当前位置: 首页 > 期刊 > 《干细胞学杂志》 > 2005年第7期 > 正文
编号:11357836
Trafficking of Normal Stem Cells and Metastasis of Cancer Stem Cells Involve Similar Mechanisms: Pivotal Role of the SDF-1–CXCR4 Axis
http://www.100md.com 《干细胞学杂志》
     a Stem Cell Biology Program at James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA;

    b European Union Stem Cell Therapeutics Excellence Center, CMUJ, Krakow, Poland;

    c Department of Medicine, University of Alberta, Edmonton, Alberta, Canada

    Key Words. CXCR4 ? SDF-1 ? Stem cells ? Homing ? Metastasis

    Correspondence: Mariusz Z. Ratajczak, M.D., Ph.D., Director of Stem Cell Biology Program, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky 40202, USA. Telephone: 502-852-1788; Fax: 502-852-3032; e-mail: mzrata01@louisville.edu

    ABSTRACT

    Chemokines, small pro-inflammatory chemoattractant cytokines that bind to specific G-protein–coupled seven-span transmembrane receptors present on the plasma membranes of target cells, are the major regulators of cell trafficking . Some of them have also been reported to modulate cell survival and growth .

    More than 50 different chemokines and 20 different chemo-kine receptors have been cloned . Chemokines usually bind to multiple receptors, and the same receptor may bind to more than one chemokine. However, there is one exception to this rule: the -chemokine stromal-derived factor (SDF)-1, which binds exclusively to CXCR4 and has CXCR4 as its only receptor . This fact alone suggests that the SDF-1–CXCR4 axis plays a uniquely important biological role. Supporting this notion are murine knockout data showing that SDF-1 secreted by bone marrow (BM) stromal cells is critical for the colonization of BM by fetal liver-derived hematopoietic stem cells (HSCs) during embryogenesis . Furthermore, during adult life, it plays a pivotal role in the retention/homing of these cells into the BM microenvironment . Thus, it is not surprising that perturbation of the SDF-1–CXCR4 axis, for example, by mobilizing agents, is essential for the egress of hematopoietic stem/progenitor cells from BM into peripheral blood (PB) . On the other hand, proper functioning of the SDF-1–CXCR4 axis is crucial in directing homing/engraftment of HSC into BM after transplantation .

    However, what is often overlooked is that SDF-1–CXCR4 knockout mice, in addition to defects in colonization of BM by HSCs, also display defects in the development of heart, brain, and large vessels . We infer from this that the SDF-1–CXCR4 axis may have a more general role during organogenesis. Compelling evidence is accumulating from our and others’ work that stem cells for different organs and tissues (which we will refer to in this review as tissue-committed stem cells, TCSCs) resemble HSCs in expressing functional CXCR4 on their surfaces and because of this follow an SDF-1 gradient . Thus, from a developmental point of view, SDF-1 seems to be one of the most important motomorphogenic factors and chemoattractants not only for HSCs but for nonhematopoietic CXCR4+ tissue/organ–committed progenitor/stem cells.

    Concept of Cancer Stem Cells

    The evidence accumulates that quiescent TCSCs or cells developmentally closely related to them distributed in various organs may be a cellular origin of cancer development. To support this notion, stem cells are long-lived cells and thus become the subject of accumulating mutations that are crucial for initiation/progression of cancer. Thus, mutations that occur in normal stem cells lead to their malignant transformation and tumor initiation . The concept of cancer stem cells has been postulated by several investigators but was first experimentally documented for human leukemias . Recently, a stem cell origin of cancer was demonstrated for several solid tumors such as brain and breast cancers . Of note, because cancer stem cells, much like normal stem cells, exist in a quiescent state, they are relatively resistant to most cytostatics that target only dividing cells. Therefore, in a growing tumor, cancer stem cells represent a subpopulation of tumor cells that are capable of initiating metastasis and regrowth of new tumors after unsuccessful treatment.

    The fact that CXCR4 is expressed on normal stem cells for different organs/tissues may help to explain why several tumors that derived from these cells usually express CXCR4 on their surfaces (Table 1). This also implies that the SDF-1–CXCR4 axis may influence the biology of tumors and direct the metastasis of CXCR4+ tumor cells by chemoattracting them to organs that highly express SDF-1 (e.g., lymph nodes, lungs, liver, or bones). Supporting this notion, it has been recently reported that several CXCR4+ cancers (e.g., breast, ovarian, prostate cancers, rhabdomyosarcoma, and neuroblastoma) metastasize to the bones from the bloodstream in an SDF-1–dependent manner . In this review, we will focus on the role of the SDF–1–CXCR4 axis in regulating the trafficking/homing of normal stem cells and the metastasis of tumor stem cells and discuss the similarities of the molecular mechanisms involved in these processes. We postulate that the metastasis of cancer stem cells and trafficking/circulation of normal stem cells involve very similar processes.

    Table 1. Examples of CXCR4+ tumors that may derive from the normal CXCR4+ tissue/organ–specific stem cells

    CXCR4: A Marker of Normal and Malignant Stem Cells

    The role of the SDF-1–CXCR4 axis was initially extensively investigated with respect to hemato/lymphopoietic cells . It has been demonstrated that this axis regulates the trafficking/homing of CXCR4+ hematopoietic stem/progenitor cells, pre-B lymphocytes, and T lymphocytes . However, more recent evidence indicates that besides HSCs, functional CXCR4 is also expressed on the surface of various kinds of TCSCs , such as neural , skeletal muscle satellite , myocardial , endothelial , liver oval , and retinal pigment epithelium TCSCs, as well as primordial germ cells . Moreover, we recently found that functional CXCR4 is also expressed on murine embryonic stem cells (Fig. 1). Hence, CXCR4 can be considered a universal marker of versatile stem cells beginning with the population of pluripotent embryonic stem cells and ending with the population of TCSCs.

    Figure 1. Murine embryonic stem cells express functional CXCR4. (A): Negative reverse transcription–polymerase chain reaction; DNA instead of mRNA (lane 1). Expression of mRNA for CXCR4 (lane 2). Experiments were repeated three times with similar results. (B): Chemotaxis of ES-D3 cells to medium alone (–) or SDF-1 (300 ng/ml). The data are pooled together from three independent experiments (n = 12). Data are expressed as means +/– SD. * p < .00001 as compared with control. (C): Upper panel: Phosphorylation of MAPKp42/44 in ES-D3 cells. Cells were made quiescent and then stimulated with medium alone (lane 1) or for 10 minutes by SDF-1 (lane 2). Lower panel: total MAPK p42/44. Experiments were repeated three times with similar results. Abbreviation: SDF, stromal-derived factor.

    In support of the pivotal role of CXCR4 in stem cell trafficking during development, tissue injury, and regeneration, the expression of CXCR4 is regulated at the molecular level by several transcription factors related to organ development as well as those related to stress and tissue damage. With respect to the former, evidence from our laboratory shows that expression of CXCR4 is regulated in various tissues by paired-box transcription factors (PAX genes) . Sequence analysis of the CXCR4 promoter revealed several putative PAX gene–binding sites, and chromatin immunoprecipitation analysis confirmed that PAX genes bind to the CXCR4 promoter . Nine different PAX genes (PAX 1–9) have been described that bind to similar DNA consensus sequences. Because the pattern of expression of these genes is organ-specific, we propose that in the different TCSCs various PAX proteins may regulate CXCR4 expression in a tissue-specific manner (e.g., PAX 1 in osteoblastic TCSCs, PAX 3 in neural-crest and skeletal muscle TCSCs, PAX 5 in B lymphocytic progenitors, PAX 6 in endocrine pancreatic TCSCs, and PAX 7 in skeletal muscle satellite TCSCs). This would ensure the developmental responsiveness of TCSCs to an SDF-1 gradient and their proper trafficking during organogenesis. Interestingly, PAX genes are aberrantly expressed in several tumors that also highly express CXCR4 on their surface, such as rhabdomyosarcomas or small cell lung cancers . Besides PAX genes, CXCR4 expression may also be positively regulated by transcription factors related to stress/hypoxia and tissue damage such as NF-B , hypoxia-inducible factor (HIF-1) , glucocorticoids , lysophosphatidylcholine , TGF-?1 , VEGF , IFN- , and several interleukins (IL-2, IL-4, and IL-7) . Thus, it is likely that stress-related conditions may upregulate CXCR4 expression on both normal and malignant stem cells.

    SDF-1 as a Developmental and Postnatal Chemoattractant for Stem Cells

    The ligand for CXCR4, SDF-1 that is playing a crucial role in trafficking of normal and malignant cells has been reported in the literature to be expressed by BM endothelium, fibroblasts/osteoblasts, and functions in the homing/retention of CXCR4+ HSCs in the BM microenvironment . It is, however, also secreted by stromal and endothelial cells of other organs such as heart , skeletal muscle , liver , brain , and kidney . Moreover, its secretion increases during tissue damage such as heart infarct , limb ischemia , toxic liver damage , excessive bleeding , total body irradiation, and after-tissue damage related to chemotherapy . This suggests that SDF-1 may play a pivotal role in chemoattracting CXCR4+ TCSCs necessary for organ/tissue regeneration , implicating this chemokine as an important maintenance factor in postnatal tissue repair. Because a strong correlation exists between inflammation and tumor progression/metastasis, inflammation-driven expression of SDF-1 may also play an important role in dissemination/metastasis of cancer stem cells.

    In humans there are two identified splice variant forms of SDF-1: SDF-1 and SDF-1 ?. The former is more abundant than the latter, yet both are derived from a single gene. Studies at the molecular level show that SDF-1 expression is upregulated in endothelial cells by HIF-1 . Supporting this is the finding that the SDF-1 promoter contains two HIF-1 binding sites, and HIF-1 binds to these sequences, as revealed by chromatin immunoprecipitation analysis . Thus, HIF-1 elevates SDF-1 expression in hypoxic/damaged tissues, which leads to chemoattraction of CXCR4+ TCSCs that participate in tissue regeneration. Expression of SDF-1 has also been found to be positively regulated by NF-B . Therefore, SDF-1 appears to be an important indicator of tissue/organ hypoxia/injury and is involved in the active recruitment of TCSCs for repair of damaged tissue. Because, as stated previously, HIF-1 and NF-B also upregulate the expression of CXCR4 , these factors would appear to regulate the SDF-1–CXCR4 axis at both the ligand and receptor levels. This confers another important argument for a pivotal role of the SDF-1–CXCR4 axis in maintaining postnatal tissue repair.

