Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Jul 1.
Published in final edited form as: Curr Opin Hematol. 2011 Jul;18(4):231–238. doi: 10.1097/MOH.0b013e3283477962

Many Mechanisms Mediating Mobilization: An Alliterative Review

Jonathan Hoggatt 1, Louis M Pelus 1
PMCID: PMC3159187  NIHMSID: NIHMS313358  PMID: 21537168

Abstract

Purpose of review

Blood cell production is maintained by hematopoietic stem cells (HSC) that reside in specialized niches within bone marrow. Treatment with granulocyte-colony stimulating factor (G-CSF) causes HSC egress from bone marrow niches and trafficking to the peripheral blood, a process termed “mobilization”. Although the mobilization phenomenon has been known for some time and is utilized clinically to acquire HSC for transplant, the mechanisms mediating HSC release are not completely understood. We discuss recent advances and controversies in defining mechanisms responsible for G-CSF induced mobilization.

Recent findings

New reports define a role for resident monocytes/macrophages in maintaining niche cells, which is diminished after G-CSF treatment, suggesting a new mechanism for mobilization. While osteoblasts have been reported to be a primary component of the HSC niche, new results suggest a unique niche composed of innervated mesenchymal stem cells. Modulating bioactive lipid signaling also facilitates mobilization, and may define a future therapeutic strategy.

Summary

Hematopoietic mobilization by G-CSF is primarily mediated by alterations to the bone marrow niche by both direct and indirect mechanisms, rather than directly altering HSC function. Further understanding of the processes mediating mobilization will advance our understanding on the cellular and molecular components of the HSC niche.

Keywords: Hematopoietic stem cell, mobilization, niche, G-CSF, microenvironment

Introduction

Hematopoietic stem cells (HSC) reside within specialized bone marrow niches, “tethered” through adhesion molecule interactions to a complex of stromal cells, endosteal lining osteoblasts, and mesenchymal stem cells within a matrix of collagens, fibronectin, and proteoglycans, where they produce mature cells that ultimately exit the marrow and enter the peripheral blood. HSC and hematopoietic progenitor cells (HPC) also traffic to the peripheral blood [16], leaving open niches that can be repopulated by transplanted HSC [7*]. Based on observations that increased circulating HPC were found in patients after chemotherapy [8,9], we now know that this natural trafficking of HSC and HPC can be modulated, allowing for directed “mobilization” [3,4]. Mobilization is widely used to acquire hematopoietic cells for autologous and allogeneic transplantation, achieved through administration of chemotherapy [810], hematopoietic growth factors, chemokines, or small molecule inhibitors or antibodies against niche chemokine receptors and integrins [1113].

Granulocyte-colony stimulating factor (G-CSF) (NeupogenR) is widely used clinically to mobilize HSC and HPC for transplantation. G-CSF-mobilized HSC and HPC are associated with more rapid engraftment, shorter hospital stay [1417], and in some circumstances, superior overall survival compared to bone marrow [18]. The mobilization process is not well understood and many mechanisms mediating the effect have been proposed. This alliterative review focuses on recent findings that add to our understanding of the G-CSF mobilization response, but also introduces new controversies and questions.

CXCR4 controls (hematopoietic) cell circulation

The CXC4 receptor (CXCR4) and its ligand stromal cell-derived factor-1α (SDF-1α) are the most widely explored hematopoietic niche interaction regulating HSC and HPC retention and trafficking. Hematopoietic cells express CXCR4 and are chemo-attracted to and retained within the bone marrow by SDF-1α [1921]. Genetic knockout of either CXCR4 [22] or SDF-1α [23] is embryonic lethal in mice, with a failure to populate the bone marrow niche during development. Conditional deletion of either CXCR4 [24] or SDF-1α [25*] results in a substantial hematopoietic cell egress from the bone marrow and impaired marrow retention of gene deleted HSC and HPC after transplantation [25*,26].

While numerous reports support a key role for the CXCR4/SDF-1α axis in hematopoietic cell retention/trafficking/mobilization within the bone marrow niche, the predominate source of SDF-1α is still not clear. Osteoblasts [27], reticular cells found in endosteal and vascular niches [28], endothelial cells, and bone itself [29,30] all produce/express SDF-1α. Recently, Mendez-Ferrer and colleagues described a novel population of nestin+ mesenchymal stem cells (MSC) that express high levels of SDF-1α mRNA and suggest they form a unique hematopoietic niche [31**]. Early reports demonstrated that osteoblast SDF-1α is reduced after G-CSF treatment [30,32,33], suggesting that osteoblast-derived SDF-1α was a key regulator of hematopoietic cell retention/mobilization. However, other studies question the importance of osteoblast-derived SDF-1α [28,31**,34]. Christopher et al. recently showed reduced SDF-1α production in Col2.3-expressing osteoblasts with no reduction in Col2.3 negative stromal cells, suggesting that reduced osteoblast SDF-1α is a common mechanism of cytokine-induced mobilization [35*]. However, Mendez-Ferrer et al. [31**], using a similar approach, showed substantially reduced SDF-1α in nestin+ MSC relative to a similar population of stromal cells described by Christopher et al. [35*], although a direct comparison to defined osteoblasts was not made. This is similar to findings that a mesenchymal precursor cell may form the hematopoietic niche [36*]. Overall, these results suggest that heterogeneous cell populations expressing SDF-1α and other supportive factors create unique niches, each of which can be manipulated to facilitate mobilization. Defining the specific niche(s) responsible for SDF-1α production and HSC retention will aid in development of future therapeutic strategies.

Osteoblasts, osteoclasts, osteomacs and other operators

Osteoblasts express signaling molecules in addition to SDF-1α that may regulate HSC function and niche retention [3740]. Targeting the interaction between osteoblast vascular cell adhesion molecule-1 (VCAM-1) and HSC very late antigen-4 (VLA-4) with antibodies against VLA-4 [41,42] or VCAM-1 [43,44], or a small molecule inhibitor of VLA-4 (BIO5192) [45*], results in mobilization. Osteoblasts also express significant amounts of osteopontin (OPN), and HSC adhere to OPN via β1 integrins [46]. Intriguingly, OPN also negatively regulates HSC pool size within the marrow niche [46,47], and OPN knockout mice show enhanced endogenous and G-CSF-induced mobilization [48**].

Hematopoietic mobilization with G-CSF results in suppression of niche osteoblasts [30,33,49**], with increased osteoblast apoptosis [33] and a characteristic osteoblast "flattening" [30], and decreased endosteal niche expression of retention molecules. This suppression has been reported to result from altered sympathetic nervous system (SNS) signaling to osteoblasts [30] (discussed below), however recent reports also suggest an alternate, perhaps parallel, pathway involving bone marrow macrophages. Winkler et al. reported that the marrow niche contains supportive endosteal lining “osteomacs”, and that G-CSF treatment results in a trafficking and reduction of these osteomacs, causing osteoblast suppression [49**]. Macrophage depletion using Mafia transgenic mice or by treatment with clodronate-loaded liposomes (Clo-lip), resulted in hematopoietic mobilization. The osteomac population of cells was characterized as F4/80+ Ly-6G+ CD11b+, largely based on findings that this was the predominate population depleted after Clo-lip treatment.

