaryocyte (CFU-Meg) and erythroid (BFU-E) progenitors and the common macrophage and dendritic cell progenitor (MDP).

HSCs reside in very defined and limited microenvironments, or “niches” in the bone marrow [13], and signals within these niches direct HSC maintenance. Osteoblasts are a significant regulatory component of the endosteal bone marrow niche [1417]. Adhesion molecules, including, but not limited to, integrins, selectins, cadherins, osteopontin and CD44, as well as other receptors, contribute to HSC and HPC tethering in the bone marrow [18]. Perhaps the most important axis regulating HSC and HPC tethering and trafficking to and from the bone marrow niche, is the interaction between the CXC chemokine receptor 4 (CXCR4) and its ligand stromal cell-derived factor-1α (SDF-1α) [19,20].

Hematopoietic stem cell transplantation is routinely used to treat leukemias, cancer, hematologic diseases and metabolic disorders; however, long term blood reconstitution with some sources of HSCs is limited by inadequate number, inability to migrate/home to marrow niches, and poor engrafting efficiency and self-renewal [2123]. An appropriate bone marrow niche is required for HSCs to self-renew and differentiate and only HSCs homing, i.e., trafficking from the peripheral blood after injection to the bone marrow niche, are able to repopulate a lethally irradiated recipient long-term [24,25]. Homing is a rapid process, which is measured in hours (or at most 1–2 days) and is distinct from the concept of “engraftment”, which is more a description of the culmination of events pre- and post-homing.

Hematopoietic stem and progenitor cells normally reside within the bone marrow, while the mature cells they produce exit the marrow and enter the peripheral blood. Evidence over the last several decades clearly demonstrates that HSC and HPC also exit the bone marrow niche and traffic to the peripheral blood [2631]. This natural trafficking of HSC and HPC to the peripheral blood can be enhanced after chemotherapy treatment, or with pharmaceutical agents like granulocyte-colony stimulating factor (G-CSF) [28,29]. These “mobilized” cells can then be collected by apheresis and are widely used for autologous and allogeneic transplantation.

Pleiotropic Effects of Prostaglandins on Hematopoiesis

Numerous studies spanning the 1970s to 1990s documented physiological regulatory roles for prostaglandin E2 (PGE2) in hematopoiesis. Extensive work by us and others demonstrated that PGE inhibited the in vitro growth of human and mouse HPC, defined as colony forming unit-granulocyte/macrophage (CFU-GM) [3237]. We also showed that PGE acted as a negative regulator of myeloid expansion to counterbalance positive signaling from the colony-stimulating factors in order to maintain appropriate HPC proliferation [38,39] forming a selective feedback inhibition loop [37]. This physiological role of PGE2 in hematopoiesis and negative feedback regulation on myelopoiesis was documented by studies in mice differing in PGE synthetic capacity [39,40]; documentation of abnormal PGE2 responses in leukemia patients [32,33,36,41,42]; prognostic association of disordered PGE2 response in patients with myleodysplastic syndromes (MDS) [43]; abnormal HPC response in patients cured of germ cell tumors but progressing to acute leukemia [44]; and association of HPC response to PGE2 with clinical response to Interferon-γ in chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), and Hodgkin's disease patients [45,46]. Repetitive in vivo PGE2 administration validated inhibition of CFU-GM frequency and cell cycle rate, with decreased marrow and spleen cellularity [4749]. The effects of PGE2 on myeloid HPC were considered to be direct [50]; however, we later showed that PGE2 could induce F4/80+, Gr-1+, Mac-1+, myeloid suppressor cells, particularly when administered in vivo [49,51,52].

In contrast to effects on myelopoiesis, PGE2 stimulates erythropoiesis by increasing erythropoietin production by the kidney [53] and by enhancing proliferation of the erythroid progenitor cells (BFU-E) [5458]. Similarly, we showed that PGE2 enhances proliferation of multipotential progenitor cells that give rise to granulocytes, erythrocytes, monocytes and megakaryocytes (CFU-GEMM) [55,57]. In additional studies, we showed that PGE2 increased BFU-E and CFU-GEMM, which could be direct [59] or mediated through factors released by T cells [56,57,59].

Dendritic cell (DC) homeostasis, like all mature blood cells, is maintained via hierarchal generation from hematopoietic precursors and recent studies from our lab (P Singh and LM Pelus, unpublished) show that PGE2 regulates DC-committed progenitor cells and is required for optimal in vivo DC development.

While it is well established that prostaglandins have inhibitory effects on mature lymphocyte functions, PGE2 has thus far only been shown to inhibit B cell lymphopoiesis, having effects on pre-B cells [60,61]. Moreover, prostaglandins and cyclooxygenase enzymes present in stromal cells can regulate B lymphopoiesis [62].

Prostaglandins and Stem Cells

While much of the early biology of the effects of prostaglandins focused on progenitor cells, mainly owing to a lack of models to enumerate stem cell function, early studies clearly suggested that PGE2 likely affected HSC function and these effects were independent from effects on progenitor cells. Studies by Fehrer and Gidali in 1974 showed that treatment of murine marrow cells in vitro with PGE2 increased day 9 CFU-S in cell cycle that was cAMP independent [63]; however, in vivo dosing of PGE2 in mice led to little or no increase in hematopoiesis [64]. An increase in CFU-GM in S-phase was also seen after PGE2 pulse exposure of human marrow [65]. In 1982, we showed that short-term exposure of human or mouse bone marrow to PGE2 in vitro stimulated the production of cycling CFU-GM from a population of quiescent, non-cycling cells, most likely stem cells, which were dependent on time course and concentration of PGE2 [50] and were cAMP independent [66]. Kinetics of PGE2 exposure were critical for stimulatory versus inhibitory effects on HPC frequency and cell cycle and as little as 3 hour exposure of bone marrow cells to PGE2 increased the production of CFU-GM [66]. These findings were also recently validated with umbilical cord blood (UCB) cells, another source of HSC [67]. PGE2 treatment of purified human blood CD34+ cells was also shown to increase both myeloid and erythroid progenitor formation [68]. However, while highly suggestive, earlier studies did not directly measure HSC function. Recently, a requirement for PGE2 for development of hematopoiesis was found in a zebrafish screen [69] and ex vivo pulse exposure to the PGE2 derivative, 16,16 –dimethyl PGE2 (dmPGE2) was shown to increase the repopulating capacity of murine bone marrow cells and increase zebrafish kidney marrow recovery, validating the hypotheses proposed in the 1980’s. However, mechanisms for increased engraftment and recovery were not determined.

PGE2 increases long-term stem cell engraftment

Analysis of HSC frequency by Poisson statistics in either conventional limiting-dilution congenic transplant models in mice [69] or in a more refined limiting-dilution model in hybrid congenic mice that permits head-to-head comparison of the HSC populations [70] show a ~4-fold increase in HSC frequency as a result of short-term ex vivo exposure of marrow to dmPGE2 and strongly suggest a direct effect on HSC. At 5 months post-transplant, analysis of peripheral blood chimerism showed significantly higher levels of white blood cells derived from dmPGE2-treated marrow cells, with full myeloid cells and B and T lymphocyte reconstitution with no obvious lineage bias [70]. Transplant of marrow from primary transplant recipients into secondary recipients and secondary recipients into tertiary recipients, all without any further ex vivo manipulation, validated the self-renewal capacity of ex vivo dmPGE2-treated repopulating cells [70] and indicates that the effect of dmPGE2 pulse-exposure is stable and manifested on the long-term repopulating stem cell population (LT-HSC). Transplantation of human hematopoietic cells in immunodeficient mice offers a model system to evaluate human HSC function in vivo [71]. In a similar fashion to that shown using mouse bone marrow cells, short-term ex vivo pulse exposure to dmPGE2 was recently demonstrated to enhance engraftment of human umbilical cord blood HSC in NOD/SCID-IL2-γ-receptor null (NSG) mice [67]. It is important to note that these positive results on LT-HSC in the context of hematopoietic transplantation are the result of ex vivo treatment with dmPGE2, and are in slight contrast to an earlier in vivo study [64] and more recent study [72] using in vivo treatment with PGE2 where the enhancement by PGE2 was lost in competitive transplants in the long-term. These data suggest that the combined effects of extended in vivo PGE2 treatment acting directly through EP receptors on HSC and indirectly through modulation of the HSC niche in bone marrow has effects that differ from a short ex vivo pulse exposure of HSC to PGE2. Further studies exploring the role of in vivo versus ex vivo treatment with PGE2 and its analogues are likely to lead to refinements in clinical strategies for improving hematopoietic function and transplantation.