    We may assume that the secretion of chemoattractants such as SDF-1 in or around injured tissue is a crucial event that creates an environment facilitating the homing of circulating TCSCs for endothelium and the affected tissue necessary for organ regeneration/tissue repair . A similar mechanism, however, may in our opinion induce a pro-metastatic environment for CXCR4+ tumor stem cells in damaged organs. There is the possibility that this could lead, for example, to unwanted side effects of radiochemotherapy, and we will discuss this topic later. Conversely, the expression of SDF-1 was found to be down-regulated by steroids and TGF-?1 . The downregulation of SDF-1 expression by steroids inhibits the accumulation of CXCR4+ inflammatory cells and contributes to the anti-inflammatory effect of these compounds . Thus, steroid therapy may be envisioned as a two-edged sword that not only inhibits trafficking of inflammatory cells but may inhibit homing of stem cells to the damaged organ.

    Negative and Positive Modulators of the SDF-1–CXCR4 Axis, Implications for Trafficking of Normal and Malignant Stem Cells

    It is evident that the function of the SDF-1–CXCR4 axis must be tightly regulated in vivo by various biological mechanisms, a fact that we are only now beginning to understand. Some of these mechanisms that may be relevant for trafficking of normal stem cells and metastasis of cancer cells will be discussed below.

    Both tissue damage that is a signal for chemoattracting normal stem cells for regeneration and chronic inflammation promoting the metastasis of cancer stem cells lead to expression/accumulation of several molecules/factors in the damaged tissues. We and others have recently identified several of these factors that positively affect the sensitivity/responsiveness of CXCR4+ cells to an SDF-1 gradient (Fig. 2). Factors such as (a) anaphylatoxin C3a (C3 complement protein cleavage fragment), (b) des-Arg C3a (product of C3a degradation by carboxypeptidase), (c) platelet-derived membrane microvesicles, (d) hyaluronic acid, and (e) sphingosine-1 phosphate were found to significantly increase the chemotaxis of CXCR4+ cells to low/threshold dosages of SDF-1 . Similarly, we found that several other molecules such as fibronectin, fibrinogen, thrombin, soluble uPAR, and VCAM-1 sensitize/increase the chemotactic responses of cells to low dosages of SDF-1 as well . These observations support the concept that the SDF-1–CXCR4 axis may be modulated by various molecules related to inflammation/tissue damage (e.g., C3a anaphylatoxin, des-ArgC3a, fibronectin, hyaluronic acid), coagulation (e.g., fibrinogen, uPAR, thrombin) or cell activation (e.g., s-VCAM-1, s-ICAM-1, membrane-derived vesicles) . Because inflammation plays an important role in tumor progression, molecules generated during inflammation (e.g., C3a, fibrinogen, fibronectin, and hyaluronic acid) could have an enhancing effect on the metastatic behavior of CXCR4+ tumor cells. Our preliminary data lend support to this notion .

    Figure 2. Modulation of the SDF-1–CXCR4 axis by external factors. The SDF-1–CXCR4 axis is modulated by several external factors. On one hand, leukocyte-derived proteases or cell surface–expressed CD26/dipeptidylpeptidase IV may cleave both SDF-1 and the N-terminus of CXCR4. On the other hand, the SDF-1–CXCR4 axis is regulated/primed both positively (e.g., by C3a, des-Arg C3a, thrombin, uPAR, fibrinogen, fibronectin, hyaluronic acid, sICAM1, sVCAM1, and cell membrane–derived microvesicles) and negatively (e.g., by polyene antibiotics, LPS, heparin, MIP-1, and RANTES). All these molecules are present in tissues affected by tissue damage/inflammation and may modulate the responsiveness of CXCR4+ normal and cancer stem cells to an SDF-1 gradient. Abbreviations: LPS, lipopolysaccharide; SDF, stromal-derived factor.

    To understand the phenomenon of priming at the molecular level, we showed that this sensitization/priming of HSC chemotaxis/homing to an SDF-1 gradient is dependent on the cholesterol content of the cell membrane and incorporation of CXCR4 and small GTP-ase Rac-1 into membrane lipid rafts . We found that this colocalization of CXCR4 and Rac-1 in lipid rafts facilitated GTP binding/activation of Rac-1. Because CXCR4 could be primed by factors related to leukapheresis/mobilization that increase its association with membrane lipid rafts (Fig. 1), allowing HSCs to better sense the SDF-1 gradient, this may explain in part why HSCs from mobilized PB engraft faster in patients than those from BM or cord blood . Our recent data show that similarly as observed for normal hematopoietic cells , depletion of membrane cholesterol severely affects responsiveness of CXCR4+ cancer cells to an SDF-1 gradient (manuscript in preparation). Thus, drugs that perturb lipid raft formation by depleting cholesterol from cells such as polyene antibiotics (e.g., amphotericin B, nystatin) could potentially negatively affect the meta-static properties of cancer cells. To support this notion further, we envision that one of the noted anticancer effects of statins , compounds that inhibit intracellular cholesterol synthesis, could also be explained by decreasing membrane cholesterol concentrations and as a result of this the formation of lipid rafts . This, however, requires further studies in vivo in animal models.

    Conversely, it has been reported that CXCR4 signaling may be desensitized in B lymphocytes and T lymphocytes by MIP-1? or RANTES, which activate another G-protein–coupled chemokine receptor, CCR5 . The molecular mechanism of this heterologous cross-desensitization between chemokine receptors is still unclear, but it is likely that it involves RGS proteins (regulators of G-protein signaling). Whether a similar desensitization mechanism also occurs on normal and malignant CXCR4+ HSCs and TCSCs requires more investigation. Interestingly, we found that the SDF-1–CXCR4 axis in HSCs in addition to polyene antibiotics (e.g., amphotericin B, nystatin) and statins may also be negatively modulated by soluble heparin and lipopolysaccharide (LPS). It is likely that, whereas heparin restricts SDF-1 availability in the intercellular environment, LPS probably interferes directly with the binding of SDF-1 to CXCR4 .

    Furthermore, the N-terminus of CXCR4 and its first extracellular loop are crucial for SDF-1 binding, and this explains why this part of the receptor is modulated by several mechanisms. Figure 1 shows different mechanisms by which the SDF-1–CXCR4 axis is modulated. Hyposulfation of N-terminal tyrosine residues or enzymatic processing/cleavage of the CXCR4 N-terminus by leukocyte-derived proteases inhibits CXCR4 signaling . Like CXCR4, SDF-1 may also be N-terminally truncated by cell-surface–expressed CD26/dipeptidylpeptidase IV . As a result, truncated (3–68 aa) SDF-1, in contrast to full-length SDF-1 (1–68 aa), does not possess chemotactic activity and may even act as an antagonist of CXCR4 . Thus, leukocyte-derived proteases released during inflammation may negatively affect the function of the SDF-1–CXCR4 axis in CXCR4+ cells such as monocytes and lymphocytes, which accumulate in tissues affected by inflammation. The same process, however, may also inhibit the homing responses of normal CXCR4+ stem cells. Thus, tissue damage/inflammation may exert pleiotropic effects on trafficking of stem cells. Accordingly, whereas damaged tissues on one hand secrete SDF-1 and other potential chemoattractants for stem cells, on the other hand their highly proteolytic environment may affect proper homing of circulating normal stem cells and affect their regenerative potential.

    Based on all these investigations, a new and more complex picture of the mechanisms that regulate the activity of the SDF-1–CXCR4 axis is emerging. The functionality of the CXCR4 receptor on normal and malignant cells is modulated by factors such as (a) the level of receptor expression on the cell surface, (b) the sulfation status of its N-terminus, (c) the expression and biological availability of SDF-1 in tissues, (d) cleavage of the CXCR4 N-terminus on the cells and SDF-1 in the extracellular space by serine proteases and matrix metalloproteinases (MMPs), (e) heterologous desensitization by the CCR5 receptor, and (f) the modulation of CXCR4 incorporation into membrane lipid rafts positively by molecules related to inflammation/tissue remodeling and negatively by polyene antibiotics (Fig. 2). Thus, all of these versatile mechanisms may modulate the responsiveness of CXCR4+ normal and malignant stem cells to an SDF-1 gradient and affect their trafficking or metastasis, respectively.

    The SDF-1–CXCR4 Axis Regulates Cell Trafficking: Biological Effects of SDF-1–Mediated Signaling

    Extensive studies have been done on the biological effects and signaling pathways of the SDF-1–CXCR4 axis in normal and malignant cells (Fig. 2). As reported, SDF-1’s major biological effects are related to the ability of this chemokine to induce (a) motility, (b) chemotactic responses, (c) adhesion, and (d) secretion of MMPs and angiopoietic factors (e.g., VEGF) in cells bearing cognate CXCR4 . SDF-1 also increases adhesion of cells to VCAM-1, ICAM-1, fibronectin, and fibrinogen by activating/modulating the function of several cell-surface integrins . There is some controversy regarding whether SDF-1, besides regulating the trafficking of cells, also directly affects their proliferation and survival. In some studies, SDF-1 has been reported to increase the proliferation of astrocytes and myeloid progenitor cells . Moreover, it has been suggested that SDF-1 may be secreted by hematopoietic stem/progenitor cells and be involved in autocrine/paracrine regulation of their development and survival . Interestingly, SDF-1 was also found to be expressed in solid tumors, for example, in some of human rhabdomyosarcoma cell lines .

    Generally, the SDF-1–CXCR4 axis activates several signaling pathways in target cells crucial for both their trafficking and interaction with the intercellular environment. Having discussed in the previous section the biological consequences of CXCR4 incorporation into membrane lipid rafts, we will now describe the signaling pathways in cells stimulated by SDF-1 which we summarized in Figure 3. It is believed that after binding to CXCR4, SDF-1 triggers CXCR4 dimerization . Shortly after being activated, CXCR4 becomes physically associated with Gi protein , and tyrosine residues within the C-terminus become phosphorylated, probably through activation and association with the receptor of JAK2 and JAK3 kinases . Subsequently, the activated CXCR4–SDF-1 complex is rapidly internalized from the cell surface in a mechanism involving G protein-coupled receptor kinases (GRKs) followed by the binding of ?-arrestin . Internalized CXCR4 may again be re-expressed on the cell surface. We recently found, however, that the internalization of CXCR4 from the surface of HSCs could be inhibited by heparin (not published). Likewise, a recent study reported inhibition of internalization of CXCR4 after crosslinking of L-selectin when specific antibodies were used or after the addition of L-selectin–binding ligands such as fucoidan or sulfatide . Besides receptor internalization during desensitization, which terminates receptor signaling, endocytosis of CXCR4 may be necessary to activate several pathways and functions such as chemotaxis and activation of the MEK kinase–MAPK p42/44 cascade . Recent experiments generated with mutated CXCR4 signaling domains do not, however, support this notion. Surprisingly, it was found that SDF-1–mediated phosphorylation of MAPK p42/44 also occurs in cells transfected with a mutant receptor that does not undergo internalization .

