Stem Cell Migration - Ingenta Connect

7 downloads 0 Views 210KB Size Report
1Department of Medicine, Hematology and Oncology, University Hospital Muenster, Albert-Schweitzer-Strasse 33,. D-48129 Muenster, Germany. 2Institute of ...
Current Stem Cell Research & Therapy, 2007, 2, 89-103

89

Stem Cell Migration: A Quintessential Stepping Stone to Successful Therapy Corinna Weidt*,1 , Bernd Niggemann2 , Benjamin Kasenda2 , Theodore L. Drell3 , Kurt S. Zänker2 and Thomas Dittmar *,2 1Department

of Medicine, Hematology and Oncology, University Hospital Muenster, Albert-Schweitzer-Strasse 33, D-48129 Muenster, Germany 2Institute 3i3

of Immunology, Witten/ Herdecke University, Stockumer Str. 10, D-58448 Witten, Germany

Research SKM Oncology, Taunusstr. 9, D-65183 Wiesbaden, Germany Abstract: Migration is an innate and fundamental cellular function that enables hematopoietic stem cells (HSCs) and endothelial progenitors (EPCs) to leave the bone marrow, relocate to distant tissue, and to return to the bone marrow. An increasing number of studies demonstrate the widening scope of the therapeutic potential of both HSCs and endothelial cells. Therapeutic success however not only relies upon their ability to repair damaged tissue, but is also fundamentally dependent on the migration to these areas. Extensive in vivo and in vitro research efforts have shown that the most significant effects seen on HSC migration are initiated by the chemokine SDF-1α. In this review we will elucidate the many cellular and systemic factors of HSC and EPC cell migration and their modi operandi.

Keywords: Hematopoietic stem cell migration, homing, mobilization. INTRODUCTION Hematopoietic stem cells are of increasing interest due to an accumulating evidence of their therapeutic potential. This self-renewal and trans-differentiation capacity is however worthless, if these cells loose their capacity to migrate to regions of interest. Because transplantation protocols utilize intravenous injections, diseases that require hematopoietic stem cell transplantation would fail to rescue lethally irradiated recipients if their homing potential was impaired. Moreover, current research is investigating whether cells of both hematopoietic and angiogenic potential, such as hematopoietic stem cells (HSCs) and endothelial progenitor cells (EPCs), may be recruited in both ischemic and malignant tissues. This, on the one hand, will provide important information to aid the therapeutic reconstitution of damaged tissue, and on the other, prevent angiogenesis in malignant disease. One of the most prominent chemokines that initiate stem cell migration is the stromal cell-derived factor-1α (SDF1α). It provides a gradient toward which stem cells migrate, and in which they are retained in the bone marrow microenvironment. In this review we will report about the cellular and molecular pathways that regulate in the migration of HSCs and EPCs to and from the bone marrow, ischemic and malignant tissue. STROMAL CELL-DERIVED FACTOR-1 Chemokines are a group of small peptides that by definition initiate the migration of effector cells. To date *Address correspondence to these authors at the 1 Department of Medicine, Hematology and Oncology, University Hospital Muenster, Albert-Schweitzer-Strasse 33, D-48129 Muenster, Germany; Tel: +49 251 8356225; Fax: +49 251 8356709; E-mail: [email protected] 2 Institute of Immunology, Witten/ Herdecke University, Stockumer Str. 10, D-58448 Witten, Germany; Tel: +49 2302 926165; Fax: +49 2302 926158; E-mail: [email protected] 1574-888X/07 $50.00+.00

more than 50 chemokines and 18 chemokine receptors have been identified. Chemokines bind to several receptors as well as several receptors binding to several ligands [1, 2]. SDF-1α is a member of the CXC chemokine family, and has been shown to be essential for chemotaxis of both progenitor and mature blood cells, and is involved in Blymphopoiesis and myelopoiesis. It binds to the CXCR4 receptor, also known as CD184, which belongs to the class of G-protein coupled receptors, whereby chemokine binding will initiate the migration of lymphocytes [3], hematopoietic stem cells [4, 5], and tumor cells [6-8]. A recent study of Balabanian and colleagues indicated that SDF-1α also binds to and signals through the orphan receptor RDC1 in Tlymphocytes [9]. SDF-1α was isolated originally from a murine bonemarrow stromal cell line, and was reported to function as a pre-B-cell growth factor [10]. At present three SDF-1α isoforms, SDF-1α, SDF-1β, and SDF-1γ , are known. SDF1α and SDF-1β are encoded by a single gene, and are generated by alternative splicing [11]. SDF-1β (93 amino acids) has an additional 4 amino acids at the C-terminal end, whereby SDF-1α is made up of 89 amino acids. Sequence alignment analyses have revealed that SDF-1α is highly conserved between species. For instance, murine SDF-1α and SDF-1β sequences are more than 92% identical to those of human origin [11]. The SDF-1 gene was mapped to chromosome 10q, in contrast to other members of the intercrine family, which are localized on chromosome 4q and 17q. It has thus been hypothesized that SDF-1α may have important functions distinct from those of the other members of the intercrine family [11]. The third known isoform, SDF-1γ , was originally isolated from the rat [12]. Compared to mice and humans in which SDF-1α/β is expressed ubiquitously [11], Gleichmann and colleagues found that neurons and Schwann cells are the main regions of both SDF-1β and SDF-1γ mRNA expression in the rat [12]. SDF-1β is the © 2007 Bentham Science Publishers Ltd.

90

Current Stem Cell Research & Therapy, 2007, Vol. 2, No. 1

predominant isoform in embryonic and early postnatal rat nerve tissue, whereas SDF-1γ is expressed at higher levels in adulthood. Regulation of SDF-1 gene expression may be induced by the transcription factor hypoxia-inducible factor-1 (HIF-1) in endothelial cells. This results in selective in vivo expression of SDF in direct proportion to reduced oxygen tension [13]. Further characterization of the SDF promoter region has shown that the proximal promoter has 6 putative Sp1binding motifs [14]. Promotor regulation was demonstrated in three tested cell lines by using phorbol myristate acetate, ionomycin, gamma irradiation, and interferon gamma. In bone marrow, SDF-1α availability has been shown to be regulated by the uptake of SDF-1α via CXCR4 expressing cells, which then translocate the chemokine into the bone marrow. This transporter function is characteristic of both endothelial and stromal cells [15]. CXCR4 The SDF-1 receptor CXCR4 was independently described by several authors, thus explaining the various alternative names, such as neuropeptide Y (NPY) Y3 receptor [16], D2S201E [17], leukocyte-derived seventransmembrane domain receptor (LESTR) [18], or fusin [19]. All studies revealed a single open reading frame (ORF) of 352 amino acids that shared features common to many other 7-transmembrane G protein-coupled receptors, such as the human interleukin-8 (IL-8) receptor (36 – 37% homology), and bovine NPY receptor (92 to 93% homology). Bleul et al. and Oberlin et al. reported that SDF-1α is a ligand for this receptor, which they referred to as CXCR4 [20, 21]. Both groups showed that binding of SDF-1α to its’ receptor CXCR4 blocked the infection by lymphocytetropic HIV-1 strains in vitro. Similar results were obtained by Feng et al., showing that blocking of the CXCR4 receptor using anti-CXCR4 antibodies strongly inhibited HIV-1 infection of normal human CD4+ target cells [19]. Moreover, transient expression of the CXCR4 gene allowed non-human cells co-expressing recombinant CD4 to undergo Env-CD4-mediated cell fusion and productive HIV-1 infection, revealing that CXCR4 acts as a cofactor for HIV-1 infection in CD4 + T cells. CXCR4 is expressed in a variety of tissues, including brain, endothelial and epithelial cells, and tissues of hematopoietic origin [17]. Studies performed with CXCR4 knock-out mice revealed that CXCR4 is required for several physiological processes during late embryonic and early postnatal stages, as well as in the adult. CXCR4-deficient mice exhibited hematopoietic and cardiac defects [22], or died perinatally with defects in both the hematopoietic and nervous system [23]. Tachibana et al. showed that CXCR4 is essential for vascularization of the gastrointestinal tract [24]. Similar observations were made with SDF-1α deficient mice, highlighting the importance of the SDF-1α/CXCR4 signaling. Additionally, a reduced B-lymphopoiesis and myelopoiesis were observed in the fetal liver, whereby bone marrow myelopoiesis was completely ablated in these mice. Both groups showed that fetal cerebellar development was markedly different compared to wild type animals. For

Weidt et al.

instance, numerous proliferating granule cells abnormally invaded within the cerebellar anlage. The hypothesis that the SDF-1α/CXCR4 axis is involved in neuronal cell migration and patterning in the central nervous system was recently substantiated by the finding that CXCR4 mRNA is expressed at sites of neuronal and progenitor cell migration in the hippocampus at late embryonic and early postnatal stages [25]. The morphology of the hippocampal dentate gyrus is dramatically altered in CXCR4 knock-out mice due to an underlying defect in the stream of post mitotic cells and secondary dentate progenitor cells that migrate toward and from the dentate gyrus. The lack of CXCR4 signaling led to reduced migratory activity of neuronal progenitor cells, whereby neurons appeared to differentiate prematurely before reaching their target [25]. Mice deficient in CXCR4 showed a massive loss of spinal cord motoneurons and dorsal root ganglion neurons. This led to a reduced innervation of the developing mouse fore and hind limbs. Increased death of motoneurons in CXCR4deficient animals seems to result from impaired limb myogenesis and a subsequent loss of muscle-derived neurotrophic support [26]. CXCR4 AND SDF-1 The interaction between CXCR4 and its’ ligand SDF-1α plays a pivotal role in regulating the retention, migration, and mobilization of HSCs during steady state homeostasis and injury [27]. Administration of anti-CXCR4 antibodies prevented the engraftment of murine bone marrow by human SCID repopulating stem cells [28], whereas cytokine mediated CXCR4 up-regulation led to increased SDF-1α mediated migration, in vivo homing, and repopulation of HSCs [28-30]. Similarly, CXCR4 over-expression by human cord blood and mobilized peripheral blood CD34+ cells by lentiviral gene transfer resulted in an increased proliferation, SDF-1α mediated migration, and bone marrow engraftment of these cells [31, 32]. The expression of CXCR4 on HSCs is very dynamic and known to be regulated by both cytokines and chemokines. Exposure of cord blood derived and mobilized peripheral blood derived CD34+ HSCs to stem cell factor (SCF; also named c-kit-ligand) or a combination of SCF and IL6 led to a CXCR4 up-regulation, concomitant with increased motility of SCID repopulating cells (SRC) [28]. Similar results were achieved with hepatocyte growth factor (HGF), alone or in combination with SCF [29]. Sorted CD34+ CXCR4 - HSCs express intracellular CXCR4, which can be up-regulated and expressed on the cell surface in response to stimulation with 5 cytokines (SCF, Flt3-ligand, IL6, IL-3, and G-CSF), thereby converting these cells into definitive SRC with high CXCR4 expression levels (both intracellular and cell surface) and SDF-1α induced migration [33]. In contrast, our findings showed that incubation of CD133 HSCs from cord blood with Flt3-ligand alone or in combination with IL6 for 5 days resulted in a CXCR4 down-regulation in both populations (unpublished observations). However, despite a reduced CXCR4 expression, CD133 cord blood HSCs incubated with Flt3ligand and IL6 responded to SDF-1α with an increased locomotory activity and an altered migration pattern. This is

Stem Cell Migration

Current Stem Cell Research & Therapy, 2007, Vol. 2, No. 1 91

in contrast to cells solely incubated with Flt3-ligand, where only a slight increase in the migratory activity was detected [34]. Thus, cell-surface level expression of the SDF-1α receptor alone does not necessarily correlate with elevated migration or successful engraftment. On this note, a recent publication by Bertheboud and colleagues demonstrated that the regulators of G protein signaling (RGS) are responsible for negative regulation of G protein coupled receptors and regulate the activation of SDF1α signaling. The authors were able to show that an RGS16 over-expression in megakaryocytes inhibited SDF-1αinduced migration, mitogen activated protein kinase and protein kinase B activation [35]. MOBILIZATION CELLS