PGE2 increases HSC CXCR4 and migration to SDF-1α and homing efficiency

Effects on homing, apoptosis or proliferation can positively or negatively alter HSC function and hematopoietic transplantation. Since we previously showed that PGE2 can have dual effects on hematopoiesis [50,66] we sought to define its mechanism of action in enhancement of hematopoietic engraftment to better understand the potential clinical utility of transient ex vivo dmPGE2 exposure. We demonstrated that pulse-exposure of mouse and human stem and progenitor cells to dmPGE2 increases CXCR4 expression and significantly enhances migration to SDF-1α indicating that CXCR4 up-regulation on HSC coincides with enhanced migratory function. This enhancing effect of dmPGE2 on chemotaxis to SDF-1α was also blocked by AMD3100, a selective CXCR4 antagonist, further indicating a primary role for the CXCR4 receptor. These results are consistent with previous reports indicating that PGE2 increases CXCR4 expression in microvascular endothelial cells through stimulation of transcription factor binding to Sp1-binding sites [73], and that EP3/EP4 signaling in tumor stromal cells modulates CXCR4 signaling [74]. In addition, it has been reported that a PGE2 mediated increase in cAMP increases human CD34+ cell CXCR4 expression through a PKC-zeta signaling pathway [75].

As described earlier, successful hematopoietic reconstitution requires that administered HSC traffic/home to bone marrow niches where they can engraft, and the CXCR4/SDF-1α axis is a critical component of this homing process. We hypothesized that dmPGE2-induced enhancement of CXCR4 expression and migration to SDF-1α may increase in vivo homing of HSC, providing a mechanistic insight into the enhanced hematopoietic engraftment. Pulse-exposure of enriched mouse HSC to dmPGE2 ex vivo increased their bone marrow homing efficiency by 2-fold compared to cells treated with control vehicle when directly compared head-to-head in congenic hybrid mice. Increased homing of more differentiated cells was not observed when similarly evaluated, suggesting that the enhanced homing effect of dmPGE2 is specific to HSC. Similarly, dmPGE2 pulse exposure of human cord blood HSC significantly enhanced their homing efficiency in NSG mice [70,76].

PGE2 decreases apoptosis

While dmPGE2 enhanced the homing ability of HSC 2-fold over control, the enhancement seen in hematopoietic engraftment was 4-fold, suggesting other mechanisms mediating HSC engraftment were involved. Apoptosis is an important regulatory process in normal and malignant hematopoiesis and PGE2 signaling has been implicated in anti-apoptotic effects in many cell types [7779]. Pulse-exposure to dmPGE2 reduced Annexin-V and active caspase-3 levels in mouse and human HSC, suggesting that the enhancement of HSC function by dmPGE2 could result from enhanced HSC survival [70,76]. Consistent with reduced levels of active caspase-3, intracellular levels of the endogenous caspase-3 inhibitor Survivin were significantly higher in both mouse and human HSC, consistent with our previous findings that Survivin regulates apoptosis and proliferation in HSC [80,81] and studies by others that PGE2 can increase Survivin levels in cancer cells [82,83]. QRT-PCR analysis of treated HSC also showed similarly elevated levels of Survivin mRNA [70].

PGE2 increases entry of HSC into cell cycle

We previously showed that Survivin regulates HSC cell cycle entry and progression [80,81]. Early studies by us and others also showed that PGE2 can regulate the cell cycle of hematopoietic progenitors [50,63,65]. This suggests that an increase in HSC self-renewal and proliferation might contribute to the enhanced engraftment by dmPGE2-pulsed cells. In vitro exposure of highly purified primitive mouse LT-HSC to dmPGE2 significantly increases the proportion of LT-HSC in cell cycle (G1+ S/G2M) compared to controls. No significant effect on the cell cycle rate of HPC or more differentiated cells was observed, strongly suggesting that the effects of dmPGE2 on cell cycle rate are selective to HSC. To confirm the effect of dmPGE2 on enhancement of HSC cell cycle observed in vitro, bone marrow cells were pulsed with dmPGE2 and injected into congenic mice treated with BrdU post-transplant, and the proportion of donor BrdU+ HSC determined 16 hours later. A ~2-fold increase in the proportion of homed HSC in S+G2/M phase was observed for cells pulsed with dmPGE2 prior to transplant, confirming that short-term exposure of HSC to dmPGE2 stimulates HSC to enter and progress through cell cycle in vivo [70]. These studies suggest a model in which the coordinated effects of dmPGE2 treatment on homing, survival and proliferation lead to the 4-fold enhanced hematopoietic engeraftment.

Other Eicosanoids also affect Hematopoiesis

While most studies have focused on the effects of prostaglandins on hematopoiesis, the effects of other eicosanoids has not gone without notice. Like prostaglandins, leukotriene B4 (LTB4) and the cysteinyl leukotrienes are produced in the marrow microenvironment [84], by hematopoietic stromal cell cultures and by freshly isolated bone marrow mononuclear cells [85]. The 5’-Lipoxygenase (5’LOX) enzyme is also detected in hematopoietic progenitor cells [86].

LTB4 is a potent stimulator of granulocyte chemotaxis [87,88], while LTD4 stimulates chemotaxis and transendothelial migration of human HSC [86]. While PGE2 inhibits myeloid progenitor cells in vitro, LTB4, LTC4 and LTD4 increase mouse and human progenitor cells [8991]. Cyclooxygenase (COX) inhibitors also enhance myeloid progenitor cell proliferation whereas 5'-LOX inhibitors reduce myeloid progenitor cell proliferation [89,90,92]. Moreover, PGE stimulates early and late erythroid progenitor cells [57,58], while LTB4 and LTC4 inhibit them, and LOX inhibitors enhance erythroid progenitors [93]. In mice, dual COX inhibition enhances HPC recovery [92], while selective 5’-LOX inhibitors decrease CFU-GM and blast colony forming cells [89], suggesting an effect on a cell population more primitive than the CMP and CLP. The addition of LTB4 to UCB cells cultured with growth factors reduces total HSC produced and enhances HPC proliferation, whereas antagonism of the LTB4 receptor enhances production of HSC and blocks HPC proliferation [94]. Recently, deletion of 12/15-lipoxygenase was shown to reduce canonical Wnt signaling, leading to a reduction in HSC quiescence and hematopoietic defects [95].