Two recent reports also suggest a role for resident macrophages/monocytes in mobilization. Chow et al. also demonstrated that macrophage depletion with Clo-lip results in mobilization [50**]; however, in slight contrast to osteomacs, these cells were defined as Gr-1negative F4/80+ CD115mid CD169+. In an animal model where CD169+ cells are specifically depleted, mobilization was enhanced. The authors suggest that CD169+ macrophages express a soluble, yet to be identified, factor that supports niche cells, specifically nestin+ MSC. Similarly, Christopher et al. utilized G-CSF receptor (G-CSFR) knockout mice crossbred to mice expressing G-CSFR under the control of the CD68 (macrosialin) promoter [51**], which restricts G-CSFR expression to the monocyte/macrophage lineages. Treatment of these mice with G-CSF showed a reduction in marrow macrophages, coincident with HPC mobilization. Intriguingly, we previously reported that treatment of mice with an anti-Gr-1 antibody to deplete neutrophils significantly reduced the G-CSF mobilization response [52], yet the animal model used by Christopher et al. showed mobilization to G-CSF despite neutropenia. As noted by Christopher et al., the antibody we used to deplete neutrophils (clone RB6-8C5) would also target monocyte and osteomac populations, possibly accounting for the differing effects seen. However, if this were the case, one would expect increased mobilization due to depletion of these populations, not decreased mobilization. In addition, the anti-Gr-1 antibody would not target the CD169+ macrophages [50**]. More likely, mobilization is a complex process coordinated by a balance between cell populations in the bone marrow rather than specifically a function of a single cell lineage. Thus, model systems that target specific populations would be sufficient to induce mobilization, but not necessarily exclusive. Clearly, the cellular players and factors warrant further exploration.

As a prime example of cellular balance, osteoblasts and osteoclasts regulate bone formation/bone resorption, within the bone marrow niche. Kollet et al. reported that RANK ligand treatment, which increases osteoclast activity, produces a moderate mobilization of HPC [53]. Similar results are also seen in an independent report [54*]. Correspondingly, stress models such as bleeding or LPS treatment increased the number of osteoclasts that was coincident with HPC mobilization. G-CSF mobilization was decreased after inhibition of osteoclasts, by treatment with calcitonin or using a genetic knockout model of PTPε, further suggesting osteoclast involvement in G-CSF-mediated mobilization. The authors proposed that osteoclast-derived proteolytic enzymes, such as Cathepsin K, degrade important niche interaction components including SDF-1α and OPN [53]. In a more recent study they demonstrated reduced osteoclast maturation and activity in CD45 knockout mice that correlated with reduced mobilization to RANK ligand and G-CSF [55].

In contrast, an earlier report demonstrated that while G-CSF increases osteoclast number and bone resorption in both BALB/c mice and humans, the increase in osteoclasts did not occur until 10–15 days or 6–8 days, respectively, after treatment with G-CSF [56], a finding also observed by others using similar systems [32,57]. Since G-CSF mobilization is typically evaluated after 4 to 5 days, the importance of osteoclasts to G-CSF induced mobilization remains unclear. Furthermore, treatment of mice with bisphosphonates, which inhibit osteoclast activity and/or number, prior to G-CSF administration does not result in impaired mobilization [49**,56], and in fact, in one case, bisphosphonate treatment increased mobilization [49**]. Of note, the endosteal bone surface, particularly underneath resorbing osteoclasts, is a significant source of extracellular calcium and studies by Adams et al. demonstrated that HSC expression of calcium sensing receptors mediates chemoattraction to soluble Ca2+ [58]. Calcium sensing receptor knockout mice had reduced marrow HSC content and increased peripheral blood HSC, perhaps suggesting that increased G-CSF mobilization seen in zoldronate treated mice resulted from reduced calcium retention signaling within the niche. While increased osteoclast activity can clearly induce mobilization, their role in G-CSF-mediated mobilization is not sufficiently defined and may not be a primary mechanism of mobilization. Figure 1 summarizes some of the key cellular components involved in HSC retention and subsequent mobilization following G-CSF treatment.

Figure 1.

Figure 1

Schematic representation of HSC niche retention and G-CSF induced mobilization. At steady state (left panel), HSC reside in bone marrow niches, closely associated with cells such as osteoblasts or nestin+ MSC that express SDF-1α and other supportive factors. Resident macrophages provide a positive supporting factor maintaining osteoblast and MSC activity. During G-CSF mobilization (right panel) G-CSF acts directly on the resident macrophages, causing them to transit away from the endosteal niche and reduces their number, and alters SNS signaling. This results in an attenuation of osteoblast function and a characteristic flattening, and reduction of supportive factors like SDF-1α in the marrow, with an increase in plasma SDF-1α. The role of both activated osteoclasts and neutrophils still remains unclear.

Hypoxia harbors hematopoiesis

Hematopoietic stem cells reside in hypoxic niches within the bone marrow [5961], and HSC that reside in hypoxic niches have greater hematopoietic repopulating ability [62*]. Stabilization of the transcription factor hypoxia inducible factor 1-α (HIF-1α) is a known physiological response to hypoxia and HIF-1α has been shown to up-regulate erythropoietin (EPO) production [63]; numerous cell proliferation and survival genes [64-66]; the angiogenic growth factor VEGF [67]; and other genes. The hypoxic niche may maintain HIF-1α activity thereby maintaining HSC [68], a hypothesis supported by the fact that hypoxic conditions expand human HSC [69] and HPC populations [7072] in vitro. In response to G-CSF, HIF-1α expression is increased [73*] and both the hypoxic environment and HIF-1α expand within the marrow compartment [74], with increased VEGF production; however, marrow vascular density and permeability are not increased [62]. In addition, HIF-1α is stabilized in peripheral blood HSC acquired from G-CSF mobilized donors [75]. HIF-1α also increases SDF-1α production [76] and CXCR4 expression [77], suggesting that hypoxia may be a physiological regulator of this important signaling axis. Of note, one report suggests that decreased osteoblast SDF-1α expression and reduced CXCR4 expression on bone marrow cells can be facilitated by osteoclasts activated by the hypoxia-mimetic CoCl2 in vitro [54]. Recently, a population of marrow-derived very small embryonic-like stem cells was reported to mobilize as a result of intermittent hypoxia, coincident with an increase in plasma SDF-1α [78*], further suggesting that hypoxia may regulate HSC retention/trafficking.

HIF-1α also prevents hematopoietic cell damage caused by reactive oxygen species (ROS) [79*], suggesting that the hypoxic niche helps maintain HSC lifespan. In slight contrast, another report demonstrated that enhanced c-Met activity promotes mobilization by activating mTOR and increasing ROS production in HSC and HPC [80**], while inhibition of mTOR with rapamycin reduced HSC mobilization [80**,81], suggesting that a small amount of ROS may be necessary for optimal mobilization. Knockout of the thioredoxin-interacting protein gene results in increased mobilization under stress conditions [82*], suggesting a role for oxygen tension and ROS in mobilization.