    Figure 3. Signal transduction pathways activated by the SDF-1–CXCR4 axis. Interaction of SDF-1 with G-protein–coupled seven-span transmembrane receptor CXCR4 activates several pathways in cells, with activation varying between cell types. Activation of these pathways in CXCR4+ cells (e.g., normal and malignant stem cells) regulates locomotion, chemotaxis, adhesion, and secretion. The potential direct effect of SDF-1 on cell proliferation/survival remains controversial. Abbreviation: SDF, stromal-derived factor.

    The most important pathways involved in signaling from activated CXCR4 include activation of calcium flux, focal adhesion components such as proline-rich kinase-2 (Pyk-2), Crk-associated substrate (p130Cas), focal adhesion kinase (FAK), paxillin, Nck, Crk, Crk-L, protein kinase C, phospholipase C- (PKC ), the MAPK p42/44–ELK-1 and PI-3K–AKT–NF-B axes . Recently, the major role of atypical isoform of PKC (PKC) has been proposed to play a crucial role in stem cell trafficking . Strong phosphorylation of focal adhesion components and MAPK p42/44 and serine-threonine kinase AKT is observed within seconds of CXCR4 activation . CXCR4 signaling also involves the Ras-activated signaling pathway, several src-related kinases such as Src, Lyn, Fyn, Lck, T-cell activating molecule ZAP-70, and vav . Recent evidence from other and our laboratories indicates the role of small GTPases in CXCR4 signaling. Activation of these GTPases is crucial for cell migration to an SDF-1 gradient . On the other hand, a recent report indicates that the RasGAP-associated docking protein p62Dok-1 negatively regulates chemotaxis of the Jurkat T-cell line .

    JAK2, JAK3 , and Tyk-2 may also associate in some cell types with CXCR4 and are activated, probably by transphosphorylation, in a Gi-independent manner . Consequently, several members of the STAT family of transcription factors may become recruited and phosphorylated. However, involvement of STAT proteins in signaling from activated CXCR4 may depend on the cell type. For example, whereas STAT 2 and 4 but not STAT 1, 3, 5, and 6 become activated in the hematopoietic CTS progenitor cell line , STAT 1, 2, 3, and 5 but not STAT 4 or 6 become activated in MOLT4 cells . Surprisingly, in our work we did not find that SDF-1 induces tyrosine phosphorylation of STAT 1, 3, 5 and 6, either in human CD34+ primary hematopoietic cells or in many established human lymphohematopoietic cell lines .

    Strong evidence exists that the protein-tyrosine phosphatases SHIP1 and SHIP2, as well as membrane-expressed hematopoietic phosphatase CD45, are also involved in the modulation of CXCR4 signaling . Hematopoietic cells derived from mice lacking SHIP1 showed altered patterns of chemotactic response to SDF-1 . SHIP2 also associated with CXCR4 and regulated SDF-1-induced migration of T and pre-B cells . Similarly, lymphocytes negative for CD45 phosphatase expression also show reduced chemotaxis to SDF-1. Moreover, after stimulation by SDF-1, CD45 was found to associate with CXCR4 within lipid rafts , an interaction that could be inhibited by pretreatment of cells with ?-cyclodextrin, an inhibitor of lipid raft formation.

    To better elucidate the role of CXCR4 domains in SDF-1–mediated signaling, studies were performed on a human embryonic kidney (HEK)-293 cell line stably expressing wild-type or mutated forms of three different intracellular loops or the C-terminus of CXCR4 . These data show that the third intracellular loop of CXCR4 alone supports the binding of Gi proteins and is involved in calcium mobilization and MAPK p42/44 activation but does not trigger CXCR4 internalization after SDF-1 binding. This latter observation indicates that MAPK p42/44 phosphorylation is independent of CXCR4 internalization. Furthermore, the second intracellular loop was found to be dispensable for Gi signaling. Interestingly, the second, third, and C-terminal intracellular domains were essential to transduce SDF-1–mediated chemotaxis . Finally, involvement/cooperation of several CXCR4 intracellular domains in this process suggests that chemotaxis is mediated by several complementary signaling events and not by activation of a single signaling pathway .

    No CXCR4/SDF-1 deficiencies have been described in humans. Based on murine knockout data, we assume such defects to be lethal in the embryo. In contrast, however, an activating mutation of CXCR4 in patients suffering from warts, hypogammaglobulinemia, immunodeficiency, and myelokathexis (WHIM) syndrome has been reported recently . The molecular basis of all these mutations are truncations in the C-terminus of CXCR4 . Lymphoblastoid cells derived from these patients have greater calcium flux than cells from normal donors, consistent with dysregulated signaling by the mutant receptor. This latter supports the idea that the C-terminal portion of CXCR4 is crucial for receptor desensitization and protects its hyperactivity . It would be interesting to determine whether the WHIM mutation has any impact on trafficking of HSCs and TCSCs.

    A New Perspective on BM as the Home of Circulating Normal CXCR4+ HSCs and CXCR4+ Tissue-Committed Stem/Progenitor Cells

    Below, we will present a concept that normal CXCR4+ stem/progenitor cells accumulate during ontogenesis in BM, circulate in PB, and compete for SDF-1–positive niches in peripheral tissues.

    Role of the SDF-1–CXCR4 Axis in Accumulation of CXCR4+ Stem/Progenitors in BM ? The BM tissue itself develops relatively late during ontogenesis at a time when fetal marrow replaces fetal liver as the hematopoietic organ. This developmental process establishes hematopoiesis in the human BM by the end of the second trimester of gestation. At this time, fibroblasts and osteoblasts in early bones begin to express/secrete SDF-1 that chemoattracts CXCR4+ HSCs from the fetal liver into BM . The important message from developmental studies is that CXCR4+ HSCs from the fetal liver colonize the BM microenvironment (to which they are chemoattracted by an SDF-1 gradient) as the first wave of CXCR4+ stem cells. Our recent data suggest that during ontogenesis, in addition to HSCs, other TCSCs (e.g., for muscles, neurons, liver, heart, endocrine pancreas, and kidney tubular epithelium) and perhaps even more primitive PSCs (precursors for various TCSCs) accumulate gradually in a SDF-1–dependent manner in the BM environment, where they find an environment conducive to their survival . Thus, BM can be envisioned not only as the home of HSCs but as the home of a small population of versatile CXCR4+ TCSCs that may serve in adult life as a reserve/mobile pool of stem cells for tissue/organ regeneration .

    To investigate this idea further, we hypothesized that CXCR4+ stem/progenitor cells residing in BM can be recovered, like CXCR4+ HSCs, from a suspension of BM mononuclear cells (BMMNCs) using chemotactic isolation to an SDF-1 gradient . To confirm that cells isolated by a chemotactic SDF-1 gradient are enriched in TCSCs, real-time reverse transcription–polymerase chain reaction (RT-PCR) and immunohistochemical staining were used to identify the isolated population of cells. We found that cells isolated in this way are enriched in mRNA for markers for early skeletal muscle (Myf-5, MyoD, myogenin), heart muscle (Nkx2.5/Csx, GATA-4, MEF-2C), neural (Nestin, GFAP), liver (CK19, -fetoprotein), and endocrine pancreas (Nkx6.1, Pdx1, Ptf1) . Furthermore, using immunohistochemical staining, we also detected in these cells the presence of various proteins characteristic of TCSCs, such as Myf-5, nestin, Nkx2.5/Csx, and GATA-4 . More important, the tissue/organ commitment and stem/progenitor nature of BM-derived CXCR4+ TCSCs was demonstrated in in vitro cultures, where these cells were found to be able to differentiate into cardiomyocytes , grow neurospheres , and form endothelial colonies . Of note, CXCR4+ BMMNCs also highly expressed mRNA for transcription factors of pluripotent stem cells (PSCs) such as mRNA for Oct-4, Nanog, and Rex-1, suggesting that some PSCs could possibly be present among cells enriched for TCSCs or TCSCs may express some markers typical of PSCs .

    These data not only explain a pivotal role of the SDF-1–CXCR4 axis in accumulating these cells in BM, but provide another explanation of the phenomenon of stem cell plasticity. Accordingly, because TCSCs have been found in BM aspirates, it may explain why it is possible to grow endothelial , skeletal muscle , heart , neural , liver , and endocrine pancreatic cells from BMMNCs . This may also explain the phenomenon of sex-mismatched BM transplants in humans that results in donor-derived hepatocytes and cholangiocytes , Purkinje cells , epidermal cells , or even intestinal epithelial cells . We envision that TCSCs accumulate during ontogenesis in BM as a reserve pool of stem cells for regeneration .

    The Role of the SDF-1–CXCR4 Axis in Postnatal Circulation of CXCR4+ Stem/Progenitor Cells ? However circulation of stem cells plays an important role during development, we believe that versatile stem cells may also circulate under physiological conditions at low levels in PB, thereby maintaining a pool of cells in distant parts of the body during postnatal life. This is especially important for hematopoietic, muscle, and neural stem cells that are distributed/reside in niches dispersed at distant locations around the body. The phenomenon of stem cell circulation in blood is well documented for HSCs , endothelial TCSCs , and recently for skeletal muscle satellite and skeleton TCSCs . However, the percentage of nonhematopoietic TCSCs in PB is significantly lower than that of early HSCs.