While the cannabinoids have effects on mature cells of the immune system [96,97] it is becoming clear that they also have effects on earlier hematopoietic cells. Anandamide can act as a synergistic growth factor for hematopoietic progenitor cells [98] but promotes erythroid apoptosis [99]. 2-arachidonoylglycerol (2-AG) also stimulates progenitor cells [98] and hematopoietic cells expressing the cannabinoid CB2 receptor migrate in response to 2-AG [100]. Recently, 2-AG has also been shown to increase CFU-GEMM colony formation and cell migration [101], and activation of cannabinoid receptors on murine embryonic stem cells (ESC) promotes hematopoietic differentiation [102].

It is clear that prostaglandins, leukotrienes and cannabinoids have important roles in hematopoietic homeostasis, and evaluating their responses is critical to understanding eicosanoid function and development of eicosanoid-based therapeutic strategies for improvements in hematopoietic transplantation. While this review has focused on a few key players, there are abundantly more bioactive eicosanoids which regulate hematopoiesis with potential therapeutic benefit to cure diseases (as reviewed in [85,103,104]). While cyclooxygenase enzymes and their products are expressed in early and late erythroid progenitors and clearly regulate erythropoiesis [105], cytochrome P450 derived eicosanoids also effect erythropoiesis. Studies by Abraham et al. demonstrated that picomolar concentrations of the cytochrome P450 arachidonate metabolites, 19- and 20-hydroxyeicosatetranoic (HETE) acid, increased CFU-E growth 4 to 6 fold, with 20-HETE being considerably more potent [106]. Recently, 20-HETE was also shown to regulate the chemotactic response of endothelial progenitor cells to SDF-1α [107], possibly suggesting a role of yet another eicosanoid in trafficking of hematopoietic cells. Intriguingly, even the lipid substrate for eicosanoid enzymes can alter biologic effects, as omega-3 derived fatty acids vary considerably from omega-6 derived products [108]. Recently, it was demonstrated that mice fed a diet high in omega-3 fatty acids had reduced CFU-M and CFU-GM, yet had increased common myeloid progenitors [109]. Clearly, furthering our understanding of the interactions and molecular pathways of the plethora of eicosanoids and their role in hematopoietic regulation are likely to lead to advances in clinical therapies.

The Yin and Yang of Prostaglandins and Endocannabinoids in Hematopoiesis

In many physiological systems, prostaglandins, leukotrienes and endocannabinoids exhibit compensatory or opposing roles [110]. Prostaglandins and leukotrienes have numerous opposing roles in pulmonary fibrosis [111], whereas in other systems they act coordinately [112]. We and others have shown that cannabinoids reduce signaling through the CXCR4 receptor [110,113,114] and as a consequence reduce neutrophil migration [115,116]. This is in contrast to PGE2 that up regulates CXCR4 expression [70,76,110], suggesting that prostaglandins and endocannabinoids can act in opposing fashion in hematopoiesis. Analysis of cannabinoid receptor expression using numerous antibodies and flow cytometry showed that they are expressed on mouse and human HSC. Utilizing the dual cannabinoid receptor agonist CP55940 we found that it reduced both CXCR4 and the adhesion molecule very late antigen-4 (VLA-4) expression on mouse HSC, while dmPGE2 increased expression of CXCR4 and VLA-4 expression [110]. These data support a yin and yang role for cannabinoids and prostaglandins on hematopoietic cells and the reduction of CXCR4 by cannabinoid agonism suggested that they may facilitate un-tethering of HSC and HPC from bone marrow niches that could be used to mobilize stem cells. In a further series of studies, we found that single administration of the CB2 selective agonist GP1a and the dual CB1/CB2 agonist CP55940 but not the CB1 selective agonist ACEA significantly mobilized hematopoietic progenitor cells to the peripheral blood and that the addition of a single dose of CP55940 to a standard 4-day mobilizing regimen of G-CSF significantly increased progenitor cell mobilization compared to G-CSF alone [110]. Two separate reports by Jiang et al. have also now demonstrated that cannabinoid signaling mediates mobilization, and that endocannabinoids are expressed in bone marrow and increase HPC migration and proliferation in vitro [117,118].

Summary

In vivo analysis of the effects of short-term pulse exposure to dmPGE2 clearly demonstrates that it has direct and stable effects on HSC function. Enhancement of HSC frequency and engraftment by dmPGE2 results from effects on HSC homing and cell cycle activity involving up-regulation of CXCR4 and Survivin, with increased chemotactic response to SDF-1α and reduced apoptosis. The ability to facilitate homing, survival and proliferation of HSC by short-term ex vivo dmPGE2 exposure offers an exciting clinical translation strategy to improve hematopoietic transplantation, especially in transplant settings characterized by low HSC numbers, such as umbilical cord blood and some mobilized peripheral blood stem cell products. Our experimental preclinical limiting dilution transplant studies show that equivalent engraftment is achieved with four-fold fewer dmPGE2-treated cells compared to untreated cells. Homing and migration studies utilizing UCB HSC also clearly support potential translation of short-term dmPGE2 exposure to human hematopoietic grafts. Clinical analysis of the ability of dmPGE2 to enhance engraftment of UCB is currently ongoing [119].

The available data suggest that LTB4 signaling decreases HSC self-renewal and increases differentiation, while blocking LTB4 receptors increases self-renewal and blocks differentiation. Thus, the use of a leukotriene receptor antagonist or LOX inhibitor in the post-transplant setting may favor self-renewal.

The effects of cannabinoids on hematopoietic stem and progenitor cell mobilization suggest that they can be modulated for therapeutic benefit. The fact that signaling through the CB receptors on hematopoietic cells can have effects opposite to PGE2 suggests several improvements to therapeutic strategies. Since CB signaling inhibits CXCR4 mediated migration while dmPGE2 exposure up regulates CXCR4 expression, combination use of ex vivo dmPGE2 exposure plus CB receptor antagonism may improve hematopoietic stem cell homing. Similarly, while dmPGE2 production within the bone marrow may provide signals enforcing cell retention through up regulation of CXCR4 and VLA-4, inhibition of PGE2 receptor signaling coordinate with agonizing CB receptors may facilitate acquisition of mobilized hematopoietic stem and progenitor cells for transplantation.

The availability of FDA approved pharmaceuticals that specifically regulate biosynthesis and signaling of prostaglandins, leukotrienes, cannabinoids and other eicosanoids will facilitate rapid translation of eicosanoid based therapeutic research, both at the level of improving the yield of a hematopoietic graft or measured in terms of graft performance.

Highlights.

  • Short-term pulse exposure of hematopoietic stem cells (HSC) to dmPGE2 enhances their frequency and engraftment resulting from effects on HSC homing and cell cycle activity involving up-regulation of CXCR4 and Survivin, with increased chemotactic response to SDF-1α and reduced apoptosis.

  • Short-term pulse exposure of hematopoietic stem cells to dmPGE2 has direct and stable effects on HSC function.