OF

HEMATOPOIETIC

STEM

Mobilization/recruitment and the homing of HSCs are mirror processes that are regulated by the interplay of cytokines, chemokines and proteases [36]. In brief, the process of HSC mobilization is characterized by both a loss of cell to cell contacts due to down-regulation and degradation of cell adhesion molecules, and a desensitization of the SDF-1α/CXCR4 axis. In contrast, up-regulation of cell adhesion molecules and activation of the SDF1α/CXCR4 axis is essential for stem cell homing, as we shall shortly see. Mobilization of HSCs from bone marrow is achieved by the application of cytokines, such as granulocyte-colony stimulating factor (G-CSF) and granulocyte-macrophagecolony-stimulating factor (GM-CSF) [37, 38], Flt-3 ligand [39-41], SCF [42, 43], and vascular endothelial growth factor (VEGF) [44], as well as several chemokines such as IL-8 [45-48], growth regulated oncogene-β (GROβ) [49], and macrophage inflammatory protein-2 (MIP-2) [50]. The aforementioned cytokines require 5 to 6 days for a peak level response to be reached; chemokines however induce mobilization within the time span of 30 minutes to a few hours. G-CSF G-CSF induced mobilization is a multi-step process that depends on both the disruption of cell-cell contacts by cleavage of cell adhesion molecules, and the CXCR4/SDF1α axis by inactivation of the SDF-1α receptor. Levesque and colleagues demonstrated that following GCSF application, active neutrophil granulocyte proteases, e.g. such as elastase and cathepsin G, accumulate within the bone marrow [51, 52]. An increase in the concentration of these proteases coincided with a down-regulation of the SCF receptor (CD117)[51], as well as a sharp reduction of the VCAM-1/CD106 expression in the bone marrow [52]. Conjointly, accumulation of neutrophil granulocyte secreted proteases within the bone marrow coincides with the cleavage of the N-terminus of CXCR4 on HSCs resident in the bone marrow and mobilized into the peripheral blood [53]. Recent findings indicate that CD26/Dipetidylpeptidase IV (DPPIV) plays an important role in G-CSF mediated HSC mobilization [54-57]. CD26/DPPIV is a membrane-

bound extracellular peptidase expressed on a subset of CD34 positive HSCs. It inactivates SDF-1α by cleaving it at position-2 proline [56]. Inhibition of CD26/DPPIV activity enhanced the migratory response of HSC to SDF-1α [54], thus pointing toward a regulatory role of CD26/DPPIV in SDF-1α mediated chemotaxis. Treatment of mice with CD26 inhibitors during G-CSF induced mobilization resulted in a reduced number of progenitor cells in the periphery, compared to the G-CSF regimen alone [54]. Similar results were obtained in CD26 (-/-) mice treated with G-CSF. Due to the absence of CD26/DPPIV activity, the number of HSCs in the peripheral blood was significantly lessened, compared to the corresponding number of HSCs detected in G-CSF treated wild-type mice [55]. It nevertheless remains to be determined whether CD26/DPPIV activity is directly triggered by G-CSF. Furthermore, the same group was able to show that endogenous CD26 expression on donor cells negatively regulates homing and engraftment [57]. FLT3-LIGAND Flt3 is a type III tyrosine kinase receptor expressed on primitive HSCs. Its ligand, aptly called Flt3-ligand, induces HSC mobilization in mice and acts synergistically with GCSF or GM-CSF. In a recent study by Fukuda and colleagues, Flt3-ligand was shown to enhance SDF-1α induced migration in the short term. Long term exposure to Flt3-ligand however caused a down-regulation of CXCR4 [39]. These findings suggest that Flt3-ligand plays an extrinsic regulatory role in the migration of HSCs that may be relevant in both homing to the bone marrow as well as mobilization. IL-8 In contrast to G-CSF-induced accumulation of elastase and cathepsin G in the bone marrow, IL-8 induced mobilization has been proposed to be dependent upon matrix metalloprotease-9 (MMP-9) activity. MMPs belong to the group of gelatinases/type IV collagenases and have been found to be of relevance to HSC migration and mobilization. The MMP family comprises 16 members [58] and are produced as latent pro-enzymes which are proteolytically processed to attain their activated status. The gelatinases predominantly degrade denatured collagens (gelatin) and also degrade collagen types IV, V, VII and X, as well as fibronectin and laminin (for a review see [59]). MMP-9 has been implicated in HSC mobilization in both mice and monkeys [45, 60-62]. Studies on rhesus monkeys showed that a single intravenous injection of IL-8 resulted in a dramatic increase in the plasma levels of MMP9, followed by a 10- to 100-fold increase of HSCs in the peripheral blood within 30 minutes after injection [60]. Pretreatment of the animals with a specific MMP-9 antibody blocked the IL-8 induced HSC mobilization. The mobilization of cells via the IL-8 and MMP pathway is not well understood. In a study by Pruijt and colleagues [63], the authors showed that the IL-8 induced mobilization was restored in neutropenic mice by injection with polymorphonuclear cells and non-purified mononuclear cells.

92

Current Stem Cell Research & Therapy, 2007, Vol. 2, No. 1

Weidt et al.

Furthermore, IL-8 induced mobilization was not impaired in MMP-9 deficient mice, demonstrating that MMP-9 is dispensable for mobilization.

caused the release of soluble SCF and permitted the transfer of HSCs from a quiescent to a proliferative niche [66], thus implying an active role of SDF-1α in HSC mobilization (Fig. ( 1)).

GRO

Mobilization via an increase in plasma SDF-1α levels has furthermore been demonstrated with both fucan sulfate and dextransulfate [68]. The authors were able to show that a single injection of these sulfated glycans bind to SDF-1α, and thereby inhibit SDF-1/heparin binding, and likewise cause an increase in plasma concentrations. Homologously MMP-9 deficient mice did not prevent mobilization, indicating that MMP-9 it is not essential for this mechanism.

CXCR2 mediated mobilization is initiated via the chemokine CXCL2, also known as GROβ, and has been shown to be mediated via neutrophil derived proteases [49]. Mobilization induced by GROβ/CXCL2 was associated with elevated plasma and bone marrow levels of MMP-9, whereas mobilization and the associated increase in MMP-9 levels did not take place in neutrophil granulocyte-depleted mice [49]. Interestingly, the authors reported that GROβ/ CXCL2 acted synergistically with G-CSF. HSC mobilization by both G-CSF and GROβ/CXCL2 correlated with a synergistic rise in the plasma levels of MMP-9 and a simultaneous increase in peripheral blood neutrophil granulocytes. In contrast, marrow levels of neutrophil granulocytes and neutrophil granulocyte-derived proteases were decreased. Synergistic mobilization was absent in MMP-9-deficient or neutrophil granulocyte-depleted mice and completely blocked in animals pretreated with antiMMP-9 [49]. SDF-1 The chemokine SDF-1α is to date one of the best described and widely confirmed chemoattractants for HSCs [4, 5]; several studies indicated that this chemokine is pivotal in HSC mobilization. Aiuti and colleagues reported that the migration capacity of G-CSF mobilized HSCs from peripheral blood in response to SDF-1α was lower than of HSCs from bone marrow, and suggested that an altered response to SDF-1α may be associated with HSC mobilization [5]. Furthermore, exposure of adult mice to the SDF-1β analog methionin-SDF-1β (Met-SDF-1β) resulted in HSC mobilization with a more than 30-fold increase compared to the PBS control [64]. Over-expression of SDF1α in the peripheral circulation of SCID mice via intravenous injection of an adenoviral vector expressing SDF-1α (AsSDF1) has also been shown to result in HSC mobilization [65, 66]. Although these studies implicate a direct role of SDF-1α in HSC mobilization, the reported effects are predominantly attributed to an impairment of the CXCR4/SDF-1α axis or a change in the SDF-1α concentration in peripheral blood and bone marrow. For example, the decreased migration rate of mobilized peripheral blood HSCs to SDF-1α [5] is attributed to a desensitization of the CXCR4/SDF-1α axis due to SDF-1α cleavage, mediated by G-CSF induced activation of neutrophil granulocyte-derived elastase [67]. Similarly, administration of Met-SDF, which is resistant to cleavage causes a prolonged CXCR4 desensitization, which again impairs CXCR4/SDF-1α signaling [64]. In contrast to the desensitization of the CXCR4/SDF-1α axis, mobilization of HSCs by adenovirus-driven SDF-1α was attributed to a change in the SDF-1α gradient between bone marrow and peripheral blood [66]. However, the same group reported that SDF-1α over-expression resulted in an activation of MMP-9 induced in bone marrow cells, which

OTHER MOBILIZING FACTORS More recent data has shown that more factors other than the above mentioned proteases and cytokines are involved in the mobilization process. For example, the calcium sensing receptor is expressed on HSCs, a method by which cells are able to respond to extracellular calcium concentrations. It serves to retain HSCs close to the endosteal surface [69]. The activation of noradrenergic neurons has also been shown to decrease the production of SDF-1α. Ablation of adrenergic neurotransmission indicated that norepinephrine signaling controls G-CSF induced osteoblast suppression, SDF down-regulation and HSC mobilization [70]. The regulation of stem cell release and expansion has not only been shown to be regulated by osteoblasts, but by osteoclasts as well. Zhang and colleagues found that the number of osteoblastic cells correlates with the number of HSCs, whereby long term HSCs were found to be attached to these cells, and bone morphogenic protein indirectly controlled the number of stem cells [71]. Recently the role of osteoclasts in stem cell homeostasis has been investigated by Kollet and colleagues. The mobilization of HSCs associated increase in osteoclast number led the authors to investigate a possible relationship between stem cell release from the endosteal niche [72]. Stimulation of the osteoclasts was then shown to cause a release of stem cells into the circulation in a CXCR4 and MMP-9 dependent manner. The cytokine RANKL caused an altered expression of the proteases MMP-9 and cathepsin K, which degrades membrane bound SCF, and also caused a decrease in osteopontin. In a clinical setting it is of continual concern to increase the mobilization efficacy of HSCs for therapeutic purposes. Concurrently several strategies for the mobilization of stem cells are being tested in the clinical setting. King and colleagues have shown that a truncated form of the CXC chemokine GROβ (SB-251353) alone or in combination with G-CSF mobilize hematopoietic stem cells with long term repopulating ability [73]. Likewise a combination of the CXCR4 antagonist AMD3100 and G-CSF to improve the efficacy of stem cell mobilization is being tested [74, 75]. AMD3100 is a bicyclam antagonist of the SDF-1α receptor CXCR4 and has been shown to induce rapid mobilization of CD34+ hematopoietic cells in mice, dogs and humans [74]. Recent findings by Broxmeyer and colleagues show that AMD3100 synergizes with G-CSF and leads to the mobilization of long-term repopulating (LTR)

Stem Cell Migration

Current Stem Cell Research & Therapy, 2007, Vol. 2, No. 1 93

Fig. (1). Stem cell homing and mobilization to and from the bone marrow endosteum. HSC mobilization and homing are mirror processes that strongly depend on the activation state of cell-cell contacts and the SDF-1α/ CXCR4 axis. HSC mobilization from bone marrow mediated by cytokines, e.g. G-CSF or chemokines such as IL-8 is caused by proteases (MMPs, elastase, and cathepsin G) that degrades adhesion molecules, membrane bound c-kit, and SDF-1α and its receptor CXCR4. In contrast, up-regulation of cell adhesion molecules and activation of the SDF-1α/ CXCR4 axis is essential for stem cell homing. The blue coloring represents the SDF-1α gradient in the bone marrow.