Nervous Niche

The marrow microenvironment is highly innervated [83], and catecholamine signaling can alter numerous physiological processes of immune cells [84]. A seminal study by Katayama et al. demonstrated that G-CSF mobilization was reduced in chemically sympathectomized mice; mice treated with the β-blocker propanolol; or mice genetically deficient in the gene for dopamine β-hydroxylase (Dbh), an enzyme that converts dopamine into norepinephrine, demonstrating that mobilization requires peripheral β2-adrenergic signals [30]. This study also demonstrated that G-CSF attenuated osteoblast function, via the sympathetic nervous system (SNS), resulting in osteoblasts having a marked flattened appearance. Intriguingly, β2-adrenergic signaling can regulate osteoclast differentiation [85] and nestin+ MSC are highly innervated [31], perhaps suggesting that SNS signaling alters niche cell components through multiple mechanisms. In addition to the niche, human CD34+ cells express β2-adrenergic and dopamine receptors that are upregulated by G-CSF [86], and neurotransmitters serve as direct chemoattractants to hematopoietic cells [86] and increase CXCR4 expression [87]. Epinephrine treatment also results in mobilization [86].

Signaling from β3-adrenergic receptors regulate circadian oscillations mediating norepinephrine release, CXCR4 expression, and SDF-1α production, leading to rhythmic egress of hematopoietic cells from the niche [88,89]. Both β2 and β3-adrenergic signals cooperate to regulate G-CSF-induced mobilization, although knockout of both only partially blocks mobilization [90*] and does not abrogate mobilization by Clo-lip [50], suggesting multiple/parallel pathways regulating mobilization. β2-adrenergic signaling also up-regulates osteoblast vitamin D receptors (VDR) and it was recently demonstrated that expression of VDR is necessary for the G-CSF-induced suppression of osteoblast function and that HSC mobilization is reduced in VDR knockout mice [91*]. VDR is also regulated by circadian rhythms [92], demonstrating further complexity in interconnected mobilization mechanisms.

Fatty acids for the future

Studies exploring the role of SNS signaling in G-CSF-mediated mobilization began in mice defective in ceramide galactosyltransferase (CGT), an enzyme that synthesizes galactocerebrosides, major lipid components of myelin sheaths and are important for proper nerve conduction [30]. Mice deficient in the related enzyme galactocerbrosidase (GALC) have a defective niche in which transplanted HSC fail to engraft [93*]. Sphingosine-1-phosphate (S1P) and ceramide, which affect a wide variety of cellular processes, are part of the GALC metabolic pathway [94]. HSC and HPC express the S1P receptor S1P1 [95], and S1P1 signaling alters CXCR4/SDF-1α signaling and chemotaxis [9597]. S1P directs trafficking of immature B cells [98*] and trafficking of HSC and HPC from blood, bone marrow and lymph tissues [1]. A recent report by Ratajczak et al. suggests that plasma S1P increases following G-CSF administration and directs peripheral chemoattraction, facilitating mobilization [99**]. An increase in S1P in peripheral blood coordinate with a decrease in marrow was recently reported, suggesting the formation of an S1P gradient driving mobilization [100], or perhaps even a dual lipid action mediate by both S1P and ceramide concentrations in peripheral blood and bone, respectively [101]. If S1P signaling through S1P1 regulates G-CSF mobilization, then co-treatment with an S1P1 antagonist, like FTY720, would be expected to reduce G-CSF mobilization. This was indeed reported in one case [100], however others report no decreases in G-CSF mobilization in FTY720-treated mice [1,102]. Further studies evaluating the role of S1P and ceramide will aid in determining their functional importance in hematopoietic mobilization, and whether agents modifying S1P1 signaling, like the S1P agonist SEW2871, can be utilized as an adjunct therapeutic agent in mobilization.

In addition to the sphingolipids, other bioactive lipids have the capacity to regulate hematopoiesis, including eicosanoids. We recently reported that prostaglandin E2 (PGE2) regulates CXCR4 expression on HSC and HPC and facilitates their chemoattraction to SDF-1α and homing [103]. Like many lipid systems, homeostasis is maintained by a balance of signaling amongst eicosanoids with several of the eicosanoids acting in opposing manners. We also showed that agonism of cannabinoid receptors, receptors for the eicosanoid-related endocannabinoids, act in an opposing fashion to PGE2 signaling, decreasing adhesion molecule expression and CXCR4, and enhancing G-CSF mobilization [104**]. Two separate reports by Jiang et al. have also demonstrated that cannabinoid signaling mediates mobilization, and that endocannabinoids are expressed in bone marrow and increase HPC migration and proliferation in vitro [105**,106*]. Another report demonstrated that cannabinoids mobilize myeloid-derived suppressor cells, with a possible role for endogenous G-CSF production [107*]. It is interesting to note that cannabinoid receptors are physically associated with β2-adrenergic receptors and modulate adrenergic signaling [108*,109*], possibly suggesting a SNS/cannabinoid mechanistic link. With the abundance of FDA approved compounds modulating eicosanoid signaling, additional studies exploring their role in mobilization are likely to lead to new therapeutic strategies.

Conclusion

While significant progress has been made in defining the mechanisms mediating mobilization by G-CSF, a complete understanding remains elusive. Do osteoblasts cells comprise the important hematopoietic niche suppressed by G-CSF administration, or is it innervated nestin+ MSC, or a heterogeneous population of cells forming several unique niches? Experimental animal models designed to explore mobilization have significantly advanced the field, but manipulation of a specific systems while sufficient to mimic or block mobilization by G-CSF, may not tell the whole story. While both genetic alteration of the SNS or macrophages can mediate mobilization, neither regulates the niche exclusively during mobilization and other parallel or intersecting pathways are likely involved. Combination therapies manipulating multiple mechanistic pathways should be explored to not only maximally mobilize hematopoietic cells for transplant, but possibly to mobilize other potential therapeutically efficacious cell populations.

Key Points.

  • G-CSF reduces resident monocytes/macrophages, causing niche suppression and mobilization.

  • The niche consists of a heterogeneous population of cells that can coordinately be altered during mobilization.

  • Sphingosine-1-phosphate, endocannabinoids and other bioactive lipids mediate HSC retention and trafficking to and from the bone marrow.

  • The mechanisms regulating G-CS- induced mobilization alter multiple, parallel mechanistic pathways, suggesting combinatorial therapies should be explored.

Acknowledgments

Supported by NIH grants HL069669 and HL096305 (to LMP). JH is supported by training grant HL007910.