    Supporting this concept are our recent data that mRNA for several early markers for muscle (Myf-5, Myo-D), neural (GFAP, nestin), and liver (CK19, fetoprotein) can be detected in circulating (adherent cell-depleted) PBMNCs . Furthermore, we recently observed that the number of circulating CXCR4+ TCSCs in PB is increased by the administration of agents similar to those used for mobilization of HSCs (e.g., G-CSF) or triggered by stress factors related to tissue/organ injury (e.g., heart infarct or stroke) . Thus, pharmacological mobilization could be envisioned not only as a tool for increasing the number of HSCs in PB, but as a process that mobilizes TCSCs into PB. TCSCs, in addition to BM, are probably also mobilized into PB from various tissue/organ–specific niches. Furthermore, an increase in the number of these cells in PB during tissue/organ injury supports the idea that they play an important role in tissue repair. Therefore, it is likely that regeneration processes involve the recruitment not only of local stem/progenitor cells that reside in the damaged organ, but of TCSCs (specific for the damaged organ) that reside in distant niches of the same tissue/organ (e.g., in remote healthy muscle tissue) or, as we recently postulated, TCSCs that reside as a reserve pool in BM .

    The Role of the SDF-1–CXCR4 Axis in Postnatal Tissue Distribution of CXCR4+ Stem/Progenitor Cells ? Because TCSCs are CXCR4+, these cells respond to an SDF-1 gradient, and there is growing evidence that the SDF-1–CXCR4 axis plays an important role in regulating their trafficking. SDF-1 is not only constitutively expressed by BM fibroblasts/osteoblasts and enriched in BM stem cell niches , but expressed in stem cell niches located in various tissues/organs; this may explain the presence of heterogenous populations of CXCR4+ TCSCs in various organs such as HSCs in muscle or neural tissue and, conversely, muscle or neural progenitors in BM .

    In support of this concept is our recent finding that both CXCR4+ muscle stem/progenitor satellite cells as well as CXCR4+ HSCs could be chemoattracted in an SDF-1–dependent manner by media conditioned by either BM or muscle fibroblasts . The effect of these media was less visible in our study when (a) the media were precleared by anti-SDF-1 antibodies, (b) CXCR4 on HSPCs or muscle satellite cells was blocked by antiCXCR4 antibodies, or (c) cells were preincubated with the small peptide CXCR4 antagonist T140 before chemotaxis . These data provide evidence that both HSCs and muscle satellite TCSCs follow an SDF-1 gradient. Because neural, oval liver, and myocardium TCSCs are also chemoattracted by SDF-1, this chemokine alone or in combination with other factors appears to affect the homing of circulating stem cells into tissue-specific niches. Thus, the concept of circulating CXCR4+ stem cells and their competition for SDF-1–positive niches explains why stem cells specific to other organs are detectable in certain organs/tissues in addition to the organ-specific stem cells . Of course, this does not mean that other tissue-specific chemoattractants are not involved, which would account for why circulating stem cells have a higher affinity for niches in organs to which they are committed .

    The SDF-1–CXCR4 Axis Regulates Crucial Steps Involved in Mobilization, Trafficking, and Homing of Normal and Metastasis of Malignant Stem Cells

    Mobilization/trafficking/homing of normal stem cells and metastasis of malignant stem cells is a multistep process (Fig. 4). First, normal stem cells that egress from their tissue/organ niches or cancer stem cells that will establish distant metastases have to detach from the primary tumor mass and enter the PB or lymph. At a certain point, they sense a chemoattracting gradient that directs their homing (normal stem cell) or metastasis (cancer stem cell). On receiving this signal, they actively attach to the endothelium and subsequently penetrate the microvessel wall in a mechanism involving the secretion of metalloproteinases (MMPs). At the new distant site, stem cells may engraft and find an environmental niche that will protect them from apoptosis and allow them to expand to regenerate damaged tissue or establish a metastatic tumor (normal and cancer stem cells, respectively) (Fig. 4). The role of the SDF-1–CXCR4 axis in the consecutive steps of stem cell trafficking will be discussed below.

    Figure 4. The role of the SDF-1–CXCR4 axis in migration/circulation of normal stem cells and metastasis of cancer stem cells. Migration of normal stem cells and metastasis of malignant stem cells is a multistep process in which cells (I) leave their stem cell niches (normal stem cells) or primary tumor (cancer stem cells) and enter circulation, (II) arrive at the site of homing (normal stem cells) or metastasis (malignant stem cells) via the peripheral blood or lymph, (III) adhere to the endothelium, (IV) invade tissues, and (V) proliferate and expand at a location that provides a supportive environment for them. We hypothesize that SDF-1 plays a crucial role in this process, chemoattracting CXCR4+ normal or tumor stem cells. Abbreviations: SC, stem cell; SDF, stromal-derived factor.

    The SDF-1–CXCR4 Axis and Mobilization/Egress of Stem Cells ? To get into circulation, a normal stem cell has to egress from its niche in the organ or, as in the case of a cancer stem cell, detach from the primary tumor (Fig. 4). The influence of SDF-1 on detachment/egress of cells from their tissue niches was studied best in a model of mobilization of normal HSPCs . This intriguing process, however, is not yet fully understood. Mobilization may be enhanced by both decreasing the concentration of endogenous SDF-1 (e.g., after infusion of G-CSF or cyclophosphamide) in BM or, in contrast, by stimulating BM with exogenous SDF-1 . In the first event, proteolytic enzymes released by marrow accessory cells may degrade SDF-1 expressed in BM and inactivate CXCR4 by partial proteolysis. As a result, cells will be released from BM into PB. In the second scenario, exogenous SDF-1 may stimulate the release of MMP-9 by HSPCs and consequently enhance their translocation from the endosteal niche to the endothelial niche within the BM environment . HSPCs residing in the endothelial niche could subsequently be easily mobilized into PB by responding to a transendothelial SDF-1 gradient. To further support this last possibility, small peptide analogs of SDF-1 (e.g., CTCE0021) were recently used for mobilization . An interesting phenomenon was also recently described, so called chemofugotaxis/reverse chemotaxis to SDF-1 . Whether these mechanisms are also involved in the release/egress of the metastasizing cancer cells from the primary tumor is an open question.

    The SDF-1–CXCR4 Axis, Cell Motility, and Directed Chemotaxis ? After they reach circulation, stem cells respond to chemoattractants that direct them to a new stem cell niche or, as in the case of cancer stem cells, to the organ where they may metastasize. SDF-1 is a potent chemoattractant for normal and malignant stem cells . SDF-1 both chemoattracts cells and increases their motility . This effect on cell motility corresponded to changes in or rearrangements of cytoskeletal proteins, and cells exposed to SDF-1 display significant increases in the number and thickness of F-actin bundles. Moreover, stimulation of cells by SDF-1 also led to the formation of a leading edge in migrating tumor cells. By using time-lapse monitoring of the locomotion of individual cells, we were able to observe that SDF-1 increases the motility of several human tumor cells (e.g., breast cancer, rhabdomyosarcoma, and small lung cancer cell lines) . Tumor cells migrated extensively in the presence of SDF-1, which also increased the final displacement and the average velocity of cell displacement .

    Furthermore, SDF-1, just as in CXCR4+ stem cells, has been reported to induce a directional chemotaxis of several human solid tumor cell lines . The stimulation of cell movement by SDF-1 (motility and directional migration) was inhibited in normal CD34+ hematopoietic cells as well as in several established cell lines after the PI-3K–AKT axis was blocked using Ly290042 or wortmanin and/or after inhibition of Gi signaling by preincubating cells with pertussis toxin . This role of the PI-3K–AKT pathway in cell motility is further indicated by the observation that murine cells with a disruption of a single gene that encodes Pten (phosphatase and tensin homologue), a negative regulator of AKT phosphorylation/activation, shows enhanced phosphorylation of AKT, and as a result enhanced chemotaxis to SDF-1 . SDF-1–mediated cell motility and chemotaxis, however, are affected by several other signaling events, probably involving MAPK p42/44 and phosphatases . As reported recently, whereas the chemotaxis of normal HSCs was dependent on signaling through an atypical isoform of PKC, the chemotaxis of the Jurkat T-cell line was positively regulated by nonreceptor tyrosine kinase Pyk2 and negatively by p120RasGAP-associated p62 protein Dok-1 . As we previously discussed, the second, third, and C-terminal intracellular domains of CXCR4 are needed to transduce signals involved in SDF-1–mediated chemotaxis .

    The SDF-1–CXCR4 Axis and Cell Adhesion ? Circulating stem cells have to adhere to the endothelium in the organ where they will home or metastasize (Fig. 4). It is known that SDF-1 modulates adhesion of cells to fibrinogen, fibronectin, stroma, and endothelial cells , an effect that is mediated by the activation of various adhesion molecules (e.g., integrins) on the surface of target cells rather than by an increase in their de novo expression on the cell surface . In accordance with this, SDF-1 has been reported to activate integrins LFA (lymphocyte function-associated antigen)-1, VLA (very late activation antigen)-4, and VLA-5 on human HSCs . Lending further support to this evidence, we found that SDF-1 also activates IIb/?3 integrin (CD41) on the surface of megakaryocytic cells . Furthermore, SDF-1 induces firm adhesion and transendothelial migration of human CD34+ hematopoietic cells which are dependent on LFA-1/intracellular adhesion molecule (ICAM)-1 and VLA-4/vascular adhesion molecule (VCAM)-1 interactions. These interactions could be inhibited by pertussis toxin and cytochalasin D, indicating the involvement of Gi protein downstream signaling and the requirement of an intact cytoskeleton .

    Interestingly, an alternative possibility was recently suggested postulating that adhesion molecules may trigger signals for both enhanced CXCR4 expression and increased function . Activation of L-selectin on the surface of CXCR4+ cells upregulated both expression of and signaling from CXCR4. In light of these findings, we infer that interactions between CXCR4 and adhesion molecules are more complex than initially thought and seem in fact to constitute a two-way street. In the model proposed, CXCR4 signaling regulates the function of adhesion molecules on the cell surface, and, in turn, activation of certain adhesion molecules may enhance the signaling/expression of CXCR4 .

    By means of similar mechanisms, SDF-1 may also increase the adhesion of tumor cells . To support this notion, SDF-1 has been demonstrated, for example, to increase adhesion of neuroblastoma cells activated by TNF- and INF- HUVEC , prostate cancer cells to osteosarcoma layer and marrow-derived endothelium , and rhabdomyosarcoma cells to laminin and fibronectin . Furthermore, small lung cancer cells were reported to adhere to BM-derived stroma in response to SDF-1 in a VLA-4/VCAM-1–dependent manner .