cells [76]. Moreover lymphoma and myeloma cells do not appear to be mobilized by this drug [75]. AMD3100 is currently being tested in phase I-III clinical trials. HOMING OF HEMATOPOIETIC STEM CELLS Homing, as defined by Lapidot et al., is a descriptive term used for the crossing of circulating HSCs across the blood/bone endothelial barrier into the bone marrow compartment within a fairly short time span of hours to days. Successful homing is measured by the successful reconstitution of hematopoiesis [77]. HSC homing is a multi-step process requiring an interplay of adhesion molecules, cytokines and chemokines, and extracellular matrix degrading proteases. This resembles the process of leukocyte/ lymphocyte extravasation [78], as

well as trans-endothelial migration of tumor cells during the hematogenous metastasis of cancer [79]. Extravasating cells (1) adhere to the vascular endothelium, (2) transmigrate across the endothelial lining and the underlying basement membrane, and (3) migrate into the surrounding tissue. Although prefatory work on molecular pathways guiding extravasation of HSCs has provided some insight, the mechanisms explaining this complex process remain nebulous [80]. Studies investigating the mechanisms of homing were first published in the early 90s. These provided a first indication that galactose and mannose specific lectins and their receptors were important for successful bone marrow engraftment [81]. As in the case of lymphocytes/ leucocytes, the rolling phase of extravasation is mediated by E- and Pselectins [82, 83]. Subsequent firm adhesion is mediated through intercellular adhesion molecule-1 (ICAM-

94

Current Stem Cell Research & Therapy, 2007, Vol. 2, No. 1

1)/leukocyte function-associated antigen-1 (LFA-1), vascular adhesion molecule-1 (VCAM-1)/very late antigen-4 (VLA-4) ligand pairs, and junctional adhesion molecules, such as platelet endothelial cell adhesion molecule (PECAM) [83]. The importance of VLA-4 (α 4β1 integrin) and VCAM interactions have been demonstrated by the use of a blocking anti-α 4-antibody; results showed that HSC homing was inhibited by more than 90% [80, 84]. Similar results were achieved with hypomorphic VCAM-1 mice with domain 4 deletion (D4D) and low expression of VCAM-1. Experiments with the latter showed that homing was virtually abrogated when the animals were treated with antiVCAM-1 antibody and were given anti-CD11a-treated cells [80]. However, β2-integrins and selectins may be used as a fall-back if the dominant VLA-4/ VCAM-1 (α 4β1/ VCAM1) interaction is compromised [80]. Furthermore α4 integrins have been shown to selectively mediate the homing of cells to the bone marrow, but not to the spleen (as was demonstrated in transplantation studies using α4 (-/-) cells) [85].

Weidt et al.

Central to the regulation of this process is SDF-1α, which has been shown to trigger the firm adhesion to LFA-1 to ICAM, as well as the VLA-4 to VCAM-1 under shear flow conditions [86]. Studies by Peled and colleagues indicated that the SDF-1α induced polarization and extravasation of CD34+ /CXCR4 + HSCs through the extracellular matrix underlining the endothelium is dependent upon both VLA-4 and VLA-5 [87]. A myriad of publications have shown that polarization per se is a prerequisite for migration [88], as has been shown in leucocytes as well as in tumor cells. SDF-1α causes a relocalization of CXCR4 to the leading edge of transfected KG1a cells upon contact with HUVECS. CXCR4 has furthermore been shown to be co-localized with lipid rafts, and is found at cell to cell interaction sites when in contact with the endothelial cell surface [89] (Fig. (2)). Polarization was also induced when cells stimulated with SDF-1α were brought together with immobilized hyaluronic acid (HA). CD44 was found at the leading edge of pseudopodia and is involved in adhesion and immobilization of cells [90]. Pretreatment of these cells with blocking anti-CD44 antibodies

Fig. (2). Migrating HSCs show a polarized phenotype. A confocal laser scanning microscope picture of a CD34 cord blood derived progenitor cell embedded in a 3D collagen lattice. Anti βtubulin (green) and phalloidin (red) staining reveals the polarization of the migrating cell. The fibers of the collagen lattice appear in blue.

Stem Cell Migration

prevented these morphologic alterations implying that CD44-HA interaction is a prerequisite for polarization. Pilarski and colleagues found that the HA receptor RHAMM on CD34 cells leads to an increase in motility, whereby CD44 leads to an increase in adhesion. CD44-HA has furthermore been shown to play a role in the lodging of HSCs in the spleen [90, 91] and bone marrow [90-92]. However, studies performed on CD44 deficient mice showed that these animals have no hematopoietic abnormalities and that CD44-/- HSCs are capable of homing to the bone marrow of normal or CD44-/- animals [93]. Inconsistencies in these results may be readily explained by the expression of various CD44 isoforms [94]. A recent study has shown that HSCs express a novel glycoform of CD44 named HCELL (hematopoietic cell E-/L-selectin ligand) and most likely plays a pivotal role in HSC trafficking to the bone marrow [95]. Cytokines trigger the adhesion of HSCs during homing. Several studies have shown that various cytokines such as GM-CSF, IL-3, and SCF temporarily increase the adhesiveness of HSCs by activating the β1-integrins VLA-4 and VLA-5. Cytokines (Flt3-ligand, SCF, IL-3, IL6, HGF) up-regulate CXCR4 expression on HSCs in vitro and in vivo [29, 86, 96], thereby enhancing the intracellular signals generated through the SDF-1α/ CXCR4 axis [97]. Our investigations and those published by Fukuda et al. have shown that prolonged exposure of CD133 + or CD34+ HSCs from cord blood to Flt3-ligand leads to a down-regulation of CXCR4 expression [34, 39]. Interestingly, short-term (2 day) exposure of CD34+ HSCs with Flt3-ligand inhibited the SDF-1α mediated phosphorylation of MAPKp42/p44, CREB, and Akt, and impaired migration toward CXCL12 [39], whereas long-term (5 day) Flt3-ligand incubation of CD133+ HSCs responded to SDF-1α with an increased migration rate, despite reduced CXCR4 expression [34]. Moreover, similar results were observed for cord-blood derived CD133+ HSCs cultured in Flt3-ligand and IL6. Prolonged incubation time led to a decrease in CXCR4 expression levels, whereas the migratory response to SDF1α was enhanced. These cells displayed an altered migration pattern, as well as a recruitment of non-moving cells. The mechanisms responsible for the altered migration pattern of cells cultured in IL6 remain to be elucidated. Some research has however shown that the differentiation of HSCs is regulated by IL6 [98-101]. It may thus be speculated that these phenotypical changes could be attributed to an altered gene expression profile. Transendothelial migration of cells presupposes the degradation of the basement membrane, which requires the production of matrix-degrading enzymes, especially those capable of degrading type IV collagen such as MMPs [58]. Both MMP-2 and MMP-9 are known to be expressed by all cell types that migrate through the endothelial cell layer including leukocytes [78, 102-105], tumor cells [58, 106114], and HSCs [27, 29, 115, 116]. Their expression and secretion are stimulated by various chemokines and cytokines, as discussed in the previous section. Two recent studies by Zheng and colleagues showed that CD34+ HSC from cord blood exhibited significantly lower expression levels of CD49e, CD49f, CXCR4 as well as MMP-2 and MMP-9, compared to their counterparts from peripheral blood and bone marrow, which may be a reason for delayed

Current Stem Cell Research & Therapy, 2007, Vol. 2, No. 1 95

hematopoietic reconstitution after umbilical cord blood transplantation [117, 118]. However, upon incubation with recombinant human SCF, these cells gained increased expression of the homing related molecules, including CXCR4 and MMP-2/-9, and showed an increased ex vivo transmigratory and in vivo homing potential [117, 118]. SDF-1 /CXCR4 MIGRATION

SIGNAL

TRANSDUCTION

AND

Cell migration is an essential component of both successful mobilization and homing. Its’ initiation, as discussed in the previous sections, is controlled by a multitude of extracellular parameters. These include extrinsic initiation, such as the extracellular matrix, signaling via chemokine receptors and receptor tyrosine kinases [119]. These pathways ultimately lead to an activation of actin associated proteins, reorganization of the actin cytoskeleton, as well as the activation of focal adhesion kinases. The SDF-1 receptor CXCR4 belongs to the group of 7transmembrane receptors, also known as serpentine receptors, and are linked to G proteins [19, 20], as described in previous sections. SDF-1α induced chemotaxis in HSCs may be inhibited by the addition of pertussis toxin, indicating that it is associated with a Gα i-protein subtype [5, 120, 121]. In the case of both T lymphocytes and CD34 positive cells, the PI3-kinase, phospholipase C-γ (PLC-γ )/ protein kinase C (PKC) cascade and MAPKp42/44 (ERK-1/2) are activated upon SDF-1α binding [122, 123]. Studies on human T cell lines indicated that SDF-1α triggers CXCR4 dimerization and activates the JAK/STAT pathway, which suggests gene regulation [124]. Similarly, Ganju and colleagues have reported that SDF-1α treatment led to increased NF-κB activity in nuclear extracts of CXCR4 transfectants, indicating that changes of the gene expression level can be initiated via two independent signal transduction pathways down-stream of the SDF-1α/ CXCR4 axis [122]. In contrast, Lee and colleagues have demonstrated that NF-κB is activated after addition of SDF1α [97]. The same group has however demonstrated that MAPK p42/44 (ERK-1/2), ribosomal S6 kinase (p90RSK) and Akt are synergistically activated by SDF-1α in combination with GM-CSF, SCF, TPO, or Flt3-ligand, and that this correlated with an enhanced survival of cord blood derived CD34+ HSCs [97]. The actin cytoskeleton is one of the central mechanical components responsible for the motility of cells, and its’ analysis is an effective method of determining a migratory phenotype. Actin polymerization in migration is induced by the PI3-kinase and the PLC-γ / PKC cascade in a variety of cell types [119, 125-127], including HSCs. So far several groups have convincingly demonstrated that SDF-1α induces actin polymerization [5, 39] as well as tyrosine phosphorylation of several components of focal adhesion complexes such as paxillin, the related adhesion focal tyrosine kinase (RAFTK/ pyk2), p130cas , Crk-II, and Crk-L [122, 123]. Inhibition of PI3-kinase signaling using wortmannin partially inhibited the SDF-1α induced migration and tyrosine phosphorylation of paxillin [122], further underpinning the role PI3-kinase in cell migration. In a recent study by Petit and co-workers, SDF-1α induced cell migration was shown to activate an atypical

96

Current Stem Cell Research & Therapy, 2007, Vol. 2, No. 1

PKC subtype known as PKC-ζ [128]. Activation of this subtype is not dependent upon calcium or diacylglycerole (DAG) [129, 130]. Furthermore cell polarization, adhesion to bone marrow stromal cells and chemotaxis were all shown to be PKC-ζ-dependent. PKC-ζ phosphorylation, translocation to the plasma membrane, and kinase activity, was furthermore shown to be dependent upon PI3-kinase activity, Pyk-2 and MAPKp42/44 (ERK-1/2). MMP-9 was also found to be activated after PKC-ζ phosphorylation [128]. Moreover, in vivo studies showed that engraftment, but not homing, of human CD34+ HSCs was also dependent upon PKC-ζ activity [128]. Cancelas and colleagues were recently able to demonstrate that a post engraftment deletion of the Rho GTPases Rac1 and Rac2 led to a massive mobilization of HSCs from the bone marrow. Rac1(-/-) mutants were not able to rescue hematopoiesis after transplantation, but a deletion of Rac1 did not prevent steady state hematopoiesis. The authors were thus able to demonstrate that Rac proteins regulate HSC motility in both homing and mobilization [131]. The examination of HSC migration however should not only be limited to an analysis of signal transduction pathways, but should also include a study of the migratory phenotype. For the general study of migration Maheshweri and Lauffenburger proposed that cell population migration should be deconstructed in terms of a mathematical model, comprising of cell population parameters such as random motility, chemotaxis/haptotaxis, and chemotaxis/haptotaxis coefficients [132]. They recognized that individual cell paths can be analyzed in a model of individual cell parameters which include translocation, speed, and directional persistence in time, which in turn depend upon membrane extension and retraction, cell to substratum adhesion, cell contractile force and front vs. rear symmetry. Commonplace migration analyses are carried out in a 2D environment, which regrettably offer but an indication of the situation in vivo, and are thus insufficient for the study of the aforementioned parameters. In the benchmark work by Friedl and colleagues, leukocytes were introduced into a 3D environment, in which the migratory parameters such as speed, directionality, persistence, trajectory, and percentage locomoting cells were analyzed and determined for CD4 and CD8 lymphocytes [133]. Comparative experiments using lymphocytes and tumor cells demonstrated that these parameters were determined, and seen to be characteristic for different cell types [134]. Video microscopic observations concomitantly present the opportunity to further scrutinize the relationship between morphology and migration, and have successfully been implemented in the case of tumor cells and cord blood derived CD133 + cells [34, 134]. In a recent comparative study of both 2D and 3D migration [135], the authors were able to determine that the 2D model predictions were qualitatively similar to the 3D situation, but that new effects such as matrix sterics and mechanics arose in the 3D situation. In our work we analyzed various migratory parameters in a 3D collagen matrix. These parameters included the time active within the period of the assay, the velocity of the cells (speed without pauses), the overall speed (with pauses), the pause length, pause frequency, and the overall distance migrated. Upon stimulation with SDF-1α and following extensive single