References

  • 1.Massberg S, Schaerli P, Knezevic-Maramica I, et al. Immunosurveillance by hematopoietic progenitor cells trafficking through blood, lymph, and peripheral tissues. Cell. 2007;131:994–1008. doi: 10.1016/j.cell.2007.09.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.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]
  • 3.Wright DE, Wagers AJ, Gulati AP, et al. Physiological migration of hematopoietic stem and progenitor cells. Science. 2001;294:1933–1936. doi: 10.1126/science.1064081. [DOI] [PubMed] [Google Scholar]
  • 4.Abkowitz JL, Robinson AE, Kale S, et al. Mobilization of hematopoietic stem cells during homeostasis and after cytokine exposure. Blood. 2003;102:1249–1253. doi: 10.1182/blood-2003-01-0318. [DOI] [PubMed] [Google Scholar]
  • 5.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]
  • 6.Goodman JW, Hodgson GS. Evidence for stem cells in the peripheral blood of mice. Blood. 1962;19:702–714. [PubMed] [Google Scholar]
  • *7.Bhattacharya D, Czechowicz A, Ooi AG, et al. Niche recycling through division-independent egress of hematopoietic stem cells. J Exp Med. 2009;206:2837–2850. doi: 10.1084/jem.20090778. This report utilized both stringent phenotypic HSC markers and transplantation assays to demonstrate that 1–5% of the total pool of HSCs enters the circulation each day. They show that daily transplantations of purified HSC engraft stronger than a single bolus transplant, and suggest that HSC niches are transiently filled and unfilled daily. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kurnick JE, Robison WA. Colony growth of human peripheral white blood cells in vitro. Blood. 1971;37:136–141. [PubMed] [Google Scholar]
  • 9.McCredie KB, Hersh EM, Freireich EJ. Cells capable of colony formation in the peripheral blood of man. Science. 1971;171:293–294. doi: 10.1126/science.171.3968.293. [DOI] [PubMed] [Google Scholar]
  • 10.Kessinger A, Armitage JO. The evolving role of autologous peripheral stem cell transplantation following high-dose therapy for malignancies. Blood. 1991;77:211–213. [PubMed] [Google Scholar]
  • 11.Fruehauf S, Seggewiss R. It's moving day: factors affecting peripheral blood stem cell mobilization and strategies for improvement [corrected] Br J Haematol. 2003;122:360–375. doi: 10.1046/j.1365-2141.2003.04483.x. [DOI] [PubMed] [Google Scholar]
  • 12.Papayannopoulou T. Current mechanistic scenarios in hematopoietic stem/progenitor cell mobilization. Blood. 2004;103:1580–1585. doi: 10.1182/blood-2003-05-1595. [DOI] [PubMed] [Google Scholar]
  • 13.To LB, Haylock DN, Simmons PJ, Juttner CA. The biology and clinical uses of blood stem cells. Blood. 1997;89:2233–2258. [PubMed] [Google Scholar]
  • 14.Kennedy MJ, Davis J, Passos-Coelho J, et al. Administration of human recombinant granulocyte colony-stimulating factor (filgrastim) accelerates granulocyte recovery following high-dose chemotherapy and autologous marrow transplantation with 4-hydroperoxycyclophosphamide-purged marrow in women with metastatic breast cancer. Cancer Res. 1993;53:5424–5428. [PubMed] [Google Scholar]
  • 15.McQuaker IG, Hunter AE, Pacey S, et al. Low-dose filgrastim significantly enhances neutrophil recovery following autologous peripheral-blood stem-cell transplantation in patients with lymphoproliferative disorders: evidence for clinical and economic benefit. J Clin Oncol. 1997;15:451–457. doi: 10.1200/JCO.1997.15.2.451. [DOI] [PubMed] [Google Scholar]
  • 16.Jansen J, Thompson EM, Hanks S, et al. Hematopoietic growth factor after autologous peripheral blood transplantation: comparison of G-CSF and GM-CSF. Bone Marrow Transplant. 1999;23:1251–1256. doi: 10.1038/sj.bmt.1701806. [DOI] [PubMed] [Google Scholar]
  • 17.Nemunaitis J, Rosenfeld CS, Ash R, et al. Phase III randomized, double-blind placebo-controlled trial of rhGM-CSF following allogeneic bone marrow transplantation. Bone Marrow Transplant. 1995;15:949–954. [PubMed] [Google Scholar]
  • 18.Stem Cell Trialists' Group. Allogeneic peripheral blood stem-cell compared with bone marrow transplantation in the management of hematologic malignancies: an individual patient data meta-analysis of nine randomized trials. J Clin Oncol. 2005;23:5074–5087. doi: 10.1200/JCO.2005.09.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Aiuti A, Webb IJ, Bleul C, et al. The chemokine SDF-1 is a chemoattractant for human CD34+ hematopoietic progenitor cells and provides a new mechanism to explain the mobilization of CD34+ progenitors to peripheral blood. J Exp Med. 1997;185:111–120. doi: 10.1084/jem.185.1.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Jo DY, Rafii S, Hamada T, Moore MA. Chemotaxis of primitive hematopoietic cells in response to stromal cell-derived factor-1. J Clin Invest. 2000;105:101–111. doi: 10.1172/JCI7954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kim CH, Broxmeyer HE. In vitro behavior of hematopoietic progenitor cells under the influence of chemoattractants: stromal cell-derived factor-1, steel factor, and the bone marrow environment. Blood. 1998;91:100–110. [PubMed] [Google Scholar]
  • 22.Zou YR, Kottmann AH, Kuroda M, et al. Function of the chemokine receptor CXCR4 in haematopoiesis and in cerebellar development. Nature. 1998;393:595–599. doi: 10.1038/31269. [DOI] [PubMed] [Google Scholar]
  • 23.Nagasawa T, Hirota S, Tachibaba K, et al. Defects of B-cell lymphopoiesis and bone-marrow myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1. Nature. 1996;382:635–638. doi: 10.1038/382635a0. [DOI] [PubMed] [Google Scholar]
  • 24.Nie Y, Han YC, Zou YR. CXCR4 is required for the quiescence of primitive hematopoietic cells. J Exp Med. 2008;205:777–783. doi: 10.1084/jem.20072513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *25.Tzeng YS, Li H, Kang YL, et al. Loss of Cxcl12/Sdf-1 in adult mice decreases the quiescent state of hematopoietic stem/progenitor cells and alters the pattern of hematopoietic regeneration after myelosuppression. Blood. 2011;117:429–439. doi: 10.1182/blood-2010-01-266833. The authors developed a conditional mouse, allowing for inducible deletion of SDF-1α in adult mice. When deleted, HSC lose quiescence and egress away from endosteal niches. [DOI] [PubMed] [Google Scholar]
  • 26.Foudi A, Jarrier P, Zhang Y, et al. Reduced retention of radioprotective hematopoietic cells within the bone marrow microenvironment in CXCR4−/− chimeric mice. Blood. 2006;107:2243–2251. doi: 10.1182/blood-2005-02-0581. [DOI] [PubMed] [Google Scholar]
  • 27.Ponomaryov T, Peled A, Petit I, et al. Induction of the chemokine stromal-derived factor-1 following DNA damage improves human stem cell function. J Clin Invest. 2000;106:1331–1339. doi: 10.1172/JCI10329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Sugiyama T, Kohara H, Noda M, Nagasawa T. Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity. 2006;25:977–988. doi: 10.1016/j.immuni.2006.10.016. [DOI] [PubMed] [Google Scholar]