    The SDF-1–CXCR4 Axis and Cell Secretion ? Circulating stem cells have to release factors (e.g., MMPs) that will enable them to cross the endothelial barrier and home to the organ (Fig. 4). Evidence has accumulated that, under certain circumstances, normal and malignant stem cells may express and secrete several proteins such as growth factors, cytokines, or proteolytic enzymes. These factors allow stem cells to interact with their surrounding environment as well as play some role in autocrine/paracrine regulation of the stem cell pool in niches . Interactions of CXCR4 with SDF-1 lead to the activation of NF-B, which regulates cell secretion. It has been shown that normal and tumor cells secrete more MMPs (e.g., MMP-2 and MMP-9), nitric oxide, and certain angiopoietic factors such as VEGF after stimulation by SDF-1 . These factors/enzymes secreted by target cells are involved in migration of HSCs and tumor cells across vascular basement membranes (e.g., MMPs), in tumor vasculogenesis, and in crosstalk between CXCR4+ cells and endothelium (e.g., VEGF, HGF). These interactions are crucial both for trafficking of normal stem cells and for metastasis of cancer stem cells. Interestingly, both normal early stem/progenitor cells as well as cancer cells secrete several factors that promote angiogenesis. Thus, SDF-1 plays an important role in angiogenesis both by directly chemoattracting CXCR4+ endothelial TCSCs as well as by stimulating cells to secrete other angiopoietic factors (e.g., VEGF) . Similar mechanisms may operate during tumor vasculogenesis, in which SDF-1 has been reported to be upregulated in tumor and tumor-surrounding tissues under steady state conditions as well as after radiochemotherapy . Thus, in response to SDF-1 stimulation, cancer cells may also secrete several proangiopoietic factors and metalloproteinases .

    The SDF-1–CXCR4 Axis on Cell Proliferation and Survival ? Finally, normal stem cells or cancer stem cells have to survive/expand in the new distant tissue location (Fig. 4). Several chemokines have been reported to positively or negatively modulate cell growth. It has been suggested that the ratio between activated p38 stress kinase (negative regulator) and the PI-3K–AKT axis (positive regulator) as a result of chemokine receptor activation determines whether a particular chemokine will support or inhibit cell survival . Nevertheless, the direct effect of SDF-1 on cell proliferation and survival remains controversial. For example, in contrast to several other chemokines such as MIP-1 and PF-4 that have been reported to inhibit the proliferation of early hematopoietic cells , SDF-1 has been found to stimulate the proliferation and survival of hematopoietic cells under certain experimental conditions . As we previously discussed, SDF-1 has been reported to be an autocrine survival factor for purified CD34+ CD38+ BMMNCs and its prosurvival effect is PI-3K–AKT–axis dependent .

    Nevertheless, when we evaluated the effect of SDF-1 on normal human CD34+ stem/progenitor cells, lineage-expanded erythroblasts, megakaryoblasts, and myeloid cells, we found that SDF-1 did not affect cell survival or proliferation in any of them . Similarly, we did not observe an effect of SDF-1 on proliferation/survival in 26 different established human T and B lymphoid and myeloid cell lines . However, in several of these cell lines as well as in normal human CD34+ cells and megakaryoblasts, SDF-1 induced phosphorylation of MAPK p42/44 and serine-threonine kinase AKT, which are widely accepted as being associated with cell survival/proliferation.

    Interestingly, SDF-1 was found to stimulate the proliferation and survival of astrocytes and some tumor cell lines . The proliferation effect of SDF-1 on astrocytes was MAPK p42/44-dependent and sensitive to inhibition by PD98059, an inhibitor of MEK kinase which activates MAPK p42/44 . Some tumor cells (e.g., prostate cancer) were found to secrete SDF-1, and an autocrine SDF-1–CXCR4 interaction had been suggested to regulate the proliferation/survival of these cells . Moreover, SDF-1 was found to be a survival factor for glioma and glioblastoma cells, an effect that has been correlated with prolonged activation of AKT and MAPK p42/44 .

    On the other hand, several other tumor cell lines did not respond to stimulation by SDF-1 by proliferation or increased survival, making it very likely that SDF-1 is not directly involved in growth of HSCs, TCSCs, or cancer stem cells expressing CXCR4. It is possible, however, that SDF-1’s effect on cell proliferation/survival is an indirect one. By increasing cell adhesion, SDF-1 may increase cell survival of cells through signaling from adhesion contact molecules or by directing their migration may expose cells to other prosurvival factors that are coexpressed with SDF-1 in tissues. It is well known that signals generated from adhesion molecules prevent cells from undergoing apoptosis in a mechanism described as "anoikis" . Lending support to this idea is the fact that hematopoietic cells from SDF-1 transgenic mice display both enhanced survival in vitro in response to growth factor withdrawal and enhanced myelopoiesis in vivo . Finally, it is possible that SDF-1 increases cell proliferation by enhancing secretion of other autocrine growth factors.

    Implications for the Future

    The SDF-1–CXCR4 axis has emerged as an important regulator of trafficking of normal and malignant stem cells, which means that it is potentially a target for various therapeutic interventions. Modulation of this axis may be important for (a) enhancing homing of stem cells for tissue/organ regeneration, (b) mobilization of normal hematopoietic and nonhematopoietic stem cells into PB, and (c) inhibiting metastasis of CXCR4+ cancer stem cells.

    Enhancement of stem cell homing could be accomplished by employing several recently identified priming agents of the SDF-1–CXCR4 axis, such as C3a, des-Arg C3a, or hyaluronic acid, before transplantation to prime the SDF-1–dependent homing of HSPCs into BM . Such pharmacological modulation of CXCR4 signaling may also play an important role in chemoattraction of CXCR4+ TCSCs and regeneration of damaged organs and tissues by these cells. Based on this, we expect, for example, that in the future, homing and engraftment efficiency of HSCs after transplantation can be significantly improved by the use of drugs containing molecules that prime/increase the responsiveness of HSCs to an SDF-1 gradient. This strategy would be especially important in cord blood transplantation, in which the number of HSCs is limited, especially for reconstituting an adult recipient.

    On the other hand, several inhibitors of the CXCR4–SDF-1 axis could be used as new drugs to increase the mobilization of HSCs and nonhematopoietic stem cells into PB. Small-molecular inhibitors of CXCR4 such as T140 and AMD 3,100, modified recombinant SDF-1, blocking antibodies against CXCR4 or molecules perturbing incorporation of CXCR4 into membrane lipid formation (e.g., statins) are examples of such potential compounds .

    All of these blocking strategies may also have value as potent new antimetastatic drugs for CXCR4+ cancer cells, but there needs to be a careful assessment of the possible side effects resulting from such therapy that might occur during trafficking and development of hemato/lymphopoietic stem cells, maturation of lymphocytes, and/or platelet formation. Encouragingly, the first clinical trials in HIV-infected patients using blocking agents (to prevent binding of HIV to CXCR4) have proven pharmacologically safe . Strategies based on utilization of antisense oligodeoxynucleotides or double-stranded RNA-mediated interference (RNAi) could also have potential as downregulators of the expression of CXCR4 in target cells . Similarly, downregulation of HIF-1 by an siRNA strategy may lead to downregulation of SDF-1 in various tissues and inhibit spreading of CXCR4+ tumor cells . Recently, a small molecular inhibitor of transcriptional coactivation of HIF-1, called chetomin, has been identified. Interestingly, systemic administration of chetomin inhibited hypoxia-inducible transcription within tumors and inhibited tumor growth in mice .

    In this review, we have focused on a role of the SDF-1–CXCR4 axis in regulation of cell trafficking. This process, however, is regulated by several other regulatory axes that together are responsible for the efficacy and tissue specificity of this process. The most important motomorphogens that are involved, beside SDF-1, in regulating cell trafficking are hepatocyte growth factor/scatter factor , vascular endothelial growth factor , basic fibroblast growth factor , kit ligand , and leukemia inhibitory factor . We are on the beginning of a long journey to understand how all of these factors orchestrate stem cell migration.

    CONCLUSION

    We deeply apologize that because of the space limitation we were not able to cite all the excellent work of our colleagues and other investigators working in this field. This study was supported by Kentucky Lung Cancer Research Fund grant and National Institutes of Health grant R01 CA106281-01 to M.Z.R. and a CIHR grant to A.J.W.

    REFERENCES

    Schier AF. Chemokine signaling: rules of attraction. Curr Biol 2003;13: R192–R194.

    Broxmeyer HE, Cooper S, Kohli L et al. Transgenic expression of stromal cell-derived factor-1/CXC chemokine ligand 12 enhances myeloid progenitor cell survival/antiapoptosis in vitro in response to growth factor withdrawal and enhances myelopoiesis in vivo. J Immunol 2003;170:421–429.

    Zlotnik A, Yoshie O. Chemokines: a new classification system and their role in immunity. Immunity 2000;12:121–127.

    Horuk R. Chemokine receptors. Cytokine Growth Factor Rev 2001;12:313–335.

    Nagasawa T, Hirota S, Tachibana K et al. Defects of B-cell lymphopoiesis and bone-marrow myelopoiesis in mice lacking the CXC chemokine PBSF/SDF-1. Nature 1996;382:635–638.

    Ma Q, Jones D, Springer TA. The chemokine receptor CXCR4 is required for the retention of B lineage and granulocytic precursors within the bone marrow microenvironment. Immunity 1999;10:463–471.

    Bagri A, Gurney T, He X et al. The chemokine SDF-1 regulates migration of dentate granule cells. Development 2002;129:4249–4260.

    Lazarini F, Tham TN, Casanova P et al. Role of the alpha-chemokine stromal cell-derived factor (SDF-1) in the developing and mature central nervous system. Glia 2003;42:139–148.

    Zou Y, Kottmann AH, Kuroda M et al. Function of the chemokine receptor CXCR4 in haematopoiesis and in cerebellar development. Nature 1998;393:595–599.

    Tachibana K, Hirota S, Iizasa H et al. The chemokine receptor CXCR4 is essential for vascularization of the gastrointestinal tract. Nature 1998;393:591–594.

    Petit I, Szyper-Kravitz M, Nagler A et al. G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and up-regulating CXCR4. Nat Immunol 2002;3:687–694.

    Liles WC, Broxmeyer HE, Rodger E et al. Mobilization of hematopoietic progenitor cells in healthy volunteers by AMD3100, a CXCR4 antagonist. Blood 2003;102:2728–2730.