Weidt et al.

cell analysis, the migratory phenotype showed a recruitment of non-moving cells, an increase in speed and frequency of pauses, but a decrease in pause duration. This resulted in an increase in the distance migrated. The study of cell migration should be considered on both a molecular and cell population level, for both are integral components of homing and mobilization. The understanding of these parameters becomes particularly relevant in a clinical setting where engraftment and mobilization rates vary greatly from patient to patient. Both isolated HSCs as well as whole bone marrow are commonly used to restore hematopoiesis in hematopoietic maladies, whereby bone marrow transplantation is preferred in clinical transplantation. Bone marrow transplantation is currently the only therapeutic approach for several lethal malignant and non-malignant disorders. The use of cord blood as a source of progenitor cells was quickly established as an alternative. To date over 50 banks worldwide serve as a source for cord blood, and over 2000 patients have received transplantation [136]. Progenitors are also being targeted as potential vectors for gene therapy to invoke immune tolerance with the aim of facilitating allogeneic or xenogeneic organ transplants. Despite the fact that cord blood transplantation strategies have meanwhile become routine in clinical practice, engraftment is still very variable [137]. Intensive studies in the recent years indicate that the ability of hematopoietic stem cells to engraft is dependent on cell their cycle status. Studies on murine HSCs that have been exposed to a cytokine cocktail revealed that the engraftment phenotype showed marked fluctuations over 24h intervals, whereby least effective responses were recorded with cells in late S and early G2 phase [138]. Enhanced long-term donor-derived multilineage reconstitution of the peripheral blood was observed in recipients of G0/G1-phase HSCs from fetal liver compared to recipients of S/G 2/Mphase cells [139]. Similar results were observed for human cord blood derived CD34+ HSCs that were stimulated to proliferate [139]. HSCs that had entered the S/G2/M-phase lost engraftment potential and did not reenter the G0-phase. No differences in the expression level of VLA-4, VLA-5, and CXCR4 were detectable in cultured G0/G1-phase and S/G 2/M-phase cord blood CD34+ HSCs indicating that an unknown cell cycle-associated mechanism selectively prevents proliferating HSCs to successfully engraft [139]. Yamaguchi and colleagues have however demonstrated that CD34+ HSCs from various sources (bone marrow, peripheral blood, and apheresis peripheral blood samples), and in different stages of the cell cycle (G0/G1-phase versus S/G 2/M-phase), exhibited altered adhesive characteristics and VLA-4 expression [140]. In a comparative study by Ramirez and colleagues, CD34 cells were expanded for six days. The authors showed that the adhesion molecules VLA-4, VLA-5, MAC-1 (α Mβ2 integrin) were up-regulated, but that the adhesion to fibronectin was significantly decreased [141]. Different results are probably due to different culture conditions and times used: Ramirez cultured the cells for six days in IL-3, IL6, and SCF or alternatively in IL-11, SCF, and Flt3-ligand [141]. Glimm and colleagues used a five day culture with Flt3-ligand, SCF, IL-3, IL6, and G-CSF [139], and Yamaguchi on the other hand used a 4 day culture with IL-3 and SCF [140].

Stem Cell Migration

The cycling of long term culture initiating cells and primitive human colony forming cells is stopped upon addition of SDF-1α to the culture media of CD34 HSCs [142, 143]. The recovery rate of transplanted NOD/SCID mice increased two fold under this regimen [144]. Thus, blocking the cell cycle of HSCs suggests new strategies to improve the engraftment activity of ex vivo expanded HSCs. SDF-1 IN INFLAMMATORY CONDITIONS The role that SDF-1α plays in inflammatory processes has long remained unclear. SDF-1α is ubiquitously expressed in various tissues [11]; it was thus suggested to play a role in immune surveillance and in the basal extravasation of lymphocytes, rather than in inflammation [120]. A study by Gonzalo and colleagues delivered the first indication that SDF-1α is involved in inflammatory processes. They demonstrated that SDF-1α is a critical inflammatory component in allergic airway disease [145]. Administration of neutralizing antibodies to CXCR4 in a mouse model of allergic airway disease resulted in reduced eosinophilia (broncho-alveolar lavage fluid and interstitium) by half, which was accompanied by a significant decrease in airway hyper-responsiveness. A later study revealed that the influence of SDF-1α in the inflammatory component of allergic airway disease is more passive than active [146]. Exposure of human airway epithelial cells with proinflammatory cytokines resulted in CXCR4 up-regulation, thus enabling the cells to respond more efficiently to constitutively expressed SDF-1α. The treatment of an asthma mouse model with the CXCR4 antagonist AMD3100 furthermore significantly reduced airway hyper-reactivity, peribronchial eosinophilia and the overall inflammatory response [18]. The study of bacterial neurological infections have shown that SDF-1α participates in the recruitment on dendritic cells in the cerebrospinal fluid [147, 148]. Patients suffering from noninflammatory neurological diseases, as well as inflammatory neurological diseases, were seen to have elevated SDF-1α in the cerebrospinal fluid. The application of blocking antibodies led to a decrease in the recruitment of both mature and immature dendritic cells [148]. For rheumatoid arthritis it has been reported that SDF-1α plays an important role in CD4+ memory T cell accumulation in the rheumatoid arthritis synovium [149, 150]. CXCR4 expression on CD4+ T cells was enhanced by IL-15 [150] and/or TGF-β [149], whereas SDF-1α expression of the synovium and on synovial endothelial cells was increased by CD40 stimulation [150], and led to a strong integrin mediated adhesion of synovial fluid T cells to fibronectin and ICAM1 [149]. Recent findings indicate that SDF-1α is modulated by inflammatory cytokines and proteases. Gene profiling analysis of endothelial cells showed that the inflammatory cytokines TNF-α and IFN-γ inhibit the expression of proangiogenic molecules including SDF-1α [151]. Similar results were reported by Fedyk et al., showing that secretion of TNF-α and IL-1α from activated macrophages inhibited the expression of SDF-1α by human fibroblasts in vitro [152]. The biological activity of SDF-1α is not only impaired by inflammatory cytokine-mediated down-

Current Stem Cell Research & Therapy, 2007, Vol. 2, No. 1 97

regulation of the gene, but also by degradation of the chemokine by proteases, such as elastase [153], MMPs [154], and CD26/Dipeptidylpeptidase IV (DPPIV) [56]. Modulation of SDF-1α function by inflammatory cytokines and/or proteases plays a role in various biological processes. Salvucci et al. showed that down-regulation of endothelial cell-derived SDF-1α by TNF-α and IFN-γ correlated with a disruption of extracellular matrix-dependent endothelial cell tube formation, an in vitro morphogenic process that recapitulates critical steps in angiogenesis [151]. Degradation of endothelial-bound SDF-1α by elastase released by transmigrating neutrophils may prove to be involved in the regulation of leukocyte trafficking to sites of inflammation [153]. Pre-treatment of endothelial cells with purified neutrophil elastase resulted in an inhibition of T lymphocyte transmigration. Elastase selectively cleaves the aminoterminus of endothelial-bound SDF-1α, which is required for chemotactic activity. The emerging inactivated cytokine is immobilized on the endothelial cell, thereby preventing T lymphocyte transmigration. In contrast, within the bone marrow elastase plays a major role in dislodging HSCs from bone marrow stroma by degrading VLA-4, VCAM-1, CXCR4, and SDF-1α [51-53]. Due to desensitization of the CXCR4/SDF-1α axis concomitant with a loss of cell-cell contacts, CD34+ HSCs are successfully released from the bone marrow (see above). HOMING OF HSCs TO ORGANS In this section we would briefly like to discuss the role of SDF-1α and HSCs in tissue regeneration. The first work that suggested that HSCs were homed to damaged tissue by SDF-1α was published by Kollet and co-workers [29]. Previous findings that the level of HSC engraftment into the liver is amplified during liver injury or viral inflammation prompted the authors to investigate the role that SDF-1α plays in this process. Both irradiation and inflammation led to elevated SDF-1α expression in liver bile and duct epithelial cells. Conjointly, hepatic injury induced MMP-9 activity, leading to both increased CXCR4 expression and SDF-1α mediated recruitment of hematopoietic progenitors to the liver. Recruited HSCs were found localized in clusters surrounding the bile ducts, in close proximity to SDF-1α expressing epithelial cells, and had differentiated into albumin-producing cells [29]. In a recent publication by Jin and colleagues, the authors were able to show that nonendothelial CXCR4 + , VEGFR1+ progenitors constitute a major determinant of revascularization in ischemia [155]. A series of cytokines such as SCF, TPO, erythropoietin (EPO), and GM-CSF induced the release of SDF-1α from platelets. It has been shown that stress signals, such as tissue injury and/or inflammation, cause up-regulation of SDF-1α in endothelial cells, thereby recruiting HSCs. This has been demonstrated in tissues such as heart [156, 157], kidney [157], and brain [158]. Ceradini et al. found that the recruitment of CXCR4-positive progenitor cells to regenerating tissues is mediated by the hypoxic gradient, namely via HIF-1 induced expression of SDF-1α in endothelial cells [13]. The up-regulation of SDF-1α in ischemic tissue is directly proportional to reduced oxygen tension, and was correlated with increased adhesion,

98

Current Stem Cell Research & Therapy, 2007, Vol. 2, No. 1

migration, and homing of circulating HSCs to ischemic tissue. MALIGNANT DISEASE, SDF-1 AND HSCs Tumor tissue is very heterogeneous; it includes not only tumor cells but also fibroblasts, lymphocytes and macrophages. Tumor associated macrophages (TAMs) for example, have been suggested to sustain microenvironmental conditions in malignant tissue [159]. TAMs are attracted to tumor tissue via hypoxia and they themselves initiate angiogenesis [160]. They likewise secrete proinflammatory cytokines, such as tumor necrosis factor-α (TNF-α), IL-1, IL6 and also chemokines such as monocyte chemoattractant protein-1, -2, -3 (MCP-1, -2, -3) [161, 162]. Several publications have also shown that bone marrow derived cells contribute to malignant tissue. Using the angiogenic deficient Id-mutant mouse model, Lyden and colleagues were able to show that both wild type and VEGF mobilized bone marrow was able to restore tumor angiogenesis. By targeting both VEGFR1 and VEGFR2 complete ablation of tumor growth was achieved [163]. Other studies have shown that CD34-positive cells of hematopoietic nature with endothelial morphology were found in tumor tissue [164-166]. So called tie2, CD11b expressing monocytes were likewise found in tumor tissue [167]. In this study the authors were able to show that a knockout of tie2 expressing monocytes completely prevented human glioma neovascularization in the mouse brain and brought about substantial tumor regression. But the role that bone marrow derived cells may assume in a malignant environment is not yet fully explored. The aforementioned studies, as well as many others, suggest that the bone marrow derived cells may contribute to malignant tissue. If one however combines both the thought that stem cells recruit to malignant tissues with that of the tumor as an instructing milieu [168-171] not only in the sense that they contribute to neovascularization, but to the development of new malignant tissue, current paradigms of stem cell therapy should be (re)considered carefully. Stem cells recruited to tumor tissue may on the one hand remain dormant in this environment, but may also boost cancer progression. In a hypothetical process of transdifferentiation or fusion, signals from the malignant environment may cause these stem cells to both retain stem cell characteristics and attain a variety of common features of cancer cells. In an important work by Müller et al., the authors were able to demonstrate that breast cancer metastasis to the regional lymph nodes, bone marrow, liver and lung is directed by SDF-1α. The target organs were shown to predominantly express SDF-1α, and breast cancer cells themselves expressed CXCR4 [6]. The application of a neutralizing SDF-1α specific antibody effectively impaired metastasis formation in regional lymph nodes and lung. The pivotal role that SDF-1α plays has furthermore been shown for bone marrow metastasis formation in neuroblastoma [172], and in metastasis formation in prostate cancer [8, 173]. In melanoma, SDF-1α has been shown to promote tumor cell invasion across basement membranes via stimulation of MT1-MMP activity [174]. Moreover, SDF1α triggered the activation of the GTPases RhoA, Rac1, and

Weidt et al.