  • The ability to facilitate homing, survival and proliferation of HSC by short-term ex vivo dmPGE2 exposure offers an exciting clinical translation strategy to improve hematopoietic transplantation,

Acknowledgments

Supported by Grant HL09305 (to LMP), NHLBI, National Institutes of Health

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Till JE, McCulloch EA. A direct measurement of the radiation sensitivity of normal mouse bone marrow cells. Radiat Res. 1961;14:213–222. [PubMed] [Google Scholar]
  • 2.Becker AJ, McCulloch EA, Till JE. Cytological demonstration of the clonal nature of spleen colonies derived from transplanted mouse marrow cells. Nature. 1963;197:452–454. doi: 10.1038/197452a0. [DOI] [PubMed] [Google Scholar]
  • 3.Siminovitch L, McCulloch EA, Till JE. The Distibution of Colony-forming Cells Among Spleen Colonies. J Cell Physiol. 1963;62:327–336. doi: 10.1002/jcp.1030620313. [DOI] [PubMed] [Google Scholar]
  • 4.Wu AM, Till JE, Siminovitch L, McCulloch EA. A cytological study of the capacity for differentiation of normal hemopoietic colony-forming cells. J Cell Physiol. 1967;69:177–184. doi: 10.1002/jcp.1040690208. [DOI] [PubMed] [Google Scholar]
  • 5.Wu AM, Till JE, Siminovitch L, McCulloch EA. Cytological evidence for a relationship between normal hemotopoietic colony-forming cells and cells of the lymphoid system. J Exp Med. 1968 Jan 3;127:455–464. doi: 10.1084/jem.127.3.455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Harrison DE. Competitive repopulation: a new assay for long-term stem cell functional capacity. Blood. 1980;55:77–81. [PubMed] [Google Scholar]
  • 7.Harrison DE, Jordan CT, Zhong RK, Astle CM. Primitive hemopoietic stem cells: direct assay of most productive populations by competitive repopulation with simple binomial, correlation and covariance calculations. Exp Hematol. 1993;21:206–219. [PubMed] [Google Scholar]
  • 8.Taswell C. Limiting dilution assays for the determination of immunocompetent cell frequencies. I. Data analysis. J Immunol. 1981;126:1614–1619. [PubMed] [Google Scholar]
  • 9.Szilvassy SJ, Humphries RK, Lansdorp PM, Eaves AC, Eaves CJ. Quantitative assay for totipotent reconstituting hematopoietic stem cells by a competitive repopulation strategy. Proc Natl Acad Sci U S A. 1990;87:8736–8740. doi: 10.1073/pnas.87.22.8736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Szilvassy SJ, Lansdorp PM, Humphries RK, Eaves AC, Eaves CJ. Isolation in a single step of a highly enriched murine hematopoietic stem cell population with competitive long-term repopulating ability. Blood. 1989 Aug 15;74:930–939. [PubMed] [Google Scholar]
  • 11.Benveniste P, Frelin C, Janmohamed S, Barbara M, Herrington R, Hyam D, Iscove NN. Intermediate-term hematopoietic stem cells with extended but time-limited reconstitution potential. Cell Stem Cell. 2010 Jan 8;6:48–58. doi: 10.1016/j.stem.2009.11.014. [DOI] [PubMed] [Google Scholar]
  • 12.Prchal JF, Adamson JW, Steinmann L, Fialkow PJ. Human erythroid colony formation in vitro: evidence for clonal origin. J Cell Physiol. 1976;89:489–492. doi: 10.1002/jcp.1040890314. [DOI] [PubMed] [Google Scholar]
  • 13.Schofield R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells. 1978;4:7–25. [PubMed] [Google Scholar]
  • 14.Calvi LM, Adams GB, Weibrecht KW, Weber JM, Olson DP, Knight MC, Martin RP, Schipani E, Divieti P, Bringhurst FR, Milner LA, Kronenberg HM, Scadden DT. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 2003 Oct 23;425:841–846. doi: 10.1038/nature02040. [DOI] [PubMed] [Google Scholar]
  • 15.Zhang J, Niu C, Ye L, Huang H, He X, Tong WG, Ross J, Haug J, Johnson T, Feng JQ, Harris S, Wiedemann LM, Mishina Y, Li L. Identification of the haematopoietic stem cell niche and control of the niche size. Nature. 2003 Oct 23;425:836–841. doi: 10.1038/nature02041. [DOI] [PubMed] [Google Scholar]
  • 16.Visnjic D, Kalajzic Z, Rowe DW, Katavic V, Lorenzo J, Aguila HL. Hematopoiesis is severely altered in mice with an induced osteoblast deficiency. Blood. 2004 May 1;103:3258–3264. doi: 10.1182/blood-2003-11-4011. [DOI] [PubMed] [Google Scholar]
  • 17.Arai F, Hirao A, Ohmura M, Sato H, Matsuoka S, Takubo K, Ito K, Koh GY, Suda T. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell. 2004 Jul 23;118:149–161. doi: 10.1016/j.cell.2004.07.004. [DOI] [PubMed] [Google Scholar]
  • 18.Lam BS, Adams GB. Hematopoietic stem cell lodgment in the adult bone marrow stem cell niche. Int J Lab Hematol. 2010;32:551–558. doi: 10.1111/j.1751-553X.2010.01250.x. [DOI] [PubMed] [Google Scholar]
  • 19.Kollet O, Spiegel A, Peled A, Petit I, Byk T, Hershkoviz R, Guetta E, Barkai G, Nagler A, Lapidot T. Rapid and efficient homing of human CD34(+)CD38(−/low)CXCR4(+) stem and progenitor cells to the bone marrow and spleen of NOD/SCID and NOD/SCID/B2m(null) mice. Blood. 2001 May 15;97:3283–3291. doi: 10.1182/blood.v97.10.3283. [DOI] [PubMed] [Google Scholar]
  • 20.Peled A, Petit I, Kollet O, Magid M, Ponomaryov T, Byk T, Nagler A, Ben-Hur H, Many A, Shultz L, Lider O, Alon R, Zipori D, Lapidot T. Dependence of human stem cell engraftment and repopulation of NOD/SCID mice on CXCR4. Science. 1999 Feb 5;283:845–848. doi: 10.1126/science.283.5403.845. [DOI] [PubMed] [Google Scholar]
  • 21.Hall KM, Horvath TL, Abonour R, Cornetta K, Srour EF. Decreased homing of retrovirally transduced human bone marrow CD34+ cells in the NOD/SCID mouse model. Exp Hematol. 2006;34:433–442. doi: 10.1016/j.exphem.2005.12.014. [DOI] [PubMed] [Google Scholar]
  • 22.Porecha NK, English K, Hangoc G, Broxmeyer HE, Christopherson KW. Enhanced functional response to CXCL12/SDF-1 through retroviral overexpression of CXCR4 on M07e cells: implications for hematopoietic stem cell transplantation. Stem Cells Dev. 2006;15:325–333. doi: 10.1089/scd.2006.15.325. [DOI] [PubMed] [Google Scholar]
  • 23.Rocha V, Broxmeyer HE. New approaches for improving engraftment after cord blood transplantation. Biol Blood Marrow Transplant. 2010;16:S126–S132. doi: 10.1016/j.bbmt.2009.11.001. [DOI] [PubMed] [Google Scholar]
  • 24.Nibley WE, Spangrude GJ. Primitive stem cells alone mediate rapid marrow recovery and multilineage engraftment after transplantation. Bone Marrow Transplant. 1998;21:345–354. doi: 10.1038/sj.bmt.1701097. [DOI] [PubMed] [Google Scholar]