  • 29.Sipkins DA, Wei X, Wu JW, et al. In vivo imaging of specialized bone marrow endothelial microdomains for tumour engraftment. Nature. 2005;435:969–973. doi: 10.1038/nature03703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Katayama Y, Battista M, Kao WM, et al. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell. 2006;124:407–421. doi: 10.1016/j.cell.2005.10.041. [DOI] [PubMed] [Google Scholar]
  • **31.Mendez-Ferrer S, Michurina TV, Ferraro F, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010;466:829–834. doi: 10.1038/nature09262. This is the first report to describe a novel population of MSC that express the neuronal marker nestin. Using nestin-GFP mice, the authors demonstrate that labeled HSC, when transplanted, co-localize to nestin+ MSC, and demonstrate that nestin+ MSC express high levels of SDF-1α and other supportive HSC factors, which are significantly reduced after mobilization. These results suggest a unique bone marrow niche for HSC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Christopher MJ, Link DC. Granulocyte colony-stimulating factor induces osteoblast apoptosis and inhibits osteoblast differentiation. J Bone Miner Res. 2008;23:1765–1774. doi: 10.1359/JBMR.080612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Semerad CL, Christopher MJ, Liu F, et al. G-CSF potently inhibits osteoblast activity and CXCL12 mRNA expression in the bone marrow. Blood. 2005;106:3020–3027. doi: 10.1182/blood-2004-01-0272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Tokoyoda K, Egawa T, Sugiyama T, et al. Cellular niches controlling B lymphocyte behavior within bone marrow during development. Immunity. 2004;20:707–718. doi: 10.1016/j.immuni.2004.05.001. [DOI] [PubMed] [Google Scholar]
  • *35.Christopher MJ, Liu F, Hilton MJ, et al. Suppression of CXCL12 production by bone marrow osteoblasts is a common and critical pathway for cytokine-induced mobilization. Blood. 2009;114:1331–1339. doi: 10.1182/blood-2008-10-184754. This report demonstrates that osteoblasts reduce expression of SDF-1α in response to mobilizing cytokines, including Flt-3, SCF, and G-CSF. In addition, using chimeric animals, the authors show that CXCR4−/− HSC do not mobilize in response to G-CSF. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *36.Omatsu Y, Sugiyama T, Kohara H, et al. The essential functions of adipo-osteogenic progenitors as the hematopoietic stem and progenitor cell niche. Immunity. 2010;33:387–399. doi: 10.1016/j.immuni.2010.08.017. The authors developed an animal model in which a CXCL12 (SDF-1α) abundant reticular (CAR) could be inducibley ablated, causing reduced HSC number and early myeloid differentiation, and a reduction in SCF and SDF-1α. The CAR cells were shown to have both adipogenic and osteogenic potential, and the authors suggest these cells form an HSC niche. [DOI] [PubMed] [Google Scholar]
  • 37.Calvi LM, Adams GB, Weibrecht KW, et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 2003;425:841–846. doi: 10.1038/nature02040. [DOI] [PubMed] [Google Scholar]
  • 38.Zhang J, Niu C, Ye L, et al. Identification of the haematopoietic stem cell niche and control of the niche size. Nature. 2003;425:836–841. doi: 10.1038/nature02041. [DOI] [PubMed] [Google Scholar]
  • 39.Visnjic D, Kalajzic Z, Rowe DW, et al. Hematopoiesis is severely altered in mice with an induced osteoblast deficiency. Blood. 2004;103:3258–3264. doi: 10.1182/blood-2003-11-4011. [DOI] [PubMed] [Google Scholar]
  • 40.Arai F, Hirao A, Ohmura M, et al. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell. 2004;118:149–161. doi: 10.1016/j.cell.2004.07.004. [DOI] [PubMed] [Google Scholar]
  • 41.Craddock CF, Nakamoto B, Andrews RG, et al. Antibodies to VLA4 integrin mobilize long-term repopulating cells and augment cytokine-induced mobilization in primates and mice. Blood. 1997;90:4779–4788. [PubMed] [Google Scholar]
  • 42.Papayannopoulou T, Priestley GV, Nakamoto B, et al. Molecular pathways in bone marrow homing: dominant role of alpha(4)beta(1) over beta(2)-integrins and selectins. Blood. 2001;98:2403–2411. doi: 10.1182/blood.v98.8.2403. [DOI] [PubMed] [Google Scholar]
  • 43.Kikuta T, Shimazaki C, Ashihara E, et al. Mobilization of hematopoietic primitive and committed progenitor cells into blood in mice by anti-vascular adhesion molecule-1 antibody alone or in combination with granulocyte colony-stimulating factor. Exp Hematol. 2000;28:311–317. doi: 10.1016/s0301-472x(99)00151-4. [DOI] [PubMed] [Google Scholar]
  • 44.Papayannopoulou T, Priestley GV, Nakamoto B. Anti-VLA4/VCAM-1-induced mobilization requires cooperative signaling through the kit/mkit ligand pathway. Blood. 1998;91:2231–2239. [PubMed] [Google Scholar]
  • *45.Ramirez P, Rettig MP, Uy GL, et al. BIO5192, a small molecule inhibitor of VLA-4, mobilizes hematopoietic stem and progenitor cells. Blood. 2009;114:1340–1343. doi: 10.1182/blood-2008-10-184721. This report describes the use of a small molecule inhibitor of VLA-4, BIO5192, for hematopoietic mobilization. The authors also show that BIO5192 enhances mobilization of both G-CSF and AMD3100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Nilsson SK, Johnston HM, Whitty GA, et al. Osteopontin, a key component of the hematopoietic stem cell niche and regulator of primitive hematopoietic progenitor cells. Blood. 2005;106:1232–1239. doi: 10.1182/blood-2004-11-4422. [DOI] [PubMed] [Google Scholar]
  • 47.Stier S, Ko Y, Forkert R, et al. Osteopontin is a hematopoietic stem cell niche component that negatively regulates stem cell pool size. J Exp Med. 2005;201:1781–1791. doi: 10.1084/jem.20041992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **48.Grassinger J, Haylock DN, Storan MJ, et al. Thrombin-cleaved osteopontin regulates hemopoietic stem and progenitor cell functions through interactions with alpha9beta1 and alpha4beta1 integrins. Blood. 2009;114:49–59. doi: 10.1182/blood-2009-01-197988. This report demonstrates that osteopontin not only regulates proliferation of HSC within the endosteal niche, but also mediates retention through β1 integrins. Studies also describe both endogenous mobilization and enhanced G-CSF mobilization in osteopontin knockout mice, demonstrating the importance of osteopontin for HSC niche retention. [DOI] [PubMed] [Google Scholar]
  • **49.Winkler IG, Sims NA, Pettit AR, et al. Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and their depletion mobilizes HSCs. Blood. 2010;116:4815–4828. doi: 10.1182/blood-2009-11-253534. This seminal study describes a population of "osteomacs", which are macrophages that form a layer over endosteal osteoblasts. This report demonstrates that osteomacs provide support to osteoblasts, and when macrophages are ablated using Mafia mice or Clo-lip treatment, osteoblasts are suppressed and HSC mobilize. Importantly, treatment with G-CSF results in migration of the osteomacs away from the endosteal layer, causing osteoblast suppression and HSC mobilization. The authors also show that when osteoclasts are inhibited using zoldronate treatment, G-CSF mobilization is not attenuated, and in fact, increases. [DOI] [PubMed] [Google Scholar]