Cdc42 in the highly metastatic BLM melanoma cell line [174]. Furthermore, CXCR4 expression on melanoma cells was notably augmented by transforming growth factor-β1 (TGF-β1), a matrigel component, whereas anti-TGF-β1 antibodies inhibited increases in CXCR4 expression and melanoma cell invasion toward SDF-1α. It is well known that tumor tissue is hypoxic, and that under these conditions the transcription factor HIF regulates gene expression and allows tumor cells to adapt to hypoxic conditions [175, 176]. Recently the expression of SDF-1α and CXCR4 have been suggested to be regulated by HIF [13, 177]. Taken together these findings suggest that SDF-1α signaling is involved in metastasis formation. ENDOTHELIAL PROGENITOR CELL MOBILIZATION Bone marrow derived endothelial progenitors have become the focus of intense investigation in the past decade. First indications of the existence of a bone marrow derived endothelial precursor population were discovered by Asahara and colleagues [178]. They found that up to 20% of the CD34 positive population of peripheral blood was also VEGFR2 positive. They have been phenotypically characterized by antigens that to date have concomitantly been associated with hematopoietic stem and progenitor cells. These encompass CD133, CD34, c-kit, VEGRR2, CD144 (VE-Cadherin), and Sca-1 [179]. An ever increasing number of studies have provided multiple indications that endothelial precursors contribute to neo-angiogenesis in wound healing, post myocardial ischemia, cerebral ischemia, limb ischemia and tumor growth. The elucidation of mechanisms that enable the mobilization of endothelial progenitor cells (EPCs) will provide a new target for therapeutical strategies in the treatment of cardiovascular diseases and malignancies. The mobilization of these cells out of the bone marrow may be induced by G-CSF [180], GM-CSF [181], SDF-1α [182], VEGF [182, 183], angiopoietin-1 [182, 183], erythropoietin (EPO) [184, 185], estrogen [186], physical activity [187] and two members of the platelet-derived growth factor (PDGF) family, PDGF-AA and PDGF–CC [188]. In a study by Bergers and colleagues, MMP-9 was shown to render normal pancreatic islets angiogenic via VEGF. MMPs made membrane bound cytokines such as VEGF-A biologically available [189], and invoked an angiogenic switch in carcinogenesis. Furthermore, osteoclast migration may be initiated by VEGF [190], these are cells which now have been shown to regulate the release of HSCs via MMP-9 and CXCR4. Kopp and colleagues have furthermore been able to show that marrow suppression (or release of VEGF-A or placental growth factor) result in an increase in MMP-9, as well as soluble SCF within the bone marrow [179]. The increase of soluble kit ligand promotes stem cell cycling and motility. Another very interesting study published by Grunewald et al., this year [191] analyzed the role of VEGF in the recruitment of circulating mononuclear myeloid cells from the bone marrow. These cells were held in close proximity to angiogenic vessels by SDF-1α, the expression of which

Stem Cell Migration

was induced by VEGF in perivascular myofibroblasts. They enhanced the in situ proliferation of endothelial cells, and not the recruitment of cells from the bone marrow. In regard to metastasis the mobilization of VEGFR1 positive cells from the bone marrow cells have been suggested to provide the soil for metastases [192]. In light of this data, one can conclude that this population provides an ideal target for anti-angiogenic therapy, but must in light of the Kaplan study be considered carefully so as not to provide additional soil for metastases. Even though several factors that stimulate EPC mobilization are known, the cellular and molecular mechanisms remain to be elucidated. This also applies of EPC homing, which is still poorly understood. Recently, β2-integrins have been reported to be play a role in EPC homing by showing in a murine model of hind limb ischemia. Preactivation of β2-integrins expressed on EPCs by activating antibodies augments the EPC-induced neovascularization in vivo [193]. Elucidation of these mechanisms will provide a valuable means to regulate the processes involving EPCs such as ischemia and neoangiogenesis. This is still a challenge in light of technical difficulties with EPC numbers as well as the still open discussion of EPC antigen profile.

Current Stem Cell Research & Therapy, 2007, Vol. 2, No. 1 99 [2] [3] [4] [5]

[6] [7] [8] [9] [10] [11] [12]

CONCLUSION In this review, we have elucidated the current understanding of the role of migration in HSC and EPC function. We have furthermore discussed the myriad of factors, and their complex cellular and systemic regulation of HSC migration. The most prominent known regulators of migration include MMPs and elastases, c-kit, and SCF which govern the expression of SDF-1α and its receptor CXCR4. This however begs the question: How does this knowledge help nurture the current (often insufficient) therapies for various illnesses? Current therapies rely on the harvesting of HSCs from the bone marrow, isolating the clinically relevant sub-population and re-administering via intravenous injection. These therapies unfortunately do not always deliver the desired clinical benefit. In these processes, migration plays a pivotal role which is central in both the mobilization of cells from the bone marrow, and subsequent engraftment in the recipient. An understanding of migration and its initiation, regulation, and mechanisms may thus provide a promising approach to enhance and enrich current therapeutic strategies involving HSC and EPC engraftment for a range of critical illnesses.

[13]

ACKNOWLEDGEMENTS

[22]

We would like to apologize to all those authors whose invaluable work was not mentioned in the above. This work was supported by the Fritz-Bender-Foundation, Munich, Germany.

[14] [15]

[16] [17]

[18] [19] [20] [21]

[23]

[24]

REFERENCES [1]

Youn BS, Mantel C, Broxmeyer HE. Chemokines, chemokine receptors and hematopoiesis. Immunol Rev 2000; 177: 150-74.

[25]

Thelen M. Dancing to the tune of chemokines. Nat Immunol 2001; 2: 129-34. Kucia M, Jankowski K, Reca R, et al. CXCR4-SDF-1 signalling, locomotion, chemotaxis and adhesion. J Mol Histol 2004; 35: 23345. Wright DE, Bowman EP, Wagers AJ, Butcher EC, Weissman IL. Hematopoietic stem cells are uniquely selective in their migratory response to chemokines. J Exp Med 2002; 195: 1145-54. Aiuti A, Webb IJ, Bleul C, Springer T, Gutierrez-Ramos JC. 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: 111-20. Muller A, Homey B, Soto H, et al. Involvement of chemokine receptors in breast cancer metastasis. Nature 2001; 410: 50-6. Sun YX, Wang J, Shelburne CE, et al. Expression of CXCR4 and CXCL12 (SDF-1) in human prostate cancers (PCa) in vivo. J Cell Biochem 2003; 89: 462-73. Vaday GG, Hua SB, Peehl DM, et al. CXCR4 and CXCL12 (SDF1) in Prostate Cancer: Inhibitory Effects of Human Single Chain Fv Antibodies. Clin Cancer Res 2004; 10: 5630-9. Balabanian K, Lagane B, Infantino S, et al. The chemokine SDF1/CXCL12 binds to and signals through the orphan receptor RDC1 in T lymphocytes. J Biol Chem 2005; 280: 35760-6. Nagasawa T, Kikutani H, Kishimoto T. Molecular Cloning and structure of a pre-B-cell growth-stimulating factor. Proc Natl Acad Sci USA 1994; 91: 2305-9. Shirozu M, Nakano T, Inazawa J, et al. Structure and chromosomal localization of the human stromal cell-derived factor 1 (SDF1) gene. Genomics 1995; 28: 495-500. Gleichmann M, Gillen C, Czardybon M, et al. Cloning and characterization of SDF-1gamma, a novel SDF-1 chemokine transcript with developmentally regulated expression in the nervous system. Eur J Neurosci 2000; 12: 1857-66. 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: 858-64. Garcia-Moruja C, Alonso-Lobo JM, Rueda P, et al. Functional characterization of SDF-1 proximal promoter. J Mol Biol 2005; 348: 43-62. Dar A, Goichberg P, Shinder V, et al. Chemokine receptor CXCR4-dependent internalization and resecretion of functional chemokine SDF-1 by bone marrow endothelial and stromal cells. Nat Immunol 2005; 6: 1038-46. Rimland J, Xin W, Sweetnam P, et al. Sequence and expression of a neuropeptide Y receptor cDNA. Mol Pharmacol 1991; 40: 86975. Federsppiel B, Melhado IG, Duncan AM, et al. Molecular cloning of the cDNA and chromosomal localization of the gene for a putative seven-transmembrane segment (7-TMS) receptor isolated from human spleen. Genomics 1993; 16: 707-12. Loetscher M, Geiser T, O'Reilly T, et al. Cloning of a human seven-transmembrane domain receptor, LESTR, that is highly expressed in leukocytes. J Biol Chem 1994; 269: 232-7. Feng Y, Broder CC, Kennedy PE, Berger EA. HIV-1 entry cofactor: functional cDNA cloning of a seven-transmembrane, G protein-coupled receptor. Science 1996; 272: 872-7. Bleul CC, Farzan M, Choe H, et al. The lymphocyte chemoattractant SDF-1 is a ligand for LESTR/fusin and blocks HIV-1 entry. Nature 1996; 382: 829-33. Oberlin E, Amara A, Bachelerie F, et al. The CXC chemokine SDF-1 is the ligand for LESTR/fusin and prevents infection by Tcell-line-adapted HIV-1. Nature 1996; 382: 833-5. Zou YR, Kottmann AH, Kuroda M, Taniuchi I, Littman DR. Function of the chemokine receptor CXCR4 in haematopoiesis and in cerebellar development. Nature 1998; 393: 595-9. Ma Q, Jones D, Borghesani PR, et al. Impaired B-lymphopoiesis, myelopoiesis, and derailed cerebellar neuron migration in CXCR4- and SDF-1-deficient mice. Proc Natl Acad Sci USA 1998; 95: 9448-53. Tachibana K, Hirota S, Iizasa H, et al. The chemokine receptor CXCR4 is essential for vascularization of the gastrointestinal tract. Nature 1998; 393: 591-4. Lu M, Grove EA, Miller RJ. Abnormal development of the hippocampal dentate gyrus in mice lacking the CXCR4 chemokine receptor. Proc Natl Acad Sci USA 2002; 99: 7090-5.

100 [26] [27] [28] [29] [30]

[31]

[32]

[33]

[34]

[35] [36]

[37]

[38] [39] [40]

[41] [42]

[43]

[44] [45]

[46]

Current Stem Cell Research & Therapy, 2007, Vol. 2, No. 1 Odemis V, Lamp E, Pezeshki G, et al. Mice deficient in the chemokine receptor CXCR4 exhibit impaired limb innervation and myogenesis. Mol Cell Neurosci 2005; 30: 494-505. Lapidot T, Dar A, Kollet O. How do stem cells find their way home? Blood 2005; Peled A, Petit I, Kollet O, et al. Dependence of human stem cell engraftment and repopulation of NOD/SCID mice on CXCR4. Science 1999; 283: 845-8. Kollet O, Shivtiel S, Chen YQ, et al. HGF, SDF-1, and MMP-9 are involved in stress-induced human CD34+ stem cell recruitment to the liver. J Clin Invest 2003; 112: 160-9. Kollet O, Spiegel A, Peled A, et al. 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; 97: 3283-91. Brenner S, Whiting-Theobald N, Kawai T, et al. CXCR4transgene expression significantly improves marrow engraftment of cultured hematopoietic stem cells. Stem Cells 2004; 22: 112833. Kahn J, Byk T, Jansson-Sjostrand L, et al. Overexpression of CXCR4 on human CD34+ progenitors increases their proliferation, migration, and NOD/SCID repopulation. Blood 2004; 103: 2942-9. Kollet O, Petit I, Kahn J, et al. Human CD34(+)CXCR4(-) sorted cells harbor intracellular CXCR4, which can be functionally expressed and provide NOD/SCID repopulation. Blood 2002; 100: 2778-86. Weidt C, Niggemann B, Hatzmann W, Zanker KS, Dittmar T. Differential effects of culture conditions on the migration pattern of stromal cell-derived factor-stimulated hematopoietic stem cells. Stem Cells 2004; 22: 890-6. Berthebaud M, Riviere C, Jarrier P, et al. RGS16 is a negative regulator of SDF-1-CXCR4 signaling in megakaryocytes. Blood 2005; 106: 2962-8. Gazitt Y. Homing and mobilization of hematopoietic stem cells and hematopoietic cancer cells are mirror image processes, utilizing similar signaling pathways and occurring concurrently: circulating cancer cells constitute an ideal target for concurrent treatment with chemotherapy and antilineage-specific antibodies. Leukemia 2004; 18: 1-10. Winter JN, Lazarus HM, Rademaker A, et al. Phase I/II study of combined granulocyte colony-stimulating factor and granulocytemacrophage colony-stimulating factor administration for the mobilization of hematopoietic progenitor cells. J Clin Oncol 1996; 14: 277-86. Gianni AM, Siena S, Bregni M, et al. Granulocyte-macrophage colony-stimulating factor to harvest circulating haemopoietic stem cells for autotransplantation. Lancet 1989; 2: 580-5. Fukuda S, Broxmeyer HE, Pelus LM. Flt3 ligand and the Flt3 receptor regulate hematopoietic cell migration by modulating the SDF-1alpha(CXCL12)/CXCR4 axis. Blood 2005; 105: 3117-26. Molineux G, McCrea C, Yan XQ, Kerzic P, McNiece I. Flt-3 ligand synergizes with granulocyte colony-stimulating factor to increase neutrophil numbers and to mobilize peripheral blood stem cells with long-term repopulating potential. Blood 1997; 89: 3998-4004. Brasel K, McKenna HJ, Morrissey PJ, et al. Hematologic effects of flt3 ligand in vivo in mice. Blood 1996; 88: 2004-12. Bodine DM, Seidel NE, Zsebo KM, Orlic D. In vivo administration of stem cell factor to mice increases the absolute number of pluripotent hematopoietic stem cells. Blood 1993; 82: 445-55. Molineux G, Migdalska A, Szmitkowski M, Zsebo K, Dexter TM. The effects on hematopoiesis of recombinant stem cell factor (ligand for c-kit) administered in vivo to mice either alone or in combination with granulocyte colony-stimulating factor. Blood 1991; 78: 961-6. Gill M, Dias S, Hattori K, et al. Vascular trauma induces rapid but transient mobilization of VEGFR2(+)AC133(+) endothelial precursor cells. Circ Res 2001; 88: 167-74. Laterveer L, Lindley IJ, Hamilton MS, Willemze R, Fibbe WE. Interleukin-8 induces rapid mobilization of hematopoietic stem cells with radioprotective capacity and long-term myelolymphoid repopulating ability. Blood 1995; 85: 2269-75. Laterveer L, Lindley IJ, Heemskerk DP, et al. Rapid mobilization of hematopoietic progenitor cells in rhesus monkeys by a single intravenous injection of interleukin-8. Blood 1996; 87: 781-8.

Weidt et al. [47]

[48] [49]

[50] [51]

[52]

[53]

[54] [55] [56]

[57] [58] [59] [60]

[61] [62] [63] [64] [65] [66]

[67] [68]

Watanabe T, Kawano Y, Kanamaru S, et al. Endogenous interleukin-8 (IL-8) surge in granulocyte colony-stimulating factor-induced peripheral blood stem cell mobilization. Blood 1999; 93: 1157-63. van Pel M, van Os R, Velders GA, et al. Serpina1 is a potent inhibitor of IL-8-induced hematopoietic stem cell mobilization. Proc Natl Acad Sci USA 2006; 103: 1469-74. Pelus LM, Bian H, King AG, Fukuda S. Neutrophil-derived MMP9 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: 110-9. Wang J, Mukaida N, Zhang Y, et al. Enhanced mobilization of hematopoietic progenitor cells by mouse MIP-2 and granulocyte colony-stimulating factor in mice. J Leukoc Biol 1997; 62: 503-9. Levesque JP, Hendy J, Winkler IG, Takamatsu Y, Simmons PJ. Granulocyte colony-stimulating factor induces the release in the bone marrow of proteases that cleave c-KIT receptor (CD117) from the surface of hematopoietic progenitor cells. Exp Hematol 2003; 31: 109-17. Levesque JP, Hendy J, Takamatsu Y, et al. Mobilization by either cyclophosphamide or granulocyte colony-stimulating factor transforms the bone marrow into a highly proteolytic environment. Exp Hematol 2002; 30: 440-9. Levesque JP, Hendy J, Takamatsu Y, Simmons PJ, Bendall LJ. Disruption of the CXCR4/CXCL12 chemotactic interaction during hematopoietic stem cell mobilization induced by GCSF or cyclophosphamide. J Clin Invest 2003; 111: 187-96. Christopherson KW, 2nd, Cooper S, Broxmeyer HE. Cell surface peptidase CD26/DPPIV mediates G-CSF mobilization of mouse progenitor cells. Blood 2003; 101: 4680-6. Christopherson KW, Cooper S, Hangoc G, Broxmeyer HE. CD26 is essential for normal G-CSF-induced progenitor cell mobilization as determined by CD26-/- mice. Exp Hematol 2003; 31: 1126-34. Christopherson KW, Hangoc G, Broxmeyer HE. Cell Surface Peptidase CD26/Dipeptidylpeptidase IV Regulates CXCL12/Stromal Cell-Derived Factor-1alpha-Mediated Chemotaxis of Human Cord Blood CD34(+) Progenitor Cells. J Immunol 2002; 169: 7000-8. Christopherson KW, 2nd, Hangoc G, Mantel CR, Broxmeyer HE. Modulation of hematopoietic stem cell homing and engraftment by CD26. Science 2004; 305: 1000-3. Chang C, Werb Z. The many faces of metalloproteases: cell growth, invasion, angiogenesis and metastasis. Trends Cell Biol 2001; 11: S37-S43. Rundhaug JE. Matrix metalloproteinases and angiogenesis. J Cell Mol Med 2005; 9: 267-85. Pruijt JF, Fibbe WE, Laterveer L, et al. Prevention of interleukin8-induced mobilization of hematopoietic progenitor cells in rhesus monkeys by inhibitory antibodies against the metalloproteinase gelatinase B (MMP-9). Proc Natl Acad Sci USA 1999; 96: 108638. Fibbe WE, Pruijt JF, van Kooyk Y, et al. The role of metalloproteinases and adhesion molecules in interleukin-8induced stem-cell mobilization. Semin Hematol 2000; 37: 19-24. Fibbe WE, Pruijt JF, Velders GA, et al. Biology of IL-8-induced stem cell mobilization. Ann N Y Acad Sci 1999; 872: 71-82. Pruijt JF, Verzaal P, van Os R, et al. Neutrophils are indispensable for hematopoietic stem cell mobilization induced by interleukin-8 in mice. Proc Natl Acad Sci USA 2002; 99: 6228-33. Shen H, Cheng T, Olszak I, et al. CXCR-4 desensitization is associated with tissue localization of hemopoietic progenitor cells. J Immunol 2001; 166: 5027-33. Hattori K, Heissig B, Rafii S. The regulation of hematopoietic stem cell and progenitor mobilization by chemokine SDF-1. Leuk Lymphoma 2003; 44: 575-82. Hattori K, Heissig B, Tashiro K, et al. Plasma elevation of stromal cell-derived factor-1 induces mobilization of mature and immature hematopoietic progenitor and stem cells. Blood 2001; 97: 3354-60. Petit I, Szyper-Kravitz M, Nagler A, et al. G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and upregulating CXCR4. Nat Immunol 2002; 3: 687-94. Sweeney EA, Papayannopoulou T. Increase in circulating SDF-1 after treatment with sulfated glycans. The role of SDF-1 in mobilization. Ann N Y Acad Sci 2001; 938: 48-52; discussion -3.

Stem Cell Migration [69] [70] [71] [72] [73]

[74] [75]

[76]

[77]

[78] [79] [80]

[81] [82]

[83]

[84]

[85]

[86] [87]

[88] [89]

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. 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: 407-21. Zhang J, Niu C, Ye L, et al. Identification of the haematopoietic stem cell niche and control of the niche size. Nature 2003; 425: 836-41. Kollet O, Dar A, Shivtiel S, et al. Osteoclasts degrade endosteal components and promote mobilization of hematopoietic progenitor cells. Nat Med 2006; 12: 657-64. King AG, Horowitz D, Dillon SB, et al. Rapid mobilization of murine hematopoietic stem cells with enhanced engraftment properties and evaluation of hematopoietic progenitor cell mobilization in rhesus monkeys by a single injection of SB251353, a specific truncated form of the human CXC chemokine GRObeta. Blood 2001; 97: 1534-42. Larochelle A, Krouse A, Metzger M, et al. AMD3100 mobilizes hematopoietic stem cells with long-term repopulating capacity in nonhuman primates. Blood 2006; 107: 3772-8. Flomenberg N, DiPersio J, Calandra G. Role of CXCR4 chemokine receptor blockade using AMD3100 for mobilization of autologous hematopoietic progenitor cells. Acta Haematol 2005; 114: 198-205. Broxmeyer HE, Orschell CM, Clapp DW, et al. Rapid mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a CXCR4 antagonist. J Exp Med 2005; 201: 1307-18. Lapidot T. Mechanism of human stem cell migration and repopulation of NOD/SCID and B2mnull NOD/SCID mice. The role of SDF-1/CXCR4 interactions. Ann NY Acad Sci 2001; 938: 83-95. Madri JA, Graesser D. Cell migration in the immune system: the evolving inter-related roles of adhesion molecules and proteinases. Dev Immunol 2000; 7: 103-16. Heyder C, Gloria-Maercker E, Hatzmann W, Zaenker KS, Dittmar T. Visualization of tumor cell extravasation. Contrib Microbiol 2006; 13: 200-8. Papayannopoulou T, Priestley GV, Nakamoto B, Zafiropoulos V, Scott LM. Molecular pathways in bone marrow homing: dominant role of alpha(4)beta(1) over beta(2)-integrins and selectins. Blood 2001; 98: 2403-11. Tavassoli M, Hardy CL. Molecular basis of homing of intravenously transplanted stem cells to the marrow. Blood 1990; 76: 1059-70. Spertini O, Cordey AS, Monai N, Giuffre L, Schapira M. Pselectin glycoprotein ligand 1 is a ligand for L-selectin on neutrophils, monocytes, and CD34+ hematopoietic progenitor cells. J Cell Biol 1996; 135: 523-31. Naiyer AJ, Jo DY, Ahn J, et al. Stromal derived factor-1-induced chemokinesis of cord blood CD34(+) cells (long-term cultureinitiating cells) through endothelial cells is mediated by E-selectin. Blood 1999; 94: 4011-9. Papayannopoulou T, Craddock C, Nakamoto B, Priestley GV, Wolf NS. The VLA4/VCAM-1 adhesion pathway defines contrasting mechanisms of lodgement of transplanted murine hemopoietic progenitors between bone marrow and spleen. Proc Natl Acad Sci USA 1995; 92: 9647-51. Scott LM, Priestley GV, Papayannopoulou T. Deletion of alpha4 integrins from adult hematopoietic cells reveals roles in homeostasis, regeneration, and homing. Mol Cell Biol 2003; 23: 9349-60. Peled A, Grabovsky V, Habler L, et al. The chemokine SDF-1 stimulates integrin-mediated arrest of CD34(+) cells on vascular endothelium under shear flow. J Clin Invest 1999; 104: 1199-211. Peled A, Kollet O, Ponomaryov T, et al. The chemokine SDF-1 activates the integrins LFA-1, VLA-4, and VLA-5 on immature human CD34(+) cells: role in transendothelial/stromal migration and engraftment of NOD/SCID mice. Blood 2000; 95: 3289-96. Servant G, Weiner OD, Herzmark P, et al. Polarization of chemoattractant receptor signaling during neutrophil chemotaxis. Science 2000; 287: 1037-40. van Buul JD, Voermans C, van Gelderen J, et al. Leukocyteendothelium interaction promotes SDF-1-dependent polarization of CXCR4. J Biol Chem 2003; 278: 30302-10.

Current Stem Cell Research & Therapy, 2007, Vol. 2, No. 1 101 [90] [91] [92] [93]

[94]

[95] [96] [97]

[98]

[99]

[100]

[101]

[102]

[103]

[104] [105] [106] [107]

[108] [109] [110] [111] [112]

Avigdor A, Goichberg P, Shivtiel S, et al. CD44 and hyaluronic acid cooperate with SDF-1 in the trafficking of human CD34+ stem/progenitor cells to bone marrow. Blood 2004; 103: 2981-9. Vermeulen M, Le Pesteur F, Gagnerault MC, et al. Role of adhesion molecules in the homing and mobilization of murine hematopoietic stem and progenitor cells. Blood 1998; 92: 894-900. Khaldoyanidi S, Denzel A, Zoller M. Requirement for CD44 in proliferation and homing of hematopoietic precursor cells. J Leukoc Biol 1996; 60: 579-92. Oostendorp RA, Ghaffari S, Eaves CJ. Kinetics of in vivo homing and recruitment into cycle of hematopoietic cells are organspecific but CD44-independent. Bone Marrow Transplant 2000; 26: 559-66. Screaton GR, Bell MV, Jackson DG, et al. Genomic structure of DNA encoding the lymphocyte homing receptor CD44 reveals at least 12 alternatively spliced exons. Proc Natl Acad Sci USA 1992; 89: 12160-4. Sackstein R. The bone marrow is akin to skin: HCELL and the biology of hematopoietic stem cell homing. J Invest Dermatol 2004; 122: 1061-9. Rosu-Myles M, Gallacher L, Murdoch B, et al. The human hematopoietic stem cell compartment is heterogeneous for CXCR4 expression. Proc Natl Acad Sci USA 2000; 97: 14626-31. Lee Y, Gotoh A, Kwon HJ, et al. Enhancement of intracellular signaling associated with hematopoietic progenitor cell survival in response to SDF-1/CXCL12 in synergy with other cytokines. Blood 2002; 99: 4307-17. Audet J, Miller CL, Rose-John S, Piret JM, Eaves CJ. Distinct role of gp130 activation in promoting self-renewal divisions by mitogenically stimulated murine hematopoietic stem cells. Proc Natl Acad Sci USA 2001; 98: 1757-62. Hirano T, Ishihara K, Hibi M. Roles of STAT3 in mediating the cell growth, differentiation and survival signals relayed through the IL-6 family of cytokine receptors. Oncogene 2000; 19: 254856. Jacobsen SE, Ruscetti FW, Okkenhaug C, et al. Distinct and direct synergistic effects of IL-1 and IL-6 on proliferation and differentiation of primitive murine hematopoietic progenitor cells in vitro. Exp Hematol 1994; 22: 1064-9. Kopf M, Ramsay A, Brombacher F, et al. Pleiotropic defects of IL-6-deficient mice including early hematopoiesis, T and B cell function, and acute phase responses. Ann NY Acad Sci 1995; 762: 308-18. Bar-Or A, Nuttall RK, Duddy M, et al. Analyses of all matrix metalloproteinase members in leukocytes emphasize monocytes as major inflammatory mediators in multiple sclerosis. Brain 2003; 126: 2738-49. Vaday GG, Hershkoviz R, Rahat MA, et al. Fibronectin-bound TNF-alpha stimulates monocyte matrix metalloproteinase-9 expression and regulates chemotaxis. J Leukoc Biol 2000; 68: 737-47. Amorino GP, Hoover RL. Interactions of monocytic cells with human endothelial cells stimulate monocytic metalloproteinase production. Am J Pathol 1998; 152: 199-207. Leber TM, Balkwill FR. Regulation of monocyte MMP-9 production by TNF-alpha and a tumour-derived soluble factor (MMPSF). Br J Cancer 1998; 78: 724-32. Cockett MI, Murphy G, Birch ML, et al. Matrix metalloproteinases and metastatic cancer. Biochem soc Symp 1998; 63: 295-313. Kato Y, Yamashita T, Ishikawa M. Relationship between expression of matrix metalloproteinase-2 and matrix metalloproteinase-9 and invasion ability of cervical cancer cells. Oncol Rep 2002; 9: 565-9. Curran S, Murray GI. Matrix metalloproteinases in tumour invasion and metastasis. J Pathol 1999; 189: 300-8. Ellerbroek SM, Stack MS. Membrane associated matrix metalloproteinases in metastasis. Bioessays 1999; 21: 940-9. Kleiner DE, Stetler-Stevenson WG. Matrix metalloproteinases and metastasis. Cancer Chemother Pharmacol 1999; 43 Suppl: S42-S51. Polette M, Birembaut P. Membrane-type metalloproteinases in tumor invasion. Int J Biochem Cell Biol 1998; 30: 1195-202. Stamenkovic I. Matrix metalloproteinases in tumor invasion and metastasis. Semin Cancer Biol 2000; 10: 415-33.

102 [113] [114] [115]

[116]

[117]

[118]

[119] [120] [121]

[122]

[123]

[124]

[125] [126]

[127]

[128] [129] [130] [131] [132] [133]

Current Stem Cell Research & Therapy, 2007, Vol. 2, No. 1 Westermarck J, Kahari VM. Regulation of matrix metalloproteinase expression in tumor invasion. FASEB J 1999; 13: 781-92. Yoon SO, Park SJ, Yun CH, Chung AS. Roles of matrix metalloproteinases in tumor metastasis and angiogenesis. J Biochem Mol Biol 2003; 36: 128-37. Janowska-Wieczorek A, Marquez LA, Nabholtz JM, et al. Growth factors and cytokines upregulate gelatinase expression in bone marrow CD34(+) cells and their transmigration through reconstituted basement membrane. Blood 1999; 93: 3379-90. Lapidot T, Petit I. Current understanding of stem cell mobilization: the roles of chemokines, proteolytic enzymes, adhesion molecules, cytokines, and stromal cells. Exp Hematol 2002; 30: 973-81. Zheng Y, Sun A, Han ZC. Stem cell factor improves SCIDrepopulating activity of human umbilical cord blood-derived hematopoietic stem/progenitor cells in xenotransplanted NOD/SCID mouse model. Bone Marrow Transplant 2005; 35: 137-42. Zheng Y, Watanabe N, Nagamura-Inoue T, et al. Ex vivo manipulation of umbilical cord blood-derived hematopoietic stem/progenitor cells with recombinant human stem cell factor can up-regulate levels of homing-essential molecules to increase their transmigratory potential. Exp Hematol 2003; 31: 1237-46. Entschladen F, Zänker KS. Locomotion of tumor cells: a molecular comparison to migrating pre- and postmitotic leukocytes. J Cancer Res Clin Oncol 2000; 126: 671-81. Bleul CC, Fuhlbrigge RC, Casasnovas JM, Aiuti A, Springer TA. A highly efficacious lymphocyte chemoattractant, stromal cellderived factor 1 (SDF-1). J Exp Med 1996; 184: 1101-9. 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: 100-10. Ganju RK, Brubaker SA, Meyer J, et al. The alpha-chemokine, stromal cell-derived factor-1alpha, binds to the transmembrane G-protein-coupled CXCR-4 receptor and activates multiple signal transduction pathways. J Biol Chem 1998; 273: 23169-75. Wang JF, Park IW, Groopman JE. Stromal cell-derived factor1alpha stimulates tyrosine phosphorylation of multiple focal adhesion proteins and induces migration of hematopoietic progenitor cells: roles of phosphoinositide-3 kinase and protein kinase C. Blood 2000; 95: 2505-13. Vila-Coro AJ, Rodriguez-Frade JM, Martin De Ana A, et al. The chemokine SDF-1alpha triggers CXCR4 receptor dimerization and activates the JAK/STAT pathway. FASEB J 1999; 13: 1699710. Clark EA, Brugge JS. Integrins and Signal Transduction Pathways: The Road Taken. Science 1995; 268: 233-9. Dittmar T, Brandt BH, Lang K, Zänker KS, Entschladen F. Lessons from tumor and immunocompetent cells. The quantitative engagement of ligand-receptor interactions modulates stop-andgo behavior as well as proliferation. Medicina (B.Aires) 2000; 60 Suppl 2: 27-33. Dittmar T, Husemann A, Schewe Y, et al. Induction of cancer cell migration by epidermal growth factor is initiated by specific phosphorylation of tyrosine 1248 of c-erbB-2 receptor via EGFR. FASEB J 2002; 16: 1823-5. Petit I, Goichberg P, Spiegel A, et al. Atypical PKC-zeta regulates SDF-1-mediated migration and development of human CD34+ progenitor cells. J Clin Invest 2005; 115: 168-76. Mochly-Rosen D. Localization of Protein Kinases by Anchoring Proteins: A Theme in Signal Transduction. Science 1995; 268: 247-51. Hofmann J. The potential for isoenzyme-selective modulation of protein kinase C. FASEB J 1997; 11: 649-69. Cancelas JA, Lee AW, Prabhakar R, et al. Rac GTPases differentially integrate signals regulating hematopoietic stem cell localization. Nat Med 2005; 11: 886-91. Maheshwari G, Lauffenburger DA. Deconstructing (and reconstructing) cell migration. Microsc Res Tech 1998; 43: 35868. Friedl P, Noble PB, Zänker KS. Lymphocyte locomotion in threedimensional collagen gels. Comparison of three quantitative methods for analysing cell trajectories. J Immunol Methods 1993; 165: 157-65.

Weidt et al. [134]

[135] [136]

[137] [138] [139]

[140]

[141]

[142]

[143]

[144]

[145]

[146]

[147] [148] [149]

[150]

[151]

[152] [153]

Niggemann B, Drell TL 4th , Joseph J, et al. Tumor cell locomotion: differential dynamics of spontaneous and induced migration in a 3D collagen matrix. Exp Cell Res 2004; 298: 17887. Zaman MH, Kamm RD, Matsudaira P, Lauffenburger DA. Computational model for cell migration in three-dimensional matrices. Biophys J 2005; 89: 1389-97. Benito AI, Diaz MA, Gonzalez-Vicent M, Sevilla J, Madero L. Hematopoietic stem cell transplantation using umbilical cord blood progenitors: review of current clinical results. Bone Marrow Transplant 2004; 33: 675-90. Chao NJ, Emerson SG, Weinberg KI. Stem cell transplantation (cord blood transplants). Hematology (Am Soc Hematol Educ Program) 2004; 354-71. Habibian HK, Peters SO, Hsieh CC, et al. The fluctuating phenotype of the lymphohematopoietic stem cell with cell cycle transit. J Exp Med 1998; 188: 393-8. Glimm H, Oh IH, Eaves CJ. Human hematopoietic stem cells stimulated to proliferate in vitro lose engraftment potential during their S/G(2)/M transit and do not reenter G(0). Blood 2000; 96: 4185-93. Yamaguchi M, Ikebuchi K, Hirayama F, et al. Different adhesive characteristics and VLA-4 expression of CD34(+) progenitors in G0/G1 versus S+G2/M phases of the cell cycle. Blood 1998; 92: 842-8. Ramirez M, Segovia JC, Benet I, et al. Ex vivo expansion of umbilical cord blood (UCB) CD34(+) cells alters the expression and function of alpha 4 beta 1 and alpha 5 beta 1 integrins. Br J Haematol 2001; 115: 213-21. Cashman J, Clark-Lewis I, Eaves A, Eaves C. Stromal-derived factor 1 inhibits the cycling of very primitive human hematopoietic cells in vitro and in NOD/SCID mice. Blood 2002; 99: 792-9. Cashman J, Dykstra B, Clark-Lewis I, Eaves A, Eaves C. Changes in the proliferative activity of human hematopoietic stem cells in NOD/SCID mice and enhancement of their transplantability after in vivo treatment with cell cycle inhibitors. J Exp Med 2002; 196: 1141-9. Glimm H, Tang P, Clark-Lewis I, von Kalle C, Eaves C. Ex vivo treatment of proliferating human cord blood stem cells with stroma-derived factor-1 enhances their ability to engraft NOD/SCID mice. Blood 2002; 99: 3454-7. Gonzalo JA, Lloyd CM, Peled A, et al. Critical involvement of the chemotactic axis CXCR4/stromal cell-derived factor-1 alpha in the inflammatory component of allergic airway disease. J Immunol 2000; 165: 499-508. Eddleston J, Christiansen SC, Zuraw BL. Functional expression of the C-X-C chemokine receptor CXCR4 by human bronchial epithelial cells: regulation by proinflammatory mediators. J Immunol 2002; 169: 6445-51. Pashenkov M, Soderstrom M, Link H. Secondary lymphoid organ chemokines are elevated in the cerebrospinal fluid during central nervous system inflammation. J Neuroimmunol 2003; 135: 154-60. Pashenkov M, Teleshova N, Kouwenhoven M, et al. Recruitment of dendritic cells to the cerebrospinal fluid in bacterial neuroinfections. J Neuroimmunol 2002; 122: 106-16. Buckley CD, Amft N, Bradfield PF, et al. Persistent induction of the chemokine receptor CXCR4 by TGF-beta 1 on synovial T cells contributes to their accumulation within the rheumatoid synovium. J Immunol 2000; 165: 3423-9. Nanki T, Hayashida K, El-Gabalawy HS, et al. Stromal cellderived factor-1-CXC chemokine receptor 4 interactions play a central role in CD4+ T cell accumulation in rheumatoid arthritis synovium. J Immunol 2000; 165: 6590-8. Salvucci O, Basik M, Yao L, Bianchi R, Tosato G. Evidence for the involvement of SDF-1 and CXCR4 in the disruption of endothelial cell-branching morphogenesis and angiogenesis by TNF-alpha and IFN-gamma. J Leukoc Biol 2004; 76: 217-26. Fedyk ER, Jones D, Critchley HO, et al. Expression of stromalderived factor-1 is decreased by IL-1 and TNF and in dermal wound healing. J Immunol 2001; 166: 5749-54. Rao RM, Betz TV, Lamont DJ, et al. Elastase release by transmigrating neutrophils deactivates endothelial-bound SDF1alpha and attenuates subsequent T lymphocyte transendothelial migration. J Exp Med 2004; 200: 713-24.

Stem Cell Migration [154] [155] [156] [157]

[158]

[159] [160] [161] [162] [163]

[164]

[165]

[166] [167]

[168] [169] [170]

[171] [172]

[173]

Current Stem Cell Research & Therapy, 2007, Vol. 2, No. 1 103

McQuibban GA, Butler GS, Gong JH, et al. Matrix metalloproteinase activity inactivates the CXC chemokine stromal cell-derived factor-1. J Biol Chem 2001; 276: 43503-8. Jin DK, Shido K, Kopp HG, et al. Cytokine-mediated deployment of SDF-1 induces revascularization through recruitment of CXCR4+ hemangiocytes. Nat Med 2006; 12: 557-67. Askari AT, Unzek S, Popovic ZB, et al. Effect of stromal-cellderived factor 1 on stem-cell homing and tissue regeneration in ischaemic cardiomyopathy. Lancet 2003; 362: 697-703. Ratajczak MZ, Kucia M, Reca R, et al. Stem cell plasticity revisited: CXCR4-positive cells expressing mRNA for early muscle, liver and neural cells 'hide out' in the bone marrow. Leukemia 2004; 18: 29-40. Stumm RK, Rummel J, Junker V, et al. A dual role for the SDF1/CXCR4 chemokine receptor system in adult brain: isoformselective regulation of SDF-1 expression modulates CXCR4dependent neuronal plasticity and cerebral leukocyte recruitment after focal ischemia. J Neurosci 2002; 22: 5865-78. Mantovani A, Bottazzi B, Colotta F, Sozzani S, Ruco L. The origin and function of tumor-associated macrophages. Immunol Today 1992; 13: 265-70. Bingle L, Brown NJ, Lewis CE. The role of tumour-associated macrophages in tumour progression: implications for new anticancer therapies. J Pathol 2002; 196: 254-65. Balkwill F, Foxwell B, Brennan F. TNF is here to stay! Immunol Today 2000; 21: 470-1. Coussens LM, Werb Z. Inflammation and cancer. Nature 2002; 420: 860-7. Lyden D, Hattori K, Dias S, et al. Impaired recruitment of bonemarrow-derived endothelial and hematopoietic precursor cells blocks tumor angiogenesis and growth. Nat Med 2001; 7: 1194201. Young MR, Kolesiak K, Wright MA, Gabrilovich DI. Chemoattraction of femoral CD34+ progenitor cells by tumorderived vascular endothelial cell growth factor. Clin Exp Metastasis 1999; 17: 881-8. Young MR, Petruzzelli GJ, Kolesiak K, et al. Human squamous cell carcinomas of the head and neck chemoattract immune suppressive CD34(+) progenitor cells. Hum Immunol 2001; 62: 332-41. Udani VM, Santarelli JG, Yung YC, et al. Hematopoietic stem cells give rise to perivascular endothelial-like cells during brain tumor angiogenesis. Stem Cells Dev 2005; 14: 478-86. De Palma M, Venneri MA, Galli R, et al. Tie2 identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vessel formation and a mesenchymal population of pericyte progenitors. Cancer Cell 2005; 8: 211-26. Jang YY, Collector MI, Baylin SB, Diehl AM, Sharkis SJ. Hematopoietic stem cells convert into liver cells within days without fusion. Nat Cell Biol 2004; 6: 532-9. Ianus A, Holz GG, Theise ND, Hussain MA. In vivo derivation of glucose-competent pancreatic endocrine cells from bone marrow without evidence of cell fusion. J Clin Invest 2003; 111: 843-50. Newsome PN, Johannessen I, Boyle S, et al. Human cord bloodderived cells can differentiate into hepatocytes in the mouse liver with no evidence of cellular fusion. Gastroenterology 2003; 124: 1891-900. Dittmar T, Seidel J, Zänker KS, Niggemann B. Carcinogenesis Driven by Bone Marrow-Derived Stem Cells. Contrib Microbiol 2006; 13: 156-69. Geminder H, Sagi-Assif O, Goldberg L, et al. A possible role for CXCR4 and its ligand, the CXC chemokine stromal cell-derived factor-1, in the development of bone marrow metastases in neuroblastoma. J Immunol 2001; 167: 4747-57. Mochizuki H, Matsubara A, Teishima J, et al. Interaction of ligand-receptor system between stromal-cell-derived factor-1 and CXC chemokine receptor 4 in human prostate cancer: a possible predictor of metastasis. Biochem Biophys Res Commun 2004; 320: 656-63.

Received: June 28, 2006

[174]

[175] [176] [177] [178] [179]

[180] [181] [182]

[183]

[184] [185] [186]

[187] [188] [189] [190]

[191] [192] [193]

Revised: July 27, 2006

Bartolome RA, Galvez BG, Longo N, et al. Stromal cell-derived factor-1alpha promotes melanoma cell invasion across basement membranes involving stimulation of membrane-type 1 matrix metalloproteinase and Rho GTPase activities. Cancer Res 2004; 64: 2534-43. Vaupel P, Harrison L. Tumor hypoxia: causative factors, compensatory mechanisms, and cellular response. Oncologist 2004; 9 (Suppl 5): 4-9. Vaupel P, Mayer A, Hockel M. Tumor hypoxia and malignant progression. Methods Enzymol 2004; 381: 335-54. Staller P, Sulitkova J, Lisztwan J, et al. Chemokine receptor CXCR4 downregulated by von Hippel-Lindau tumour suppressor pVHL. Nature 2003; 425: 307-11. Asahara T, Murohara T, Sullivan A, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science 1997; 275: 964-7. Kopp HG, Ramos CA, Rafii S. Contribution of endothelial progenitors and proangiogenic hematopoietic cells to vascularization of tumor and ischemic tissue. Curr Opin Hematol 2006; 13: 175-81. Urbich C, Dimmeler S. Endothelial progenitor cells: characterization and role in vascular biology. Circ Res 2004; 95: 343-53. Takahashi T, Kalka C, Masuda H, et al. Ischemia- and cytokineinduced mobilization of bone marrow-derived endothelial progenitor cells for neovascularization. Nat Med 1999; 5: 434-8. Moore MA, Hattori K, Heissig B, et al. Mobilization of endothelial and hematopoietic stem and progenitor cells by adenovectormediated elevation of serum levels of SDF-1, VEGF, and angiopoietin-1. Ann N Y Acad Sci 2001; 938: 36-45; discussion -7. Hattori K, Dias S, Heissig B, et al. Vascular endothelial growth factor and angiopoietin-1 stimulate postnatal hematopoiesis by recruitment of vasculogenic and hematopoietic stem cells. J Exp Med 2001; 193: 1005-14. Bahlmann FH, De Groot K, Spandau JM, et al. Erythropoietin regulates endothelial progenitor cells. Blood 2004; 103: 921-6. Heeschen C, Aicher A, Lehmann R, et al. Erythropoietin is a potent physiologic stimulus for endothelial progenitor cell mobilization. Blood 2003; 102: 1340-6. Iwakura A, Luedemann C, Shastry S, et al. Estrogen-mediated, endothelial nitric oxide synthase-dependent mobilization of bone marrow-derived endothelial progenitor cells contributes to reendothelialization after arterial injury. Circulation 2003; 108: 3115-21. Laufs U, Werner N, Link A, et al. Physical training increases endothelial progenitor cells, inhibits neointima formation, and enhances angiogenesis. Circulation 2004; 109: 220-6. Li X, Tjwa M, Moons L, et al. Revascularization of ischemic tissues by PDGF-CC via effects on endothelial cells and their progenitors. J Clin Invest 2005; 115: 118-27. Bergers G, Brekken R, McMahon G, et al. Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat Cell Biol 2000; 2: 737-44. Engsig MT, Chen QJ, Vu TH, et al. Matrix metalloproteinase 9 and vascular endothelial growth factor are essential for osteoclast recruitment into developing long bones. J Cell Biol 2000; 151: 87989. Grunewald M, Avraham I, Dor Y, et al. VEGF-induced adult neovascularization: recruitment, retention, and role of accessory cells. Cell 2006; 124: 175-89. Kaplan RN, Riba RD, Zacharoulis S, et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the premetastatic niche. Nature 2005; 438: 820-7. Chavakis E, Aicher A, Heeschen C, et al. Role of beta2-integrins for homing and neovascularization capacity of endothelial progenitor cells. J Exp Med 2005; 201: 63-72.

Accepted: August 17, 2006