  • 25.Lanzkron SM, Collector MI, Sharkis SJ. Homing of long-term and short-term engrafting cells in vivo. Ann N Y Acad Sci. 1999 Apr 30;872:48–54. doi: 10.1111/j.1749-6632.1999.tb08452.x. [DOI] [PubMed] [Google Scholar]
  • 26.Massberg S, Schaerli P, Knezevic-Maramica I, Kollnberger M, Tubo N, Moseman EA, Huff IV, Junt T, Wagers AJ, Mazo IB, von Andrian UH. Immunosurveillance by hematopoietic progenitor cells trafficking through blood, lymph, and peripheral tissues. Cell. 2007 Nov 30;131:994–1008. doi: 10.1016/j.cell.2007.09.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.McKinney-Freeman S, Goodell MA. Circulating hematopoietic stem cells do not efficiently home to bone marrow during homeostasis. Exp Hematol. 2004;32:868–876. doi: 10.1016/j.exphem.2004.06.010. [DOI] [PubMed] [Google Scholar]
  • 28.Wright DE, Wagers AJ, Gulati AP, Johnson FL, Weissman IL. Physiological migration of hematopoietic stem and progenitor cells. Science. 2001 Nov 30;294:1933–1936. doi: 10.1126/science.1064081. [DOI] [PubMed] [Google Scholar]
  • 29.Abkowitz JL, Robinson AE, Kale S, Long MW, Chen J. Mobilization of hematopoietic stem cells during homeostasis and after cytokine exposure. Blood. 2003 Aug 15;102:1249–1253. doi: 10.1182/blood-2003-01-0318. [DOI] [PubMed] [Google Scholar]
  • 30.Chervenick PA, Boggs DR. In vitro growth of granulocytic and mononuclear cell colonies from blood of normal individuals. Blood. 1971;37:131–135. [PubMed] [Google Scholar]
  • 31.Goodman JW, Hodgson GS. Evidence for stem cells in the peripheral blood of mice. Blood. 1962;19:702–714. [PubMed] [Google Scholar]
  • 32.Aglietta M, Piacibello W, Gavosto F. Insensitivity of chronic myeloid leukemia cells to inhibition of growth by prostaglandin E1. Cancer Res. 1980;40:2507–2511. [PubMed] [Google Scholar]
  • 33.Pelus LM, Broxmeyer HE, Clarkson BD, Moore MA. Abnormal responsiveness of granulocyte-macrophage committed colony-forming cells from patients with chronic myeloid leukemia to inhibition by prostaglandin E1. Cancer Res. 1980;40:2512–2515. [PubMed] [Google Scholar]
  • 34.Pelus LM, Broxmeyer HE, Moore MA. Regulation of human myelopoiesis by prostaglandin E and lactoferrin. Cell Tissue Kinet. 1981;14:515–526. doi: 10.1111/j.1365-2184.1981.tb00557.x. [DOI] [PubMed] [Google Scholar]
  • 35.Taetle R, Guittard JP, Mendelsohn JM. Abnormal modulation of granulocyte/macrophage progenitor proliferation by prostaglandin E in chronic myeloproliferative disorders. Exp Hematol. 1980;8:1190–1201. [PubMed] [Google Scholar]
  • 36.Taetle R, Koessler A. Effects of cyclic nucleotides and prostaglandins on normal and abnormal human myeloid progenitor proliferation. Cancer Res. 1980;40:1223–1229. [PubMed] [Google Scholar]
  • 37.Pelus LM, Broxmeyer HE, Kurland JI, Moore MA. Regulation of macrophage and granulocyte proliferation. Specificities of prostaglandin E and lactoferrin. J Exp Med. 1979 Aug 1;150:277–292. doi: 10.1084/jem.150.2.277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kurland JI, Broxmeyer HE, Pelus LM, Bockman RS, Moore MA. Role for monocyte-macrophage-derived colony-stimulating factor and prostaglandin E in the positive and negative feedback control of myeloid stem cell proliferation. Blood. 1978;52:388–407. [PubMed] [Google Scholar]
  • 39.Kurland JI, Pelus LM, Ralph P, Bockman RS, Moore MA. Induction of prostaglandin E synthesis in normal and neoplastic macrophages: role for colony-stimulating factor(s) distinct from effects on myeloid progenitor cell proliferation. Proc Natl Acad Sci U S A. 1979;76:2326–2330. doi: 10.1073/pnas.76.5.2326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kincade PW, Lee G, Fernandes G, Moore MA, Williams N, Good RA. Abnormalities in clonable B lymphocytes and myeloid progenitors in autoimmune NZB mice. Proc Natl Acad Sci U S A. 1979;76:3464–3468. doi: 10.1073/pnas.76.7.3464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Pelus LM, Gold E, Saletan S, Coleman M. Restoration of responsiveness of chronic myeloid leukemia granulocyte-macrophage colony-forming cells to growth regulation in vitro following preincubation with prostaglandin E. Blood. 1983;62:158–165. [PubMed] [Google Scholar]
  • 42.Moore MA, Mertelsmann R, Pelus LM. Phenotypic evaluation of chronic myeloid leukemia. Blood Cells. 1981;7:217–236. [PubMed] [Google Scholar]
  • 43.Gold EJ, Conjalka M, Pelus LM, Jhanwar SC, Broxmeyer H, Middleton AB, Clarkson BD, Moore MA. Marrow cytogenetic and cell-culture analyses of the myelodysplastic syndromes: insights to pathophysiology and prognosis. J Clin Oncol. 1983;1:627–634. doi: 10.1200/JCO.1983.1.10.627. [DOI] [PubMed] [Google Scholar]
  • 44.Leitner SP, Bosl GJ, Pelus LM. Abnormal colony formation and prostaglandin E responsiveness of myeloid progenitor cells in patients cured of germ cell neoplasms after combination chemotherapy. Cancer. 1987 Aug 1;60:312–317. doi: 10.1002/1097-0142(19870801)60:3<312::aid-cncr2820600307>3.0.co;2-o. [DOI] [PubMed] [Google Scholar]
  • 45.Pelus LM, Vadhan-Raj S. Modulation of responsiveness of chronic myelogenous leukemia granulocyte-macrophage colony-forming cells to growth regulation following in vivo treatment with recombinant gamma-interferon. Am J Hematol. 1988;28:21–26. doi: 10.1002/ajh.2830280105. [DOI] [PubMed] [Google Scholar]
  • 46.Vadhan-Raj S, Al Katib A, Bhalla R, Pelus L, Nathan CF, Sherwin SA, Oettgen HF, Krown SE. Phase I trial of recombinant interferon gamma in cancer patients. J Clin Oncol. 1986;4:137–146. doi: 10.1200/JCO.1986.4.2.137. [DOI] [PubMed] [Google Scholar]
  • 47.Gentile PS, Byer D, Pelus LM. In vivo modulation of murine myelopoiesis following intravenous administration of prostaglandin E2. Blood. 1983;62:1100–1107. [PubMed] [Google Scholar]
  • 48.Gentile PS, Pelus LM. In vivo modulation of myelopoiesis by prostaglandin E2. II. Inhibition of granulocyte-monocyte progenitor cell (CFU-GM) cell-cycle rate. Exp Hematol. 1987;15:119–126. [PubMed] [Google Scholar]
  • 49.Pelus LM, Gentile PS. In vivo modulation of myelopoiesis by prostaglandin E2. III. Induction of suppressor cells in marrow and spleen capable of mediating inhibition of CFU-GM proliferation. Blood. 1988;71:1633–1640. [PubMed] [Google Scholar]
  • 50.Pelus LM. Association between colony forming units-granulocyte macrophage expression of Ia-like (HLA-DR) antigen and control of granulocyte and macrophage production. A new role for prostaglandin E. J Clin Invest. 1982;70:568–578. doi: 10.1172/JCI110649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Gentile PS, Pelus LM. In vivo modulation of myelopoiesis by prostaglandin E2. IV. Prostaglandin E2 induction of myelopoietic inhibitory activity. J Immunol. 1988 Oct 15;141:2714–2720. [PubMed] [Google Scholar]
  • 52.Pelus LM. Blockade of prostaglandin biosynthesis in intact mice dramatically augments the expansion of committed myeloid progenitor cells (colony-forming units-granulocyte, macrophage) after acute administration of recombinant human IL-1 alpha. J Immunol. 1989 Dec 15;143:4171–4179. [PubMed] [Google Scholar]
  • 53.Dukes PP, Shore NA, Hammond D, Ortega JA, Datta MC. Enhancement of erythropoiesis by prostaglandins. J Lab Clin Med. 1973;82:704–712. [PubMed] [Google Scholar]
  • 54.DeGowin RL, Gibson DP. Prostaglandin-mediated enhancement of erythroid colonies by marrow stromal cells (MSC) Exp Hematol. 1981;9:274–280. [PubMed] [Google Scholar]
  • 55.Lu L, Pelus LM, Broxmeyer HE. Modulation of the expression of HLA-DR (Ia) antigens and the proliferation of human erythroid (BFU-E) and multipotential (CFU-GEMM) progenitor cells by prostaglandin E. Exp Hematol. 1984;12:741–748. [PubMed] [Google Scholar]
  • 56.Lu L, Pelus LM, Broxmeyer HE, Moore MA, Wachter M, Walker D, Platzer E. Enhancement of the proliferation of human marrow erythroid (BFU-E) progenitor cells by prostaglandin E requires the participation of OKT8-positive T lymphocytes and is associated with the density expression of major histocompatibility complex class II antigens on BFU-E. Blood. 1986;68:126–133. [PubMed] [Google Scholar]
  • 57.Lu L, Pelus LM, Piacibello W, Moore MA, Hu W, Broxmeyer HE. Prostaglandin E acts at two levels to enhance colony formation in vitro by erythroid (BFU-E) progenitor cells. Exp Hematol. 1987;15:765–771. [PubMed] [Google Scholar]
  • 58.Rossi GB, Migliaccio AR, Migliaccio G, Lettieri F, Di RM, Peschle C, Mastroberardino G. In vitro interactions of PGE and cAMP with murine and human erythroid precursors. Blood. 1980;56:74–79. [PubMed] [Google Scholar]
  • 59.Nocka KH, Ottman OG, Pelus LM. The role of marrow accessory cell populations in the augmentation of human erythroid progenitor cell (BFU-E) proliferation by prostaglandin E. Leuk Res. 1989;13:527–534. doi: 10.1016/0145-2126(89)90119-7. [DOI] [PubMed] [Google Scholar]
  • 60.Shimozato T, Kincade PW. Prostaglandin E(2) and stem cell factor can deliver opposing signals to B lymphocyte precursors. Cell Immunol. 1999 Nov 25;198:21–29. doi: 10.1006/cimm.1999.1575. [DOI] [PubMed] [Google Scholar]
  • 61.Yokota T, Meka CS, Medina KL, Igarashi H, Comp PC, Takahashi M, Nishida M, Oritani K, Miyagawa J, Funahashi T, Tomiyama Y, Matsuzawa Y, Kincade PW. Paracrine regulation of fat cell formation in bone marrow cultures via adiponectin and prostaglandins. J Clin Invest. 2002;109:1303–1310. doi: 10.1172/JCI14506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Borghesi LA, Smithson G, Kincade PW. Stromal cell modulation of negative regulatory signals that influence apoptosis and proliferation of B lineage lymphocytes. J Immunol. 1997 Nov 1;159:4171–4179. [PubMed] [Google Scholar]
  • 63.Feher I, Gidali J. Prostaglandin E2 as stimulator of haemopoietic stem cell proliferation. Nature. 1974 Feb 22;247:550–551. doi: 10.1038/247550a0. [DOI] [PubMed] [Google Scholar]
  • 64.Gidali J, Feher I. The effect of E type prostaglandins on the proliferation of haemopoietic stem cells in vivo. Cell Tissue Kinet. 1977;10:365–373. [PubMed] [Google Scholar]
  • 65.Verma DS, Spitzer G, Zander AR, McCredie KB, Dicke KA. Prostaglandin E1-mediated augmentation of human granulocyte-macrophage progenitor cell growth in vitro. Leuk Res. 1981;5:65–71. doi: 10.1016/0145-2126(81)90097-7. [DOI] [PubMed] [Google Scholar]
  • 66.Pelus LM. Prostaglandin-E-mediated modulation of human marrow CFU-GM Ia-antigen expression: kinetics and specificity. Exp Hematol. 1984;12:831–837. [PubMed] [Google Scholar]
  • 67.Goessling W, Allen RS, Guan X, Jin P, Uchida N, Dovey M, Harris JM, Metzger ME, Bonifacino AC, Stroncek D, Stegner J, Armant M, Schlaeger T, Tisdale JF, Zon LI, Donahue RE, North TE. Prostaglandin e2 enhances human cord blood stem cell xenotransplants and shows long-term safety in preclinical nonhuman primate transplant models. Cell Stem Cell. 2011 Apr 8;8:445–458. doi: 10.1016/j.stem.2011.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Dupuis F, Gachard N, Allegraud A, Praloran V, Denizot Y. Prostaglandin E2 Stimulates the Growth of Human Blood CD34+ Progenitors. Prostaglandins Other Lipid Mediat. 1998;55:179–186. [Google Scholar]
  • 69.North TE, Goessling W, Walkley CR, Lengerke C, Kopani KR, Lord AM, Weber GJ, Bowman TV, Jang IH, Grosser T, Fitzgerald GA, Daley GQ, Orkin SH, Zon LI. Prostaglandin E2 regulates vertebrate haematopoietic stem cell homeostasis. Nature. 2007 Jun 21;447:1007–1011. doi: 10.1038/nature05883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Hoggatt J, Singh P, Sampath J, Pelus LM. Prostaglandin E2 enhances hematopoietic stem cell homing, survival, and proliferation. Blood. 2009 May 28;113:5444–5455. doi: 10.1182/blood-2009-01-201335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Dick JE, Bhatia M, Gan O, Kapp U, Wang JC. Assay of human stem cells by repopulation of NOD/SCID mice. Stem Cells. 1997;15 Suppl 1:199–203. doi: 10.1002/stem.5530150826. [DOI] [PubMed] [Google Scholar]
  • 72.Frisch BJ, Porter RL, Gigliotti BJ, Olm-Shipman AJ, Weber JM, O'Keefe RJ, Jordan CT, Calvi LM. In vivo prostaglandin E2 treatment alters the bone marrow microenvironment and preferentially expands short-term hematopoietic stem cells. Blood. 2009 Nov 5;114:4054–4063. doi: 10.1182/blood-2009-03-205823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Salcedo R, Zhang X, Young HA, Michael N, Wasserman K, Ma WH, Martins-Green M, Murphy WJ, Oppenheim JJ. Angiogenic effects of prostaglandin E2 are mediated by up-regulation of CXCR4 on human microvascular endothelial cells. Blood. 2003 Sep 15;102:1966–1977. doi: 10.1182/blood-2002-11-3400. [DOI] [PubMed] [Google Scholar]
  • 74.Katoh H, Hosono K, Ito Y, Suzuki T, Ogawa Y, Kubo H, Kamata H, Mishima T, Tamaki H, Sakagami H, Sugimoto Y, Narumiya S, Watanabe M, Majima M. COX-2 and prostaglandin EP3/EP4 signaling regulate the tumor stromal proangiogenic microenvironment via CXCL12-CXCR4 chemokine systems. Am J Pathol. 2010;176:1469–1483. doi: 10.2353/ajpath.2010.090607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Goichberg P, Kalinkovich A, Borodovsky N, Tesio M, Petit I, Nagler A, Hardan I, Lapidot T. cAMP-induced PKCzeta activation increases functional CXCR4 expression on human CD34+ hematopoietic progenitors. Blood. 2006 Feb 1;107:870–879. doi: 10.1182/blood-2005-03-0941. [DOI] [PubMed] [Google Scholar]
  • 76.Pelus LM, Hoggatt J, Singh P. Pulse exposure of haematopoietic grafts to prostaglandin E2 in vitro facilitates engraftment and recovery. Cell Prolif. 2011;44 Suppl 1:22–29. doi: 10.1111/j.1365-2184.2010.00726.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Fernandez-Martinez A, Molla B, Mayoral R, Bosca L, Casado M, Martin-Sanz P. Cyclo-oxygenase 2 expression impairs serum-withdrawal-induced apoptosis in liver cells. Biochem J. 2006 Sep 15;398:371–380. doi: 10.1042/BJ20060780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.George RJ, Sturmoski MA, Anant S, Houchen CW. EP4 mediates PGE2 dependent cell survival through the PI3 kinase/AKT pathway. Prostaglandins Other Lipid Mediat. 2007;83:112–120. doi: 10.1016/j.prostaglandins.2006.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Negrotto S, Pacienza N, D'Atri LP, Pozner RG, Malaver E, Torres O, Lazzari MA, Gomez RM, Schattner M. Activation of cyclic AMP pathway prevents CD34(+) cell apoptosis. Exp Hematol. 2006;34:1420–1428. doi: 10.1016/j.exphem.2006.05.017. [DOI] [PubMed] [Google Scholar]
  • 80.Fukuda S, Pelus LM. Regulation of the inhibitor-of-apoptosis family member survivin in normal cord blood and bone marrow CD34(+) cells by hematopoietic growth factors: implication of survivin expression in normal hematopoiesis. Blood. 2001 Oct 1;98:2091–2100. doi: 10.1182/blood.v98.7.2091. [DOI] [PubMed] [Google Scholar]
  • 81.Fukuda S, Foster RG, Porter SB, Pelus LM. The antiapoptosis protein survivin is associated with cell cycle entry of normal cord blood CD34(+) cells and modulates cell cycle and proliferation of mouse hematopoietic progenitor cells. Blood. 2002 Oct 1;100:2463–2471. doi: 10.1182/blood.V100.7.2463. [DOI] [PubMed] [Google Scholar]
  • 82.Baratelli F, Krysan K, Heuze-Vourc'h N, Zhu L, Escuadro B, Sharma S, Reckamp K, Dohadwala M, Dubinett SM. PGE2 confers survivin-dependent apoptosis resistance in human monocyte-derived dendritic cells. J Leukoc Biol. 2005;78:555–564. doi: 10.1189/jlb.1004569. [DOI] [PubMed] [Google Scholar]
  • 83.Krysan K, Merchant FH, Zhu L, Dohadwala M, Luo J, Lin Y, Heuze-Vourc'h N, Pold M, Seligson D, Chia D, Goodglick L, Wang H, Strieter R, Sharma S, Dubinett S. COX-2-dependent stabilization of survivin in non-small cell lung cancer. FASEB J. 2004;18:206–208. doi: 10.1096/fj.03-0369fje. [DOI] [PubMed] [Google Scholar]
  • 84.Lindgren JA, Stenke L, Mansour M, Edenius C, Lauren L, Nasman-Glaser B, Ericsson I, Reizenstein P. Formation and effects of leukotrienes and lipoxins in human bone marrow. J Lipid Mediat. 1993;6:313–320. [PubMed] [Google Scholar]
  • 85.Dupuis F, Desplat V, Praloran V, Denizot Y. Effects of lipidic mediators on the growth of human myeloid and erythroid marrow progenitors. J Lipid Mediat Cell Signal. 1997;16:117–125. doi: 10.1016/s0929-7855(97)00007-2. [DOI] [PubMed] [Google Scholar]
  • 86.Bautz F, Denzlinger C, Kanz L, Mohle R. Chemotaxis and transendothelial migration of CD34(+) hematopoietic progenitor cells induced by the inflammatory mediator leukotriene D4 are mediated by the 7-transmembrane receptor CysLT1. Blood. 2001 Jun 1;97:3433–3440. doi: 10.1182/blood.v97.11.3433. [DOI] [PubMed] [Google Scholar]
  • 87.Claesson HE, Dahlberg N, Gahrton G. Stimulation of human myelopoiesis by leukotriene B4. Biochem Biophys Res Commun. 1985 Sep 16;131:579–585. doi: 10.1016/0006-291x(85)91276-8. [DOI] [PubMed] [Google Scholar]
  • 88.Mohle R, Bautz F, Rafii S, Moore MA, Brugger W, Kanz L. The chemokine receptor CXCR-4 is expressed on CD34+ hematopoietic progenitors and leukemic cells and mediates transendothelial migration induced by stromal cell-derived factor-1. Blood. 1998 Jun 15;91:4523–4530. [PubMed] [Google Scholar]
  • 89.Vore SJ, Eling TE, Danilowicz M, Tucker AN, Luster MI. Regulation of murine hematopoiesis by arachidonic acid metabolites. Int J Immunopharmacol. 1989;11:435–442. doi: 10.1016/0192-0561(89)90171-9. [DOI] [PubMed] [Google Scholar]
  • 90.Braccioni F, Dorman SC, O'byrne PM, Inman MD, Denburg JA, Parameswaran K, Baatjes AJ, Foley R, Gauvreau GM. The effect of cysteinyl leukotrienes on growth of eosinophil progenitors from peripheral blood and bone marrow of atopic subjects. J Allergy Clin Immunol. 2002;110:96–101. doi: 10.1067/mai.2002.125000. [DOI] [PubMed] [Google Scholar]
  • 91.Elsas PX, Queto T, Mendonca-Sales SC, Elsas MI, Kanaoka Y, Lam BK. Cysteinyl leukotrienes mediate the enhancing effects of indomethacin and aspirin on eosinophil production in murine bone marrow cultures. Br J Pharmacol. 2008;153:528–535. doi: 10.1038/sj.bjp.0707586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Kozubik A, Hofmanova J, Pospisil M, Netikova J, Hola J, Lojek A. Effects of drugs inhibiting prostaglandin or leukotriene biosynthesis on postirradiation haematopoiesis in mouse. Int J Radiat Biol. 1994;65:369–377. doi: 10.1080/09553009414550431. [DOI] [PubMed] [Google Scholar]
  • 93.Estrov Z, Halperin DS, Coceani F, Freedman MH. Modulation of human marrow haematopoiesis by leucotrienes in vitro. Br J Haematol. 1988;69:321–327. doi: 10.1111/j.1365-2141.1988.tb02369.x. [DOI] [PubMed] [Google Scholar]
  • 94.Chung JW, Kim GY, Mun YC, Ahn JY, Seong CM, Kim JH. Leukotriene B4 pathway regulates the fate of the hematopoietic stem cells. Exp Mol Med. 2005 Feb 28;37:45–50. doi: 10.1038/emm.2005.6. [DOI] [PubMed] [Google Scholar]
  • 95.Kinder M, Wei C, Shelat SG, Kundu M, Zhao L, Blair IA, Pure E. Hematopoietic stem cell function requires 12/15-lipoxygenase-dependent fatty acid metabolism. Blood. 2010 Jun 17;115:5012–5022. doi: 10.1182/blood-2009-09-243139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Croxford JL, Yamamura T. Cannabinoids and the immune system: potential for the treatment of inflammatory diseases? J Neuroimmunol. 2005;166:3–18. doi: 10.1016/j.jneuroim.2005.04.023. [DOI] [PubMed] [Google Scholar]
  • 97.Klein TW, Newton C, Larsen K, Lu L, Perkins I, Nong L, Friedman H. The cannabinoid system and immune modulation. J Leukoc Biol. 2003;74:486–496. doi: 10.1189/jlb.0303101. [DOI] [PubMed] [Google Scholar]
  • 98.Valk PJ, Verbakel S, Vankan Y, Hol S, Mancham S, Ploemacher R, Mayen A, Lowenberg B, Delwel R. Anandamide, a natural ligand for the peripheral cannabinoid receptor is a novel synergistic growth factor for hematopoietic cells. Blood. 1997 Aug 15;90:1448–1457. [PubMed] [Google Scholar]
  • 99.Bentzen PJ, Lang F. Effect of anandamide on erythrocyte survival. Cell Physiol Biochem. 2007;20:1033–1042. doi: 10.1159/000110714. [DOI] [PubMed] [Google Scholar]
  • 100.Jorda MA, Verbakel SE, Valk PJ, Vankan-Berkhoudt YV, Maccarrone M, Finazzi-Agro A, Lowenberg B, Delwel R. Hematopoietic cells expressing the peripheral cannabinoid receptor migrate in response to the endocannabinoid 2-arachidonoylglycerol. Blood. 2002 Apr 15;99:2786–2793. doi: 10.1182/blood.v99.8.2786. [DOI] [PubMed] [Google Scholar]
  • 101.Patinkin D, Milman G, Breuer A, Fride E, Mechoulam R. Endocannabinoids as positive or negative factors in hematopoietic cell migration and differentiation. Eur J Pharmacol. 2008 Oct 24;595:1–6. doi: 10.1016/j.ejphar.2008.05.002. [DOI] [PubMed] [Google Scholar]
  • 102.Jiang S, Fu Y, Williams J, Wood J, Pandarinathan L, Avraham S, Makriyannis A, Avraham S, Avraham HK. Expression and function of cannabinoid receptors CB1 and CB2 and their cognate cannabinoid ligands in murine embryonic stem cells. PLoS One. 2007;2:e641. doi: 10.1371/journal.pone.0000641. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Rizzo MT. The role of arachidonic acid in normal and malignant hematopoiesis. Prostaglandins Leukot Essent Fatty Acids. 2002;66:57–69. doi: 10.1054/plef.2001.0331. [DOI] [PubMed] [Google Scholar]
  • 104.Stenke L, Mansour M, Edenius C, Reizenstein P, Lindgren JA. Formation and proliferative effects of lipoxins in human bone marrow. Biochem Biophys Res Commun. 1991 Oct 15;180:255–261. doi: 10.1016/s0006-291x(05)81285-9. [DOI] [PubMed] [Google Scholar]
  • 105.Rocca B, Secchiero P, Celeghini C, Ranelletti FO, Ciabattoni G, Maggiano N, Habib A, Ricerca BM, Barbarotto E, Patrono C, Zauli G. Modulation of the expression and activity of cyclooxygenases in normal and accelerated erythropoiesis. Exp Hematol. 2004;32:925–934. doi: 10.1016/j.exphem.2004.07.010. [DOI] [PubMed] [Google Scholar]
  • 106.Abraham NG, Feldman E, Falck JR, Lutton JD, Schwartzman ML. Modulation of erythropoiesis by novel human bone marrow cytochrome P450-dependent metabolites of arachidonic acid. Blood. 1991 Sep 15;78:1461–1466. [PubMed] [Google Scholar]
  • 107.Guo AM, Janic B, Sheng J, Falck JR, Roman RJ, Edwards PA, Arbab AS, Scicli AG. The CYP4A/F-20-HETE System in Regulation of Endothelial Precursor Cell Derived from Human Umbilical Cord Blood. J Pharmacol Exp Ther. 2011 Apr 28; doi: 10.1124/jpet.111.179036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Lands WE. Biochemistry and physiology of n-3 fatty acids. FASEB J. 1992;6:2530–2536. doi: 10.1096/fasebj.6.8.1592205. [DOI] [PubMed] [Google Scholar]
  • 109.Varney ME, Buchanan JT, Dementieva Y, Hardman WE, Sollars VE. A high omega-3 fatty acid diet has different effects on early and late stage myeloid progenitors. Lipids. 2011;46:47–57. doi: 10.1007/s11745-010-3491-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Hoggatt J, Pelus LM. Eicosanoid regulation of hematopoiesis and hematopoietic stem and progenitor trafficking. Leukemia. 2010;24:1993–2002. doi: 10.1038/leu.2010.216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Huang SK, Peters-Golden M. Eicosanoid lipid mediators in fibrotic lung diseases: ready for prime time? Chest. 2008;133:1442–1450. doi: 10.1378/chest.08-0306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Guerrero AT, Verri WA, Jr, Cunha TM, Silva TA, Schivo IR, Dal-Secco D, Canetti C, Rocha FA, Parada CA, Cunha FQ, Ferreira SH. Involvement of LTB4 in zymosan-induced joint nociception in mice: participation of neutrophils and PGE2. J Leukoc Biol. 2008;83:122–130. doi: 10.1189/jlb.0207123. [DOI] [PubMed] [Google Scholar]
  • 113.Coopman K, Smith LD, Wright KL, Ward SG. Temporal variation in CB2R levels following T lymphocyte activation: evidence that cannabinoids modulate CXCL12-induced chemotaxis. Int Immunopharmacol. 2007;7:360–371. doi: 10.1016/j.intimp.2006.11.008. [DOI] [PubMed] [Google Scholar]
  • 114.Ghosh S, Preet A, Groopman JE, Ganju RK. Cannabinoid receptor CB2 modulates the CXCL12/CXCR4-mediated chemotaxis of T lymphocytes. Mol Immunol. 2006;43:2169–2179. doi: 10.1016/j.molimm.2006.01.005. [DOI] [PubMed] [Google Scholar]
  • 115.Nilsson O, Fowler CJ, Jacobsson SO. The cannabinoid agonist WIN 55,212-2 inhibits TNF-alpha-induced neutrophil transmigration across ECV304 cells. Eur J Pharmacol. 2006 Oct 10;547:165–173. doi: 10.1016/j.ejphar.2006.07.016. [DOI] [PubMed] [Google Scholar]
  • 116.Kurihara R, Tohyama Y, Matsusaka S, Naruse H, Kinoshita E, Tsujioka T, Katsumata Y, Yamamura H. Effects of peripheral cannabinoid receptor ligands on motility and polarization in neutrophil-like HL60 cells and human neutrophils. J Biol Chem. 2006 May 5;281:12908–12918. doi: 10.1074/jbc.M510871200. [DOI] [PubMed] [Google Scholar]
  • 117.Jiang S, Zagozdzon R, Jorda MA, Parmar K, Fu Y, Williams JS, Wood JA, Makriyannis A, Banu N, Avraham S, Groopman JE, Avraham HK. Endocannabinoids are expressed in bone marrow stromal niches and play a role in interactions of hematopoietic stem and progenitor cells with the bone marrow microenvironment. J Biol Chem. 2010 Nov 12;285:35471–35478. doi: 10.1074/jbc.M110.144758. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 118.Jiang S, Alberich-Jorda M, Zagozdzon R, Parmar K, Fu Y, Mauch P, Banu N, Makriyannis A, Tenen DG, Avraham S, Groopman JE, Avraham HK. Cannabinoid receptor 2 and its agonists mediate hematopoiesis and hematopoietic stem and progenitor cell mobilization. Blood. 2011 Jan 20;117:827–838. doi: 10.1182/blood-2010-01-265082. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 119.Durand EM, Zon LI. Newly emerging roles for prostaglandin E2 regulation of hematopoiesis and hematopoietic stem cell engraftment. Curr Opin Hematol. 2010;17:308–312. doi: 10.1097/MOH.0b013e32833a888c. [DOI] [PubMed] [Google Scholar]

RESOURCES