  • **50.Chow A, Lucas D, Hidalgo A, et al. Bone marrow CD169+ macrophages promote the retention of hematopoietic stem and progenitor cells in the mesenchymal stem cell niche. J Exp Med. 2011;208:261–271. doi: 10.1084/jem.20101688. This report, like the one above by Winkler et al., also demonstrates that a resident macrophage population regulates the HSC niche. The authors describe a slightly different phenotype than the previously described osteomacs, and use an elegant animal model to demonstrate that the CD169+ macrophages are specifically involved. The authors also suggest that a soluble factor produced by the macrophages supports niche cells, specifically nestin+ MSC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **51.Christopher MJ, Rao M, Liu F, et al. Expression of the G-CSF receptor in monocytic cells is sufficient to mediate hematopoietic progenitor mobilization by G-CSF in mice. J Exp Med. 2011;208:251–260. doi: 10.1084/jem.20101700. This group developed a sophisticated animal model in which the G-CSF receptor was only expressed on CD68 expressing monocytes. The authors demonstrate that G-CSF signaling only on monocytes is sufficient to mediate mobilization, and like similar reports, demonstrate that monocytes/macrophages produce a soluble factor which increases SDF-1α production by osteoblasts. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Pelus LM, Bian H, King AG, Fukuda S. Neutrophil-derived MMP-9 mediates synergistic mobilization of hematopoietic stem and progenitor cells by the combination of G-CSF and the chemokines GRObeta/CXCL2 and GRObetaT/CXCL2delta4. Blood. 2004;103:110–119. doi: 10.1182/blood-2003-04-1115. [DOI] [PubMed] [Google Scholar]
  • 53.Kollet O, Dar A, Shivtiel S, et al. Osteoclasts degrade endosteal components and promote mobilization of hematopoietic progenitor cells. Nat Med. 2006;12:657–664. doi: 10.1038/nm1417. [DOI] [PubMed] [Google Scholar]
  • *54.Cho KA, Joo SY, Han HS, et al. Osteoclast activation by receptor activator of NF-kappaB ligand enhances the mobilization of hematopoietic progenitor cells from the bone marrow in acute injury. Int J Mol Med. 2010;26:557–563. doi: 10.3892/ijmm_00000499. This study demonstrates in in vitro assays that osteoclasts attenuate SDF-1α production by osteoblasts, and that RANK ligand treatment in an injury model in mice increases hematopoietic mobilization. [DOI] [PubMed] [Google Scholar]
  • 55.Shivtiel S, Kollet O, Lapid K, et al. CD45 regulates retention, motility, and numbers of hematopoietic progenitors, and affects osteoclast remodeling of metaphyseal trabecules. J Exp Med. 2008;205:2381–2395. doi: 10.1084/jem.20080072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Takamatsu Y, Simmons PJ, Moore RJ, et al. Osteoclast-mediated bone resorption is stimulated during short-term administration of granulocyte colony-stimulating factor but is not responsible for hematopoietic progenitor cell mobilization. Blood. 1998;92:3465–3473. [PubMed] [Google Scholar]
  • 57.Hirbe AC, Uluckan O, Morgan EA, et al. Granulocyte colony-stimulating factor enhances bone tumor growth in mice in an osteoclast-dependent manner. Blood. 2007;109:3424–3431. doi: 10.1182/blood-2006-09-048686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Adams GB, Chabner KT, Alley IR, et al. Stem cell engraftment at the endosteal niche is specified by the calcium-sensing receptor. Nature. 2006;439:599–603. doi: 10.1038/nature04247. [DOI] [PubMed] [Google Scholar]
  • 59.Chow DC, Wenning LA, Miller WM, Papoutsakis ET. Modeling pO(2) distributions in the bone marrow hematopoietic compartment. II. Modified Kroghian models. Biophys J. 2001;81:685–696. doi: 10.1016/S0006-3495(01)75733-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kubota Y, Takubo K, Suda T. Bone marrow long label-retaining cells reside in the sinusoidal hypoxic niche. Biochem Biophys Res Commun. 2008;366:335–339. doi: 10.1016/j.bbrc.2007.11.086. [DOI] [PubMed] [Google Scholar]
  • 61.Parmar K, Mauch P, Vergilio JA, et al. Distribution of hematopoietic stem cells in the bone marrow according to regional hypoxia. Proc Natl Acad Sci U S A. 2007;104:5431–5436. doi: 10.1073/pnas.0701152104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *62.Winkler IG, Barbier V, Wadley R, et al. Positioning of bone marrow hematopoietic and stromal cells relative to blood flow in vivo: serially reconstituting hematopoietic stem cells reside in distinct nonperfused niches. Blood. 2010;116:375–385. doi: 10.1182/blood-2009-07-233437. The authors utilize in vivo Hoescht dye infusions, coupled with antibody staining for HSC markers and transplantation, to demonstrate that HSC reside in nonperfused niches. This study also demonstrates that G-CSF treatment causes HSC to migrate to more perfused locations within the bone marrow, but does not alter vascular permeability. [DOI] [PubMed] [Google Scholar]
  • 63.Semenza GL, Nejfelt MK, Chi SM, Antonarakis SE. Hypoxia-inducible nuclear factors bind to an enhancer element located 3' to the human erythropoietin gene. Proc Natl Acad Sci U S A. 1991;88:5680–5684. doi: 10.1073/pnas.88.13.5680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Feldser D, Agani F, Iyer NV, et al. Reciprocal positive regulation of hypoxia-inducible factor 1alpha and insulin-like growth factor 2. Cancer Res. 1999;59:3915–3918. [PubMed] [Google Scholar]
  • 65.Cormier-Regard S, Nguyen SV, Claycomb WC. Adrenomedullin gene expression is developmentally regulated and induced by hypoxia in rat ventricular cardiac myocytes. J Biol Chem. 1998;273:17787–17792. doi: 10.1074/jbc.273.28.17787. [DOI] [PubMed] [Google Scholar]
  • 66.Krishnamachary B, Berg-Dixon S, Kelly B, et al. Regulation of colon carcinoma cell invasion by hypoxia-inducible factor 1. Cancer Res. 2003;63:1138–1143. [PubMed] [Google Scholar]
  • 67.Levy AP, Levy NS, Wegner S, Goldberg MA. Transcriptional regulation of the rat vascular endothelial growth factor gene by hypoxia. J Biol Chem. 1995;270:13333–13340. doi: 10.1074/jbc.270.22.13333. [DOI] [PubMed] [Google Scholar]
  • 68.Lin Q, Lee YJ, Yun Z. Differentiation arrest by hypoxia. J Biol Chem. 2006;281:30678–30683. doi: 10.1074/jbc.C600120200. [DOI] [PubMed] [Google Scholar]
  • 69.Danet GH, Pan Y, Luongo JL, et al. Expansion of human SCID-repopulating cells under hypoxic conditions. J Clin Invest. 2003;112:126–135. doi: 10.1172/JCI17669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Smith S, Broxmeyer HE. The influence of oxygen tension on the long-term growth in vitro of haematopoietic progenitor cells from human cord blood. Br J Haematol. 1986;63:29–34. doi: 10.1111/j.1365-2141.1986.tb07491.x. [DOI] [PubMed] [Google Scholar]
  • 71.Broxmeyer HE, Cooper S, Gabig T. The effects of oxidizing species derived from molecular oxygen on the proliferation in vitro of human granulocyte-macrophage progenitor cells. Ann N Y Acad Sci. 1989;554:177–184. doi: 10.1111/j.1749-6632.1989.tb22419.x. [DOI] [PubMed] [Google Scholar]