    Devine SM, Flomenberg N, Vesole DH et al. Rapid mobilization of CD34+ cells following administration of the CXCR4 antagonist AMD3100 to patients with multiple myeloma and non-Hodgkin’s lymphoma. J Clin Oncol 2004;22:1095–1102.

    Peled A, Petit I, Kollet Oetal. Dependence of human stem cell engraftment and repopulation of NOD/SCID mice on CXCR4. Science 1999;283:845–848.

    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.

    Kucia M, Ratajczak J, Ratajczak MZ. Bone marrow as a source of circulating CXCR4+ tissue committed stem cells (TCSC). Biol Cell 2005;97:133–146.

    Tavor S, Petit I, Porozov S et al. CXCR4 regulates migration and development of human acute myelogenous leukemia stem cells in transplanted NOD/SCID mice. Cancer Res 2004;64:2817–2824.

    Pardal R, Clarke MF, Morrison SJ. Applying the principles of stem-cell biology to cancer. Nat Rev Cancer 2003;3:895–902.

    Reya T, Morrison SJ, Clarke MF et al. Stem cells, cancer, and cancer stem cells. Nature 2001;414:105–111.

    Lapidot T, Pflumio F, Doedens M et al. Cytokine stimulation of multilineage hematopoiesis from immature human cells engrafted in SCID mice. Science 1992;255:1137–1141.

    Singh SK, Hawkins C, Clarke ID et al. Identification of human brain tumour initiating cells. Nature 2004;432:396–401.

    Dontu G, Al-Hajj M, Abdallah WM et al. Stem cells in normal breast development and breast cancer. Cell Prolif 2003;36(suppl 1):59–72.

    Muller A, Homey B, Soto H et al. Involvement of chemokine receptors in breast cancer metastasis. Nature 2001;410:50–56.

    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;89:462–473.

    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;167:4747–4757.

    Porcile C, Bajetto A, Barbero S et al. CXCR4 activation induces epidermal growth factor receptor transactivation in an ovarian cancer cell line. Ann N Y Acad Sci 2004;1030:162–169.

    Hall JM, Korach KS. Stromal cell-derived factor 1, a novel target of estrogen receptor action, mediates the mitogenic effects of estradiol in ovarian and breast cancer cells. Mol Endocrinol 2003;17:792–803.

    Libura J, Drukala J, Majka M et al. CXCR4-SDF-1 signaling is active in rhabdomyosarcoma cells and regulates locomotion, chemotaxis, and adhesion. Blood 2002;100:2597–2606.

    Jankowski K, Kucia M, Wysoczynski M et al. Both hepatocyte growth factor (HGF) and stromal-derived factor-1 regulate the metastatic behavior of human rhabdomyosarcoma cells, but only HGF enhances their resistance to radiochemotherapy. Cancer Res 2003;63:7926–7935.

    Aiuti A, Webb IJ, Bleul C et al. The chemokine SDF-1 is a chemoattractant for human CD34+ hematopoietic progenitor cells and provides a new mechanism to explain the mobilization of CD34+ progenitors to peripheral blood. J Exp Med 1997;185:111–120.

    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 U S A 1998;95:9448–9453.

    Kim CH, Broxmeyer HE. Chemokines: signal lamps for trafficking of T and B cells for development and effector function. J Leukoc Biol 1999;65:6–15.

    Rosu-Myles M, Gallacher L, Murdoch B et al. The human hematopoietic stem cell compartment is heterogeneous for CXCR4 expression. Proc Natl Acad Sci U S A 2000;19:14626–14631.

    Pituch-Noworolska A, Majka M, Janowska-Wieczorek A et al. Circulating CXCR4+ stem/progenitor cells compete for SDF-1 positive niches in bone marrow, muscle and neural tissues: an alternative hypothesis to stem cell plasticity. Folia Histochem Cytobiol 2003;41:13–21.

    Ratajczak MZ, Majka M, Kucia M et al. Expression of functional CXCR4 by muscle satellite cells and secretion of SDF-1 by muscle-derived fibroblasts is associated with the presence of both muscle progenitors in bone marrow and hematopoietic stem/progenitor cells in muscles. STEM CELLS 2003;21:363–371.

    Damas JK, Eiken HG, Oie E et al. Myocardial expression of CC- and CXC-chemokines and their receptors in human end-stage heart failure. Cardiovasc Res 2000;47:778–787.

    Kucia M, Dawn B, Hunt G et al. Cells Expressing Early Cardiac Markers Reside in the Bone Marrow and Are Mobilized Into the Peripheral Blood After Myocardial Infarction. Circ Res 2004;95:1191–1199.

    Peichev M, Naiyer AJ, Pereira D et al. Expression of VEGFR-2 and AC133 by circulating human CD34(+) cells identifies a population of functional endothelial precursors. Blood 2000;95:952–958.

    Yamaguchi J, Kusano KF, Masuo O et al. Stromal cell-derived factor-1 effects on ex vivo expanded endothelial progenitor cell recruitment for ischemic neovascularization. Circulation 2003;107:1322–1328.

    Wojakowski W, Tendera M, Michalowska A et al. Mobilization of CD34/CXCR4+, CD34/CD117+, c-met+ stem cells, and mononuclear cells expressing early cardiac, muscle, and endothelial markers into peripheral blood in patients with acute myocardial infarction. Circulation 2004;110:3213–3220.

    Hatch HM, Zheng D, Jorgensen ML et al. SDF-1alpha/CXCR4: a mechanism for hepatic oval cell activation and bone marrow stem cell recruitment to the injured liver of rats. Cloning Stem Cells 2002;4:339–351.

    Crane IJ, Wallace CA, McKillop-Smith S et al. CXCR4 receptor expression on human retinal pigment epithelial cells from blood-retina barrier leads to chemokine secretion and migration in response to stromal cell-derived factor. J Immunol 2000;165:4372–4378.

    Ara T, Nakamura Y, Egawa T et al. Impaired colonization of the gonads by primordial germ cells in mice lacking a chemokine stromal cell-derived factor-1 (SDF-1). Proc Natl Acad Sci U S A 2003;100:5319–5323.

    Reca R, Jankowski K, Przybylski G et al. CXCR4 is a PAX family transcription factor regulated gene. Blood 2004;104a.

    Tomescu O, Xia SJ, Strezlecki D et al. Inducible short-term and stable long-term cell culture systems reveal that the PAX3-FKHR fusion oncoprotein regulates CXCR4, PAX3, and PAX7 expression. Lab Invest 2004;84:1060–1070.

    Barr FG. Gene fusions involving PAX and FOX family members in alveolar rhabdomyosarcoma. Oncogene 2001;20:5736–5746.

    Helbig G, Christopherson KW 2nd, Bhat-Nakshatri P et al. NF-B promotes breast cancer cell migration and metastasis by inducing the expression of the chemokine receptor CXCR4. J Biol Chem 2003;278:21631–21638.

    Schioppa T, Uranchimeg B, Saccani A et al. Regulation of the chemokine receptor CXCR4 by hypoxia. J Exp Med 2003;198:1391–1402.

    Curnow SJ, Wloka K, Faint JM et al. Topical glucocorticoid therapy directly induces up-regulation of functional CXCR4 on primed T lymphocytes in the aqueous humor of patients with uveitis. J Immunol 2004;172:7154–7161.

    Han KH, Hong KH, Ko J et al. Lysophosphatidylcholine up-regulates CXCR4 chemokine receptor expression in human CD4 T cells. J Leukoc Biol 2004;76:195–202.

    Franitza S, Kollet O, Brill A et al. TGF-beta1 enhances SDF-1alpha-induced chemotaxis and homing of naive T cells by up-regulating CXCR4 expression and downstream cytoskeletal effector molecules. Eur J Immunol 2002;32:193–202.

    Bachelder RE, Wendt MA, Mercurio AM. Vascular endothelial growth factor promotes breast carcinoma invasion in an autocrine manner by regulating the chemokine receptor CXCR4. Cancer Res 2002;62:7203–7206.

    Yonezawa A, Morita R, Takaori-Kondo A et al. Natural alpha interferon-producing cells respond to human immunodeficiency virus type 1 with alpha interferon production and maturation into dendritic cells. J Virol 2003;77:3777–3784.

    Iikura M, Miyamasu M, Yamaguchi M et al. Chemokine receptors in human basophils: inducible expression of functional CXCR4. J Leukoc Biol 2001;70:113–120.

    Jourdan P, Vendrell JP, Huguet MF et al. Cytokines and cell surface molecules independently induce CXCR4 expression on CD4+ CCR7+ human memory T cells. J Immunol 2000;165:716–724.

    Lapidot T, Kollet O. The essential roles of the chemokine SDF-1 and its receptor CXCR4 in human stem cell homing and repopulation of transplanted immune-deficient NOD/SCID and NOD/SCID/B2mnull mice. Leukemia 2002;16:1992–2003.

    Askari AT, Unzek S, Popovic ZB et al. Effect of stromal-cell-derived factor 1 on stem-cell homing and tissue regeneration in ischaemic cardiomyopathy. Lancet 2003;362:697–703.

    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;112:160–169.

    Schrader AJ, Lechner O, Templin M et al. CXCR4/CXCL12 expression and signalling in kidney cancer. Br J Cancer 2002;86:1250–1256.

    Abbott JD, Huang Y, Liu D et al. Stromal cell-derived factor-1alpha plays a critical role in stem cell recruitment to the heart after myocardial infarction but is not sufficient to induce homing in the absence of injury. Circulation 2004;110:3300–3305.

    Takahashi T, Kalka C, Masuda H et al. Ischemia- and cytokine-induced mobilization of bone marrow-derived endothelial progenitor cells for neovascularization. Nat Med 1999;5:434–438.

    Iwaguro H, Yamaguchi J, Kalka C et al. Endothelial progenitor cell vascular endothelial growth factor gene transfer for vascular regeneration. Circulation 2002;105:732–738.

    Ponomaryov T, Peled A, Petit I et al. Induction of the chemokine stromal-derived factor-1 following DNA damage improves human stem cell function. J Clin Invest 2000;106:1331–1339.

    Kucia M, Wysoczynski M, Reca R et al. Tissue committed stem cells (TCSC) are deposited in the bone marrow early during ontogenesis as a mobile pool of stem cells for tissue/organ regeneration – a new insight into phenomenon of stem cell plasticity and aging. Exp Hematol 2004;32:131a.

    Butler JM, Guthrie SM, Koc M et al. SDF-1 is both necessary and sufficient to promote proliferative retinopathy. J Clin Invest 2005;115:86–93.

    Korbling M, Estrov Z. Adult stem cells for tissue repair - a new therapeutic concept? N Engl J Med 2003;349:570–582.

    Isner JM, Kalka C, Kawamoto A et al. Bone marrow as a source of endothelial cells for natural and iatrogenic vascular repair. Ann N Y Acad Sci 2001;953:75–84.

    Ceradini DJ, Kulkarni AR, Callaghan MJ et al. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med 2004;10:858–864.

    Rafii S, Lyden D. Therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration. Nat Med 2003;9:702–712.

    Jin C, Fu WX, Xie LP et al. SDF-1alpha production is negatively regulated by mouse estrogen enhanced transcript in a mouse thymus epithelial cell line. Cell Immunol 2003;223:26–34.

    Wright N, de Lera TL, Garcia-Moruja C et al. Transforming growth factor-beta1 down-regulates expression of chemokine stromal cell-derived factor-1: functional consequences in cell migration and adhesion. Blood 2003;102:1978–1984.

    Kimura T, Boehmler AM, Seitz G et al. The sphingosine 1-phosphate receptor agonist FTY720 supports CXCR4-dependent migration and bone marrow homing of human CD34+ progenitor cells. Blood 2004;103:4478–4486.

    Reca R, Mastellos D, Majka M et al. Functional receptor for C3a anaphylatoxin is expressed by normal hematopoietic stem/progenitor cells, and C3a enhances their homing-related responses to SDF-1. Blood 2003;101:3784–3793.

    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;103:2981–2989.

    Wysoczynski M, Reca R, Ratajczak J et al. Incorporation of CXCR4 into membrane lipid rafts primes homing-related responses of hematopoietic stem/progenitor cells to an SDF-1 gradient. Blood 2005;105:40–48.

    Janowska-Wieczorek A, Majka M, Kijowski J et al. Platelet-derived microparticles bind to hematopoietic stem/progenitor cells and enhance their engraftment. Blood 2001;98:3143–3149.

    Chan KK, Oza AM, Siu LL. The statins as anticancer agents. Clin Cancer Res 2003;9:10–19.

    Schmidmaier R, Baumann P, Simsek M et al. The HMG-CoA reductase inhibitor simvastatin overcomes cell adhesion-mediated drug resistance in multiple myeloma by geranylgeranylation of Rho protein and activation of Rho kinase. Blood 2004;104:1825–1832.

    Kirsch C, Eckert GP, Mueller WE. Statin effects on cholesterol microdomains in brain plasma membranes. Biochem Pharmacol 2003;65:843–856.

    Hecht I, Cahalon L, Hershkoviz R et al. Heterologous desensitization of T cell functions by CCR5 and CXCR4 ligands: inhibition of cellular signaling, adhesion and chemotaxis. Int Immunol 2003;15:29–38.

    Honczarenko M, Le Y, Glodek AM et al. CCR5-binding chemokines modulate CXCL12 (SDF-1)-induced responses of progenitor B cells in human bone marrow through heterologous desensitization of the CXCR4 chemokine receptor. Blood 2002;100:2321–2329.

    Velders GA, van Os R, Hagoort H et al. Reduced stem cell mobilization in mice receiving antibiotic modulation of the intestinal flora: involvement of endotoxins as cofactors in mobilization. Blood 2004;103:340–346.

    Kishore SP, Bungum MK, Platt JL et al. Selective suppression of Toll-like receptor 4 activation by chemokine receptor 4. FEBS Lett 2005;579:699–704.

    Farzan M, Babcock GJ, Vasilieva N et al. The role of post-translational modifications of the CXCR4 amino terminus in stromal-derived factor 1 alpha association and HIV-1 entry. J Biol Chem 2002;277:29484–29489.

    Valenzuela-Fernandez A, Planchenault T, Baleux F et al. Leukocyte elastase negatively regulates stromal cell-derived factor-1 (SDF-1)/CXCR4 binding and functional by amino-terminal processing of SDF-1 and CXCR4. J Biol Chem 2002;277:15677–15689.

    Christopherson KW 2nd, Hangoc G, Broxmeyer HE. Cell surface peptidase CD26/dipeptidylpeptidase IV regulates CXCL12/stromal cell-derived factor-1 alpha-mediated chemotaxis of human cord blood CD34+ progenitor cells. J Immunol 2002;169:7000–7008.

    Christopherson KW 2nd, Hangoc G, Mantel CR et al. Modulation of hematopoietic stem cell homing and engraftment by CD26. Science 2004;305:1000–1003.

    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;95:3289–3296.

    Janowska-Wieczorek A, Marquez LA, Dobrowsky A et al. Differential MMP and TIMP production by human marrow and peripheral blood CD34(+) cells in response to chemokines. Exp Hematol 2000;28:1274–1285.

    Kijowski J, Baj-Krzyworzeka M, Majka M et al. The SDF-1-CXCR4 axis stimulates VEGF secretion and activates integrins but does not affect proliferation and survival in lymphohematopoietic cells. STEM CELLS 2001;19:453–466.

    Kucia M, Jankowski K, Reca R et al. CXCR4-SDF-1 signalling, locomotion, chemotaxis and adhesion. J Mol Histol 2004;3:233–245.

    Hidalgo A, Sanz-Rodriguez F, Rodriguez-Fernandez JL et al. Chemokine stromal cell-derived factor-1alpha modulates VLA-4 integrin-dependent adhesion to fibronectin and VCAM-1 on bone marrow hematopoietic progenitor cells. Exp Hematol 2001;29:345–355.

    Bonavia R, Bajetto A, Barbero S et al. Chemokines and their receptors in the CNS: expression of CXCL12/SDF-1 and CXCR4 and their role in astrocyte proliferation. Toxicol Lett 2003;139:181–189.

    Lataillade JJ, Clay D, Bourin P et al. Stromal cell-derived factor 1 regulates primitive hematopoiesis by suppressing apoptosis and by promoting G0/G1 transition in CD34+ cells: evidence for an autocrine/paracrine mechanism. Blood 2002;99:1117–1129.

    Vila-Coro AJ, Rodriguez-Frade JM, De Ana AM et al. The chemokine SDF-1 triggers CXCR4 receptor dimerization and activates the JAK/STAT pathway. FASEB J 1999;13:1699–1710.

    Ganju RK, Brubaker SA, Meyer J et al. The -chemokine stromal cell-derived factor-1 binds to the transmembrane G-protein-coupled CXCR-4 receptor and activates multiple signal transduction pathways. J Biol Chem 1998;273:23169–23175.

    Zhang XF, Wang JF, Matczak E et al. Janus kinase 2 is involved in stromal cell-derived factor-1 alpha-induced tyrosine phosphorylation of focal adhesion proteins and migration of hematopoietic progenitor cells. Blood 2001;97:3342–3348.

    Cheng ZJ, Zhao J, Sun Y et al. Beta-arrestin differentially regulates the chemokine receptor CXCR4-mediated signaling and receptor internalization, and this implicates multiple interaction sites between beta-arrestin and CXCR4. J Biol Chem 2000;275:2479–2485.

    Ding Z, Issekutz TB, Downey GP et al. L-selectin stimulation enhances functional expression of surface CXCR4 in lymphocytes: implications for cellular activation during adhesion and migration. Blood 2003;101:4245–4252.

    Tilton B, Ho L, Oberlin E et al. Signal transduction by CXC chemokine receptor 4. Stromal cell-derived factor 1 stimulates prolonged protein kinase B and extracellular signal-regulated kinase 2 activation in T lymphocytes. J Exp Med 2000;192:313–324.

    Wang JF, Park IW, Groopman JE. Stromal cell-derived factor-1alpha 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;95:2505–2513.

    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;115:168–176.

    Majka M, Ratajczak J, Kowalska MA et al. Binding of stromal derived factor-1alpha (SDF-1alpha) to CXCR4 chemokine receptor in normal human megakaryoblasts but not in platelets induces phosphorylation of mitogen-activated protein kinase p42/44 (MAPK), ELK-1 transcription factor and serine/threonine kinase AKT. Eur J Haematol 2000;64:164–172.

    Kremer KN, Humphreys TD, Kumar A et al. Distinct Role of ZAP-70 and Src Homology 2 Domain-Containing Leukocyte Protein of 76 kDa in the Prolonged Activation of Extracellular Signal-Regulated Protein Kinase by the Stromal Cell-Derived Factor-1alpha/CXCL12 Chemokine. J Immunol 2003;171:360–367.

    Walmsley MJ, Ooi SK, Reynolds LF et al. Critical roles for Rac1 and Rac2 GTPases in B cell development and signaling. Science 2003;302:459–462.

    Yang FC, Atkinson SJ, Gu Y et al. Rac and Cdc42 GTPases control hematopoietic stem cell shape, adhesion, migration, and mobilization. Proc Natl Acad Sci U S A 2001;98:5614–5618.

    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;64:2534–2543.

    Okabe S, Fukuda S, Kim YJ et al. Stromal cell-derived factor-1/CXCL12 induced chemotaxis of T cells involves activation of the RasGAP-associated docking protein p62Dok-1. Blood 2004;105:1204–1213.

    Fernandis AZ, Cherla RP, Ganju RK. Differential regulation of CXCR4-mediated T-cell chemotaxis and mitogen-activated protein kinase activation by the membrane tyrosine phosphatase, CD45. J Biol Chem 2003;278:9536–9543.

    Chernock RD, Cherla RP, Ganju RK. SHP2 and cbl participate in -chemokine receptor CXCR4-mediated signaling pathways. Blood 2001;97:608–615.

    Roland J, Murphy BJ, Ahr B et al. Role of intracellular domains of CXCR4 in SDF-1-mediated signaling. Blood 2003;101:399–406.

    Hernandez PA, Gorlin RJ, Lukens JN et al. Mutation in the chemokine receptor gene CXCR4 are associated with WHIM syndrome, a combined immunodeficiency disease. Nat Genet 2003;34:70–74.

    Zhang W, Navenot JM, Haribabu B et al. A point mutation that confers constitutive activity to CXCR4 reveals that T140 is an inverse agonist and that AMD3100 and ALX40-4C are weak partial agonists. J Biol Chem 2002;277:24515–24521.

    Haribabu B, Richardson RM, Fisher I et al. Regulation of human chemokine receptors CXCR4. Role of phosphorylation in desensitization and internalization. J Biol Chem 1997;272:28726–28731.

    Ratajczak MZ, Kucia M, Majka M et al. Heterogeneous populations of bone marrow stem cells - are we spotting on the same cells from the different angles? Folia Histochem Cytobiol 2004;42:139–146.

    Kucia M, Ratajczak J, Reca R et al. Tissue-specific muscle, neural and liver stem/progenitor cells reside in the bone marrow, respond to an SDF-1 gradient and are mobilized into peripheral blood during stress and tissue injury. Blood Cells Mol Dis 2004;32:52–57.

    Kucia M, Reca R, Wysoczynski M et al. A potential new application of mobilization/leukapheresis for enrichment of peripheral blood in circulating non-hematopoietic CXCR4+CD45– tissue-committed stem cells (TCSC) for organ/tissue regeneration. Blood 2004;11:151a.

    Shi Q, Rafii S, Wu MH etal. Evidence for circulating bone marrow-derived endothelial cells. Blood 1998;92:362–367.

    Jiang S, Walker L, Afentoulis M et al. Transplanted human bone marrow contributes to vascular endothelium. Proc Natl Acad Sci U S A 2004;101:16891–16896.

    LaBarge MA, Blau HM. Biological progression from adult bone marrow to mononucleate muscle stem cell to multinucleate muscle fiber in response to injury. Cell 2002;111:589–601.

    Corti S, Locatelli F, Donadoni C et al. Wild-type bone marrow cells ameliorate the phenotype of SOD1-G93A ALS mice and contribute to CNS, heart and skeletal muscle tissues. Brain 2004;127:2518–2532.

    Mezey E, Key S, Vogelsang G et al. Transplanted bone marrow generates new neurons in human brains. Proc Natl Acad Sci U S A 2003;100:1364–1369.

    Weimann JM, Charlton CA, Brazelton TR et al. Contribution of transplanted bone marrow cells to Purkinje neurons in human adult brains. Proc Natl Acad Sci U S A 2003;100:2088–2093.

    Locatelli F, Corti S, Donadoni C et al. Neuronal differentiation of murine bone marrow Thy-1- and Sca-1-positive cells. J Hematother Stem Cell Res 2003;12:727–734.

    Theise ND, Nimmakayalu M, Gardner R et al. Liver from bone marrow in humans. Hepatology 2000;32:11–16.

    Oh SH, Muzzonigro TM, Bae SH et al. Adult bone marrow-derived cells trans-differentiating into insulin-producing cells for the treatment of type I diabetes. Lab Invest 2004;84:607–617.

    Kataoka K, Medina RJ, Kageyama T et al. Participation of adult mouse bone marrow cells in reconstitution of skin. Am J Pathol 2003;163:1227–1231.

    Fang B, Shi M, Liao L et al. Multiorgan engraftment and multilineage differentiation by human fetal bone marrow Flk1+/CD31–/CD34– progenitors. J Hematother Stem Cell Res 2003;12:603–613.

    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;30:973–981.

    Kuznetsov SA, Mankani MH, Gronthos S et al. Circulating skeletal stem cells. J Cell Biol 2001;153:1133–1140.

    Hill WD, Hess DC, Martin-Studdard A et al. SDF-1 (CXCL12) is upregulated in the ischemic penumbra following stroke: association with bone marrow cell homing to injury. J Neuropathol Exp Neurol 2004;63:84–96.

    Zhang H, Vutskits L, Pepper MS et al. VEGF is a chemoattractant for FGF-2-stimulated neural progenitors. J Cell Biol 2003;163:1375–1384.

    Israsena N, Hu M, Fu W et al. The presence of FGF2 signaling determines whether beta-catenin exerts effects on proliferation or neuronal differentiation of neural stem cells. Dev Biol 2004;268:220–231.

    Erlandsson A, Larsson J, Forsberg-Nilsson K. Stem cell factor is a chemoattractant and a survival factor for CNS stem cells. Exp Cell Res 2004;301:201–210.

    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;97:3354–3360.

    Zhong R, Law P, Wong D et al. Small peptide analogs to stromal derived factor-1 enhance chemotactic migration of human and mouse hematopoietic cells. Exp Hematol 2004;32:470–475.

    Poznansky MC, Olszak IT, Evans RH et al. Thymocyte emigration is mediated by active movement away from stroma-derived factors. J Clin Invest 2002;109:1101–1110.

    Burger M, Glodek A, Hartmann T et al. Functional expression of CXCR4 (CD184) on small-cell lung cancer cells mediates migration, integrin activation, and adhesion to stromal cells. Oncogene 2003;22:8093–8101.

    Sotsios Y, Whittaker GC, Westwick J et al. The CXC chemokine stromal cell-derived factor activates a Gi-coupled phosphoinositide 3-kinase in T lymphocytes. J Immunol 1999;163:5954–5963.

    Fox JA, Ung K, Tanlimco SG et al. Disruption of a single Pten allele augments the chemotactic response of B lymphocytes to stromal cell-derived factor-1. J Immunol 2002;169:49–54.

    Yamaguchi J, Kusano KF, Masuo O et al. Stromal cell derived factor-1 effects on ex vivo expanded endothelial progenitor cell recruitment for ischemic neovascularization. Circulation 2003;107:1322–1328.

    Majka M, Janowska-Wieczorek A, Ratajczak J et al. Stromal-derived factor1 and thrombopoietin regulate distinct aspects of human megakaryopoiesis. Blood 2000;96:4142–4151.

    Taichman RS, Cooper C, Keller ET et al. Use of the stromal cell-derived factor-1/CXCR4 pathway in prostate cancer metastasis to bone. Cancer Res 2002;62:1832–1837.

    Avecilla ST, Hattori K, Heissig B et al. Chemokine-mediated interaction of hematopoietic progenitors with the bone marrow vascular niche is required for thrombopoiesis. Nat Med 2004;10:64–71.

    Fuchs E, Tumbar T, Guasch G. Socializing with the neighbors: stem cells and their niche. Cell 2004;116:769–778.

    Brusselmans K, Bono F, Collen D et al. A Novel Role for Vascular Endothelial Growth Factor as an Autocrine Survival Factor for Embryonic Stem Cells during Hypoxia. J Biol Chem 2005;280:3493–3499.

    Neuhaus T, Stier S, Totzke G et al. Stromal cell-derived factor 1 alpha (SDF-1alpha) induces gene-expression of early growth response-1 (Egr-1) and VEGF in human arterial endothelial cells and enhances VEGF induced cell proliferation. Cell Prolif 2003;36:75–86.

    Asahara T, Masuda H, Takahashi T et al. Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circ Res 1999;85:221–228.

    Kryczek I, Lange A, Mottram P et al. CXCL12 and vascular endothelial growth factor synergistically induce neoangiogenesis in human ovarian cancers. Cancer Res 2005;65:465–472.

    Spiegel A, Kollet O, Peled A et al. Unique SDF-1-induced activation of human precursor-B ALL cells as a result of altered CXCR4 expression and signaling. Blood 2004;103:2900–2907.

    Samara GJ, Lawrence DM, Chiarelli CJ et al. CXCR4-mediated adhesion and MMP-9 secretion in head and neck squamous cell carcinoma. Cancer Lett 2004;214:231–241.

    Sanchez X, Cousins-Hodges B, Aguilar T et al. Activation of HIV-1 coreceptor (CXCR4) mediates myelosuppression. J Biol Chem 1997;272:27529–27531.

    Vlahakis SR, Villasis-Keever A, Gomez T et al. G protein-coupled chemokine receptors induced both survival and apoptotic signaling pathways. J Immunol 2002;169:5546–5554.

    Su S, Mukaida N, Wang J et al. Inhibition of immature erythroid progenitor cell proliferation by macrophage inflammatory protein-1a by interacting mainly with a C-C chemokine receptor, CCR1. Blood 1997;90:605–611.

    Majka M, Janowska-Wieczorek A, Ratajczak J et al. Numerous growth factors, cytokines, and chemokines are secreted by human CD34(+) cells, myeloblasts, erythroblasts, and megakaryoblasts and regulate normal hematopoiesis in an autocrine/paracrine manner. Blood 2001;97:3075–3085.

    Broxmeyer HE, Kohli L, Kim CH et al. Stromal cell-derived factor-1/CXCL12 directly enhances survival/antiapoptosis of myeloid progenitor cell through CXCR4 and G proteins and enhances engraftment of competitive, repopulating stem cells. J Leukoc Biol 2003;73:630–638.

    Zhou Y, Larsen PH, Hao C et al. CXCR4 is a major chemokine receptor on glioma cells and mediates their survival. J Biol Chem 2002;277:49481–49487.

    Barbero S, Bonavia R, Bajetto A et al. Stromal cell-derived factor 1 cell growth through the activation of both extracellular signal-regulated kinase 1/2 and Akt. Cancer Res 2003;63:1969–1974.

    Frisch SM, Screaton RA. Anoikis mechanisms. Curr Opin Cell Biol 2001;13:555–562.

    Ratajczak MZ, Reca R, Wysoczynski M et al. Transplantation studies in C3-deficient animals reveal a novel role of the third complement component (C3) in engraftment of bone marrow cells. Leukemia 2004;18:1482–1490.

    Greene WC. The brightening future of HIV therapeutics. Nat Immunol 2004;5:867–871.

    Chen Y, Stamatoyannopoulos G, Song CZ. Down-regulation of CXCR4 by inducible small interfering RNA inhibits breast cancer cell invasion in vitro. Cancer Res 2003;63:4801–4804.

    Mazure NM, Brahimi-Horn MC, Berta MA et al. HIF-1: master and commander of the hypoxic world. A pharmacological approach to its regulation by siRNAs. Biochem Pharmacol 2004;68:971–980.

    Kung AL, Zabludoff SD, France DS et al. Small molecule blockade of transcriptional coactivation of the hypoxia-inducible factor pathway. Cancer Cell 2004;6:33–43.

    Solchaga LA, Penick K, Porter JD et al. FGF-2 enhances the mitotic and chondrogenic potentials of human adult bone marrow-derived mesenchymal stem cells. J Cell Physiol 2005; 203:398–409.

    Pitman M, Emery B, Binder M et al. LIF receptor signaling modulates neural stem cell renewal. Mol Cell Neurosci 2004;27:255–266.(Magda Kuciaa, Ryan Recaa,)