  • 72.Broxmeyer HE, Cooper S, Lu L, et al. Enhanced stimulation of human bone marrow macrophage colony formation in vitro by recombinant human macrophage colony-stimulating factor in agarose medium and at low oxygen tension. Blood. 1990;76:323–329. [PubMed] [Google Scholar]
  • *73.Liu SP, Lee SD, Lee HT, et al. Granulocyte colony-stimulating factor activating HIF-1alpha acts synergistically with erythropoietin to promote tissue plasticity. PLoS One. 2010;5:e10093. doi: 10.1371/journal.pone.0010093. This article reports that G-CSF increases HIF-1α expression, and that G-CSF and EPO can act synergistically during ischemia repair. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Levesque JP, Winkler IG, Hendy J, et al. Hematopoietic progenitor cell mobilization results in hypoxia with increased hypoxia-inducible transcription factor-1 alpha and vascular endothelial growth factor A in bone marrow. Stem Cells. 2007;25:1954–1965. doi: 10.1634/stemcells.2006-0688. [DOI] [PubMed] [Google Scholar]
  • 75.Piccoli C, D'Aprile A, Ripoli M, et al. The hypoxia-inducible factor is stabilized in circulating hematopoietic stem cells under normoxic conditions. FEBS Lett. 2007;581:3111–3119. doi: 10.1016/j.febslet.2007.05.077. [DOI] [PubMed] [Google Scholar]
  • 76.Ceradini DJ, Kulkarni AR, Callaghan MJ, et al. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med. 2004;10:858–864. doi: 10.1038/nm1075. [DOI] [PubMed] [Google Scholar]
  • 77.Staller P, Sulitkova J, Lisztwan J, et al. Chemokine receptor CXCR4 downregulated by von Hippel-Lindau tumour suppressor pVHL. Nature. 2003;425:307–311. doi: 10.1038/nature01874. [DOI] [PubMed] [Google Scholar]
  • *78.Gharib SA, Dayyat EA, Khalyfa A, et al. Intermittent hypoxia mobilizes bone marrow-derived very small embryonic-like stem cells and activates developmental transcriptional programs in mice. Sleep. 2010;33:1439–1446. doi: 10.1093/sleep/33.11.1439. While studying sleep apnea, the authors used a model of exposure to intermittent hypoxia and demonstrate that hypoxia increases SDF-1α in plasma and mobilizes very small embryonic like stem cells from the bone marrow to peripheral blood. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *79.Kirito K, Hu Y, Komatsu N. HIF-1 prevents the overproduction of mitochondrial ROS after cytokine stimulation through induction of PDK-1. Cell Cycle. 2009;8:2844–2849. doi: 10.4161/cc.8.17.9544. The authors of this study have previously shown that TPO results in an increase in mitochondrial production of ROS, which could be harmful to HSC if left unchecked. This report demonstrates that TPO increases HIF-1α expression that reduces ROS through a PDK-1 dependent mechanism. [DOI] [PubMed] [Google Scholar]
  • **80.Tesio M, Golan K, Corso S, et al. Enhanced c-Met activity promotes G-CSF-induced mobilization of hematopoietic progenitor cells via ROS signaling. Blood. 2011;117:419–428. doi: 10.1182/blood-2009-06-230359. The authors of this report evaluated the mechanisms governing hematopoietic mobilization, with an interest on stress induced mobilization as an endogenous response for host defense and repair. This report demonstrates that stress induced mobilization, or G-CSF treatment, result in c-Met activitation, causing quiescent, non-motile HPC to migrate. This effect requires ROS activity, demonstrating that at least a small level of ROS is needed for mobilization. [DOI] [PubMed] [Google Scholar]
  • 81.Vagima Y, Avigdor A, Goichberg P, et al. MT1-MMP and RECK are involved in human CD34+ progenitor cell retention, egress, and mobilization. J Clin Invest. 2009;119:492–503. doi: 10.1172/JCI36541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *82.Jeong M, Piao ZH, Kim MS, et al. Thioredoxin-interacting protein regulates hematopoietic stem cell quiescence and mobilization under stress conditions. J Immunol. 2009;183:2495–2505. doi: 10.4049/jimmunol.0804221. Using a knockout mouse for thioredoxin-interacting protein (TXNIP), the authors show that TXNIP deficiency reduces SDF-1α and OPN mediated HSC retention in the osteoblastic niche, and causes HSC exhaustion. [DOI] [PubMed] [Google Scholar]
  • 83.Artico M, Bosco S, Cavallotti C, et al. Noradrenergic and cholinergic innervation of the bone marrow. Int J Mol Med. 2002;10:77–80. [PubMed] [Google Scholar]
  • 84.Madden KS, Sanders VM, Felten DL. Catecholamine influences and sympathetic neural modulation of immune responsiveness. Annu Rev Pharmacol Toxicol. 1995;35:417–448. doi: 10.1146/annurev.pa.35.040195.002221. [DOI] [PubMed] [Google Scholar]
  • 85.Aitken SJ, Landao-Bassonga E, Ralston SH, Idris AI. Beta2-adrenoreceptor ligands regulate osteoclast differentiation in vitro by direct and indirect mechanisms. Arch Biochem Biophys. 2009;482:96–103. doi: 10.1016/j.abb.2008.11.012. [DOI] [PubMed] [Google Scholar]
  • 86.Spiegel A, Shivtiel S, Kalinkovich A, et al. Catecholaminergic neurotransmitters regulate migration and repopulation of immature human CD34+ cells through Wnt signaling. Nat Immunol. 2007;8:1123–1131. doi: 10.1038/ni1509. [DOI] [PubMed] [Google Scholar]
  • 87.Gruber-Olipitz M, Stevenson R, Olipitz W, et al. Transcriptional pattern analysis of adrenergic immunoregulation in mice. Twelve hours norepinephrine treatment alters the expression of a set of genes involved in monocyte activation and leukocyte trafficking. J Neuroimmunol. 2004;155:136–142. doi: 10.1016/j.jneuroim.2004.07.003. [DOI] [PubMed] [Google Scholar]
  • 88.Lucas D, Battista M, Shi PA, et al. Mobilized hematopoietic stem cell yield depends on species-specific circadian timing. Cell Stem Cell. 2008;3:364–366. doi: 10.1016/j.stem.2008.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Mendez-Ferrer S, Lucas D, Battista M, Frenette PS. Haematopoietic stem cell release is regulated by circadian oscillations. Nature. 2008;452:442–447. doi: 10.1038/nature06685. [DOI] [PubMed] [Google Scholar]
  • *90.Mendez-Ferrer S, Battista M, Frenette PS. Cooperation of beta(2)- and beta(3)-adrenergic receptors in hematopoietic progenitor cell mobilization. Ann N Y Acad Sci. 2010;1192:139–144. doi: 10.1111/j.1749-6632.2010.05390.x. This report follows on from previous studies by the authors, and demonstrates that β2 and β3 adrenergic receptors cooperate together during G-CSF mobilization. This study also shows that β3 signaling specifically reduces SDF-1α expression while β2 signaling induces clock gene expression. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *91.Kawamori Y, Katayama Y, Asada N, et al. Role for vitamin D receptor in the neuronal control of the hematopoietic stem cell niche. Blood. 2010;116:5528–5535. doi: 10.1182/blood-2010-04-279216. This report demonstrates that VDR knockout mice have impaired mobilization to G-CSF coincident with a lack of osteoblast suppression. The authors demonstrate that β2 adrenergic signaling increases expression of VDR and propose a model in which VDR is a mediator of SNS signaling to the niche. [DOI] [PubMed] [Google Scholar]
  • 92.Cho Y, Noshiro M, Choi M, et al. The basic helix-loop-helix proteins differentiated embryo chondrocyte (DEC) 1 and DEC2 function as corepressors of retinoid X receptors. Mol Pharmacol. 2009;76:1360–1369. doi: 10.1124/mol.109.057000. [DOI] [PubMed] [Google Scholar]
  • *93.Visigalli I, Ungari S, Martino S, et al. The galactocerebrosidase enzyme contributes to the maintenance of a functional hematopoietic stem cell niche. Blood. 2010;116:1857–1866. doi: 10.1182/blood-2009-12-256461. Using a genetic knockout of the GALC gene, the authors report that HSC fail to engraft in a GALC deficient niche. This study demonstrates that only physiological levels of GALC expression could restore the HSC deficit, as supraphysiological levels further worsened the impairment. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Hannun YA, Obeid LM. Principles of bioactive lipid signalling: lessons from sphingolipids. Nat Rev Mol Cell Biol. 2008;9:139–150. doi: 10.1038/nrm2329. [DOI] [PubMed] [Google Scholar]
  • 95.Kimura T, Boehmler AM, Seitz G, et al. The sphingosine 1-phosphate receptor agonist FTY720 supports CXCR4-dependent migration and bone marrow homing of human CD34+ progenitor cells. Blood. 2004;103:4478–4486. doi: 10.1182/blood-2003-03-0875. [DOI] [PubMed] [Google Scholar]
  • 96.Ryser MF, Ugarte F, Lehmann R, et al. S1P(1) overexpression stimulates S1P-dependent chemotaxis of human CD34+ hematopoietic progenitor cells but strongly inhibits SDF-1/CXCR4-dependent migration and in vivo homing. Mol Immunol. 2008;46:166–171. doi: 10.1016/j.molimm.2008.07.016. [DOI] [PubMed] [Google Scholar]
  • 97.Seitz G, Boehmler AM, Kanz L, Mohle R. The role of sphingosine 1-phosphate receptors in the trafficking of hematopoietic progenitor cells. Ann N Y Acad Sci. 2005;1044:84–89. doi: 10.1196/annals.1349.011. [DOI] [PubMed] [Google Scholar]
  • *98.Allende ML, Tuymetova G, Lee BG, et al. S1P1 receptor directs the release of immature B cells from bone marrow into blood. J Exp Med. 2010;207:1113–1124. doi: 10.1084/jem.20092210. The authors utilized a B-cell specific (CD19 Cre) knockout of the S1P1 receptor and demonstrate that knockout of S1P1 impairs the ability of immature B cells to exit the bone marrow into the periphery. CXCR4 antagonism also did not release immature B cells in knockout mice. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **99.Ratajczak MZ, Lee H, Wysoczynski M, et al. Novel insight into stem cell mobilization-plasma sphingosine-1-phosphate is a major chemoattractant that directs the egress of hematopoietic stem progenitor cells from the bone marrow and its level in peripheral blood increases during mobilization due to activation of complement cascade/membrane attack complex. Leukemia. 2010;24:976–985. doi: 10.1038/leu.2010.53. This report shows that S1P is released from erythrocytes and S1P concentration increases in plasma during G-CSF mobilization. The authors also report that inhibition of S1P lyase causes an accumulation of S1P in the bone marrow and prevents a bone/blood gradient, reducing hematopoietic mobilization. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Golan K, Vagima Y, Ludin A, et al. The Chemotactic Lipid S1P Regulates Hematopoietic Progenitor Cell Egress and Mobilization Via Its Major Receptor S1P1 and by SDF-1 Inhibition In a p38/Akt/mTOR Dependent Manner. Blood. 2010;116:553. [Google Scholar]
  • 101.Kim CH, Wan W, Liu R, et al. A Novel Paradigm In Stem Cell Trafficking: The Ratio of Peripheral Blood Sphingosine-1 Phosphate (S1P) to Bone Marrow Ceramide-1 Phosphate (C1P) Regulates Mobilization and Homing of Hematopoietic Stem Cells. Blood. 2010;116:554. [Google Scholar]
  • 102.Harun N, Thien M, Juarez J, et al. S1P1 Agonists for Use as Adjunct Mobilizing Agents. Blood. 2010;116:826. [Google Scholar]
  • 103.Hoggatt J, Singh P, Sampath J, Pelus LM. Prostaglandin E2 enhances hematopoietic stem cell homing, survival, and proliferation. Blood. 2009;113:5444–5455. doi: 10.1182/blood-2009-01-201335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **104.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. This article is a comprehensive review of eicosanoid signaling and regulation of hematopoiesis. A yin and yang model of eicosanoid signaling is presented, and a proof of principle is shown demonstrating opposing effects of PGE2 and cannabinoid signaling on HPC CXCR4 and VLA-4 expression. This report also demonstrates that cannabinoid receptor agonism facilitates mobilization both alone and in synergy with G-CSF. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • **105.Jiang S, Alberich-Jorda M, Zagozdzon R, et al. Cannabinoid receptor 2 and its agonists mediate hematopoiesis and hematopoietic stem and progenitor cell mobilization. Blood. 2011;117:827–838. doi: 10.1182/blood-2010-01-265082. This report shows that cannabinoid agonism mediates migration and expansion of HPC. Using transplantation assays, the authors demonstrate that cannabinoid agonism increases mobilization of HSC. When G-CSF mobilization was performed in knockout mice hematopoietic mobilization was decreased. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • *106.Jiang S, Zagozdzon R, Jorda MA, et al. 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;285:35471–35478. doi: 10.1074/jbc.M110.144758. Similar to the above report by the same authors, this study demonstrates HSC and HPC express cannabinoid receptors and that the bone marrow niche produces endocannabinoids. Cannabinoids were shown to mediate migration of HPC, and knockout of fatty acid amide hydrolase, an enzyme that degrades endocannabinoids, results in HPC expansion. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • *107.Hegde VL, Nagarkatti M, Nagarkatti PS. Cannabinoid receptor activation leads to massive mobilization of myeloid-derived suppressor cells with potent immunosuppressive properties. Eur J Immunol. 2010;40:3358–3371. doi: 10.1002/eji.201040667. This reports shows that cannabinoids expand and mobilize myeloid-derived suppressor cells; an effect that may be mediated by an increase in endogenous production of G-CSF. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *108.Hudson BD, Hebert TE, Kelly ME. Physical and functional interaction between CB1 cannabinoid receptors and beta2-adrenoceptors. Br J Pharmacol. 2010;160:627–642. doi: 10.1111/j.1476-5381.2010.00681.x. Using bioluminescence resonance energy transfer (BERT) this study demonstrates that the cannabinoid CB1 receptor is co-localized with β2 adrenergic receptors. The authors demonstrate that the signaling downstream of each receptor is altered by the co-localization to the other. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • *109.Carvalho AF, Mackie K, Van Bockstaele EJ. Cannabinoid modulation of limbic forebrain noradrenergic circuitry. Eur J Neurosci. 2010;31:286–301. doi: 10.1111/j.1460-9568.2009.07054.x. This report, similar to the one above, demonstrates that the cannabinoid CB1 receptor is associated with adrenergic receptors, and the authors suggest that cannabinoid signaling may alter adrenergic signaling using both direct and indirect mechanisms. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES