当前位置: 首页 > 期刊 > 《干细胞学杂志》 > 2004年第3期 > 正文
编号:11342417
Interactions of Chemokines and Chemokine Receptors Mediate the Migration of Mesenchymal Stem Cells to the Impaired Site in the Brain After H
http://www.100md.com 《干细胞学杂志》
     Department of Anatomy, Faculty of Medicine, National University of Singapore, Singapore

    Key Words. Mesenchymal stem cells ? Hypoglossal nerve ? Chemokines ? Chemokine receptors ? Transplantation ? Lateral ventricle

    Samuel Sam Wah Tay, Ph.D., Department of Anatomy, Faculty of Medicine, National University of Singapore, Lower Kent Ridge Road, Singapore 117597. Telephone: 65-68743210; Fax: 65-67787643; e-mail: anttaysw@nus.edu.sg

    ABSTRACT

    Bone marrow contains precursors for hematopoietic cells and stem-like cells for a variety of nonhematopoietic tissues. The precursors of nonhematopoietic tissues were initially referred to as plastic-adherent cells or colony-forming units fibroblasts because of their ability to adhere to culture dishes and form fibroblast-like colonies . In addition, these cells were referred to as mesenchymal stem cells (MSCs) or mesenchymal progenitor cells because they possess the capacity to differentiate into a variety of nonhematopoietic cells . Moreover, they have been referred to as marrow stromal cells because they arise from the supporting structures in bone marrow and can act as feeder layer for the growth of hematopoietic stem cells in culture .

    MSCs have been shown to have multipotential capacities to differentiate into osteoblasts, adipocytes, chondrocytes, myoblasts, and myotubes . Recent data have pointed to the unexpected ability of both human and rodent MSCs to differentiate into neural cells . The evidence has suggested their therapeutic potential to regenerate neural cells in the injured or diseased brain .

    Interestingly, recent studies from Chopp’s group have demonstrated the capability of in vitro propagated rat MSCs (rMSCs) to migrate selectively into damaged areas in the brain after the rMSCs were systemically or locally implanted. In the models of traumatic or ischemic brain injuries, rMSCs administered intravenously, intra-arterially, or intracerebrally preferentially migrated into the region of neurodegeneration in the brain . Identification of the molecular signals governing rMSC migration in vivo is of major importance for understanding the MSC-mediated cell therapy for traumas and diseases in the central nervous system (CNS). Although an initial study suggested a role for chemokines in both rMSC and human MSC migration in vitro , the signaling pathway relevant to their directed migration still remains unknown. In this study, we attempted to investigate the hypothesis that the interaction between chemokines and their receptors could play important roles in the migration of rMSCs to the impaired sites in the CNS in the model of left hypoglossal nerve avulsion. Unilateral hypoglossal nerve avulsion has been proven to cause severe motoneuronal death in the injured nucleus but no cell death in the contralateral side . Therefore, it would be a simple model to provide a practical comparison between the avulsed and intact nuclei to study the migration of transplanted rMSCs. Our results suggest that the interaction between fractalkine and its receptor CX3CR1 and the interaction of stromal cell-derived factor 1 (SDF-1) and its receptor CXCR4 could partially mediate the trafficking of rMSCs to the impaired nucleus in the brain.

    MATERIALS AND METHODS

    In Vitro Characterization of rMSCs

    MSCs were isolated from the femurs and tibiae of adult rats and propagated in vitro. As reported in a previous study , rMSCs grew as colonies (Fig. 1A). For transplantation studies, the rMSCs labeled by CFDA-SE displayed green fluorescence (Fig. 1B). There was no evidence of hematopoietic precursors in the cultures as fluorescent cell sorting and immunohistochemistry analysis (data not shown) demonstrated that the cells were negative for CD11b and CD45, cell-surface markers associated with lymphohematopoietic cells.

    Figure 1. Characterization of rMSCs. A) Phase-contrast microphotography of rMSCs at passage 0. B) rMSCs displayed green fluorescence after being labeled with CFDA-SE before transplantation. C) Differentiation of rMSCs into osteoblasts in vitro; rMSCs incubated in osteogenic medium for 21 days. The mineral (arrows) in the cultures was detected by staining with Alizarin red. D) Differentiation of rMSCs into adipocytes in vitro; rMSCs incubated in adipogenic medium for 21 days showed fat droplets (arrows) in the cells stained with Oil Red O. Scale bar = 100 μm.

    To assess the multipotentiality of rMSCs in the culture, the cells were subjected to in vitro differentiation assays. Under the influence of osteogenic medium, the isolated rMSCs formed aggregation or nodules and showed increased calcium accumulation as revealed by Alizarin red staining (Fig. 1C). In addition, rMSCs differentiated into adipocytes that were characterized by round morphology and accumulation of large cytoplasmic vacuoles containing lipid as shown by Oil Red O staining (Fig. 1D). Collectively, these results demonstrate that rMSCs in the cultures are morphologically and functionally characteristic of multipotential mesenchymal progenitors.

    Expression of Chemokine Receptors CCR2, CCR5, CXCR4, and CX3CR1 in rMSCs

    We examined the expression of chemokine receptors CCR2, CCR5, CXCR4, and CX3CR1 in rMSCs by RT-PCR, flow cytometry, and immunohistochemistry. The purity of microglial cultures was found to be around 95% (data not shown). We detected mRNA expression of CXCR4, CCR2, CCR5, and CX3CR1 in both microglial cells (positive control) (Fig. 2Aa-d, lane 2) and rMSCs (Fig. 2Aa-d, lane 3). The protein expression of CCR2, CCR5, CXCR4, and CX3CR1 on the surface of rMSCs was detected by flow cytometry (Fig. 2Ba-d). Immunocytochemical analysis revealed the localization of CCR2, CCR5, CXCR4, and CX3CR1 expression on the membranes and in the cytoplasm (Fig. 2Ca-d).

    Figure 2. Expression of CCR2, CCR5, CXCR4, and CX3CR1 by rMSCs at mRNA and protein levels. A) RT-PCR was used to determine mRNA expression of chemokine receptors by rMSCs. The PCR bands for CCR2, CCR5, CXCR4, and CX3CR1 were present in both microglial positive control (a-d, lane 2) and rMSCs (a-d, lane 3), whereas they were absent in negative controls (a-d, lane 4) where PCR was performed on RNA template without reverse transcription step. B) Expression of CCR2, CCR5, CXCR4, and CX3CR1 at the protein level in rMSCs; rMSCs were stained with anti-CCR2 (a), CCR5 (b), CXCR4 (c), or CX3CR1 (d) antibodies and then analyzed by flow cytometry. The shaded regions represent isotype control staining and the open regions represent the chemokine receptors staining. C) Localization of CCR2, CCR5, CXCR4, and CX3CR1 in rMSCs. Immunohistochemistry revealed the localization of CCR2 (a), CCR5 (b), CXCR4 (c), and CX3CR1 (d) in the membrane and cytoplasm of rMSCs. Scale bar = 100 μm.

    Migration of rMSCs in Response to rrfractalkine and rhSDF-1 in Heterotrimeric G-Protein-Dependent Manner In Vitro

    In view of the observations that rMSCs express CXCR4 (the sole receptor for SDF-1) and CX3CR1 (the only receptor for fractalkine), we used rhSDF-1 and the extracellular domain of rrfractalkine in an in vitro chemotaxis assay. The rrfractalkine (5 ng/ml and 50 ng/ml) and rhSDF-1 (5–500 ng/ml) significantly (p < 0.01) induced the migration of rMSCs (Figs. 3 and 4A). Interestingly, at higher concentrations of rrfractalkine (250 ng/ml and 500 ng/ml) there was no significant number of migrated rMSCs compared with the control (Fig. 3A). However, rhSDF-1 induced the migration of rMSCs in a dose-dependent manner; the optimal migration of rMSCs was observed at 250 ng/ml of rhSDF-1 (Fig. 4A). The number of migrated rMSCs to rrfractalkine and rhSDF-1 was decreased when the same effective concentrations of rrfractalkine and rhSDF-1 were added into both the top and bottom chambers in the assays (Figs. 3 and 4B). Moreover, the chemotactic effects of rrfractalkine or rhSDF-1 on rMSCs were abolished by the addition of antifractalkine or anti-SDF-1 respectively in the bottom chamber (Figs. 3 and 4C), suggesting the specificity of their chemotactic effects.

    Figure 3. Effect of rrfractalkine on migration of rMSCs. A) A significant increase in the number of migrated rMSCs was found at the concentrations of 5 and 50 ng/ml rrfractalkine (p < 0.01) compared with control. After treatment with PTx, the number of migrated rMSCs decreased to the control level at the concentrations of 5 and 50 ng/ml rrfractalkine. B) Chemotactic effect of rrfractalkine on migration of rMSCs. A significant increase in the number of migrated rMSCs was observed at the concentration of 5 ng/ml rrfractalkine. After 5 ng/ml rrfractalkine was added to both top and bottom chambers, the number of migrated rMSCs was significantly decreased. C) Specificity of the effect of rrfractalkine on migration of rMSCs. After blockage of rrfractalkine by 480 μg/ml antifractalkine antibody, the chemotactic effect of rrfractalkine on rMSCs was abolished. **p < 0.01. The results are the mean ± SE of three independent experiments.

    Figure 4. Effect of rhSDF-1 on migration of rMSCs. A) The number of migrated rMSCs increased dose-dependently at the concentration of 5–500 ng/ml (p < 0.01), compared with control. Maximum effect of rhSDF-1 was observed at the concentration of 250 ng/ml. Pretreatment of rMSCs with PTx abrogated the effect of rhSDF-1 at various concentrations. a) The migrated rMSCs in the control condition. b) The migrated rMSCs at the concentration of 250 ng/ml rhSDF-1. c) The migrated rMSCs at the concentration of 250 ng/ml rhSDF-1 after pretreatment of PTx. B) Chemotactic effect of rhSDF-1 on rMSCs. When 250 ng/ml rhSDF-1 was added to both top and bottom chambers, the number of migrated rMSCs was significantly decreased. a) The migrated rMSCs in the control condition. b) The migrated rMSCs at the concentration of 250 ng/ml rhSDF-1 in the bottom chamber only. c) The migrated rMSCs at the concentration of 250 ng/ml rhSDF-1 added to both top and bottom chambers. C) Specificity of the effect of rhSDF-1 on migration of rMSCs. When rhSDF-1 was blocked by 480 μg/ml anti-SDF-1 antibody, the chemotactic effect of rhSDF-1 on rMSCs was abolished. a) The migrated rMSCs in the control condition. b) The migrated rMSCs at the concentration of 250 ng/ml rhSDF-1. c) The migrated rMSCs at the concentration of 250 ng/ml rhSDF-1 blocked by anti-SDF-1 antibody. **p < 0.01. Scale bar = 100 μm. The results are the mean ± SE of three independent experiments.

    In order to understand whether the rrfractalkine- and rhSDF-1-induced migration of rMSCs was transmitted in a G-protein-dependent manner, the chemotaxis assay with the rrfractalkine and rhSDF-1 was performed in the presence of PTx, a specific inhibitor of heterotrimeric G-protein coupling to G-protein-coupled receptors, potently inhibiting signaling through Gai proteins . Upon pre-exposure of rMSCs to PTx, almost complete inhibition of rrfractalkine- and rhSDF-1-induced migratory response was observed (Figs. 3 and 4A). However, PTx was not observed to affect the viability of rMSCs (data not shown). These results indicate that the migratory response of rMSCs induced by rrfractalkine and rhSDF-1 is mediated by heterotrimeric G proteins, possibly Gai, suggesting that the signaling events leading to migration of rMSCs are transmitted through the CX3CR1 and CXCR4 receptors, both of which are G-protein-coupled receptors.

    Migration of rMSCs Following the Intracerebral Injection of rhSDF-1 In Vivo

    Based on the observation that rhSDF-1 has a potent migratory effect on rMSCs in vitro, we investigated whether rhSDF-1 injected intracerebrally could promote the migration of tranaplanted rMSCs in vivo. As shown by flow cytometry analysis, administration of rhSDF-1 (0.1 μg) significantly (p < 0.01) induced the accumulation of prelabeled rMSCs in the cerebral cortex area surrounding the injection site compared with the effect of PBS alone (Fig. 5).

    Figure 5. Effect of rhSDF-1 on migration of rMSCs in vivo. Immediately following jugular-vein injection of CFDA-SE–labeled rMSCs (3 x 106 cells/rat), the Wistar rat received an intracerebral injection of rhSDF-1 (0.1 μg in 10 μl) into the right cerebral cortex. As control, the opposite side was injected with PBS alone. Approximately 1 cm3 of cortical tissue block around the injected site was excised 6 hours later and proteolytically digested to produce a single-cell suspension. The number of labeled rMSCs per biopsy sample was estimated by flow cytometry and expressed as a percentage of the total number of fluorescent rMSCs injected into the jugular vein. The percentage of migrated rMSCs in the rhSDF-1-injected sites was significantly higher (p < 0.01) compared with the PBS-injected site. **p < 0.01. The results are the mean ± SE of three independent experiments.

    Selective Migration of Transplanted rMSCs Into Injured Left HN

    To further prove that transplanted rMSCs possess the directed migratory capacity to target impaired sites in the brain, we investigated their trafficking in an animal model of left hypoglossal nerve avulsion. Before transplantation, the cells were prelabeled with CFDA-SE. Examination of frozen sections by fluorescent microscopy revealed that at 1 and 2 weeks after injection of CFDA-SE–labeled MSCs into both lateral ventricles of the rat brain with left hypoglossal nerve avulsion, significantly more (p < 0.01) labeled cells were detected in the injured HN (Fig. 6C, 6E), in comparison with that of the right control side (Fig. 6D, 6F) and sham-operated control rats (Fig. 6A, 6B). Moreover, at 2 weeks after transplantation, the number of rMSCs in the avulsed HN was significantly (p < 0.01) decreased compared with that at 1 week postoperation (Fig. 6G). This observation suggests that transplanted MSCs may have the capacity for preferential migration to the site of neuronal injury.

    Figure 6. Migration of rMSCs in the model of left hypoglossal nerve avulsion. Left hypoglossal nerves of rats were avulsed and 5 μl of semisuspended prelabeled rMSCs (2 x 105 cells/μl) were then transplanted into the lateral ventricles of the avulsed and sham-operated rats. A few migrated rMSCs (arrows) were observed in the left avulsed HN (C, E), but not in the right HN (D, F) and controls (A, B), at 1 week (C, D) and 2 weeks (E, F) after operation and transplantation of labeled rMSCs into both lateral ventricles. Note: there were no labeled rMSCs in adjacent left dorsal motor nucleus of the vagus (DMV) (E). There were significantly more rMSCs in the left HN (p < 0.01) compared with right HN and control rats at 1 week after transplantation and operation (G). Two weeks later, the number of rMSCs was significantly decreased (p < 0.01) (G). **p < 0.01. Scale bar = 100 μm. The results are the mean ± SE of three independent experiments.

    Upregulated Expression of Fractalkine and SDF-1 in the Injured HN

    We examined the expression of fractalkine and SDF-1 in the HN at 1 and 2 weeks after left hypoglossal nerve avulsion. Immunohistochemical staining showed that, compared with the weak expression of fractalkine and SDF-1 in the HN of control rats (Fig. 7Aa,b; 7Ba,b) and in the right HN of avulsed rats (Fig. 7Ad, 7Bd), immunoreactivities were dramatically upregulated in the left HN at 1 week after the nerve injury (Fig. 7Ac, 7Bc). The expression of fractalkine and SDF-1 remained enhanced at 2 weeks after operation (Fig. 7Ae, 7Be). Quantification analysis of the immunopositive cells showed that the numbers of fractalkine and SDF-1 immunopositive cells in the left avulsed HN were significantly (p < 0.01) increased at 1 week after operation compared with that in the right HN and controls (Fig. 7Ag, 7Bg). Subsequently, at 2 weeks after operation, the numbers of MCP-1, fractalkine, and SDF-1 positive cells were significantly (p < 0.01) decreased (Fig. 7Ag, 7Bg).

    Figure 7. Expression of fractalkine and SDF-1 in the HN. A) Fractalkine immunostaining was upregulated in the left HN at 1 (c) and 2 (e) weeks after operation compared with the controls (a, b, d, f). Quantification analysis revealed that the number of fractalkine immuopositive cells significantly (p < 0.01) increased in the left HN at 1 week after operation compared with the controls (g). Subsequently, at 2 weeks after operation, the number significantly decreased (p < 0.01) (g). B) Expression of SDF-1 in the HN. SDF-1 immunoreactivity was enhanced in left HN at 1 week (c) and 2 weeks (e) after operation, compared with the controls (a, b, d, f). Quantification analysis revealed that the number of SDF-1 immunoreactive cells significantly (p < 0.01) increased in the left HN at 1 week after operation compared with the controls (g). Subsequently, at 2 weeks after operation, the number significantly decreased (p < 0.01) (g). **p < 0.01. Scale bar = 100 μm. The results are the mean ± SE.

    DISCUSSION

    This work was supported by a research grant (R181-000-059-213) from the National University of Singapore.

    REFERENCES

    Piersma AH, Brockbank KG, Ploemacher RE et al. Characterization of fibroblastic stromal cells from murine bone marrow. Exp Hematol 1985;13:237–243.

    Caplan AI. Mesenchymal stem cells. J Orthop Res 1991;9:641–650.

    Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science 1997;276:71–74.

    Clark BR, Keating A. Biology of bone marrow stroma. Ann N Y Acad Sci 1995;770:70–78.

    Pereira RF, O’Hara MD, Laptev AV et al. Marrow stromal cells as a source of progenitor cells for nonhematopoietic tissues in transgenic mice with a phenotype of osteogenesis imperfecta. Proc Natl Acad Sci USA 1998;95:1142–1147.

    Wakitani S, Saito T, Caplan AI. Myogenic cells derived from rat bone marrow mesenchymal stem cells exposed to 5-azacytidine. Muscle Nerve 1995;18:1417–1426.

    Azizi SA, Stokes D, Augelli BJ et al. Engraftment and migration of human bone marrow stromal cells implanted in the brains of albino rats—similarities to astrocyte grafts. Proc Natl Acad Sci USA 1998;95:3908–3913.

    Sanchez-Ramos J, Song S, Cardozo-Pelaez F et al. Adult bone marrow stromal cells differentiate into neural cells in vitro. Exp Neurol 2000;164:247–256.

    Woodbury D, Schwarz EJ, Prockop DJ et al. Adult rat and human bone marrow stromal cells differentiate into neurons. J Neurosci Res 2000;61:364–370.

    Prockop DJ, Azizi SA, Phinney DG et al. Potential use of marrow stromal cells as therapeutic vectors for diseases of the central nervous system. Prog Brain Res 2000;128:293–297.

    Lu D, Li Y, Wang L et al. Intraarterial administration of marrow stromal cells in a rat model of traumatic brain injury. J Neurotrauma 2001;18:813–819.

    Lu D, Mahmood A, Wang L et al. Adult bone marrow stromal cells administered intravenously to rats after traumatic brain injury migrate into brain and improve neurological outcome. Neuroreport 2001;12:559–563.

    Mahmood A, Lu D, Wang L et al. Treatment of traumatic brain injury in female rats with intravenous administration of bone marrow stromal cells. Neurosurgery 2001;49:1196–1203.

    Chen J, Li Y, Wang L et al. Therapeutic benefit of intracerebral transplantation of bone marrow stromal cells after cerebral ischemia in rats. J Neurol Sci 2001;189:49–57.

    Chen J, Li Y, Wang L et al. Therapeutic benefit of intravenous administration of bone marrow stromal cells after cerebral ischemia in rats. Stroke 2001;32:1005–1011.

    Wang L, Li Y, Chen X et al. MCP-1, MIP-1, IL-8 and ischemic cerebral tissue enhance human bone marrow stromal cell migration in interface culture. Hematology 2002;7:113–117.

    Wang L, Li Y, Chen J et al. Ischemic cerebral tissue and MCP-1 enhance rat bone marrow stromal cell migration in interface culture. Exp Hematol 2002;30:831–836.

    Jiang Y, Zhang M, Koishi K et al. TGF-beta 2 attenuates the injury-induced death of mature motoneurons. J Neurosci Res 2000;62:809–813.

    Javazon EH, Colter DC, Schwarz EJ et al. Rat marrow stromal cells are more sensitive to plating density and expand more rapidly from single-cell-derived colonies than human marrow stromal cells. STEM CELLS 2001;19:219–225.

    Digirolamo CM, Stokes D, Colter D et al. Propagation and senescence of human marrow stromal cells in culture: a simple colony-forming assay identifies samples with the greatest potential to propagate and differentiate. Br J Haematol 1999;107:275–281.

    Boddeke EW, Meigel I, Frentzel S et al. Cultured rat microglia express functional beta-chemokine receptors. J Neuroimmunol 1999;98:176–184.

    Boddeke EW, Meigel I, Frentzel S et al. Functional expression of the fractalkine (CX3C) receptor and its regulation by lipopolysaccharide in rat microglia. Eur J Pharmacol 1999;374:309–313.

    Ohtani Y, Minami M, Kawaguchi N et al. Expression of stromal cell-derived factor-1 and CXCR4 chemokine receptor mRNAs in cultured rat glial and neuronal cells. Neurosci Lett 1998;249:163–166.

    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.

    Bajetto A, Bonavia R, Barbero S et al. Expression of chemokine receptors in the rat brain. Ann N Y Acad Sci 1999;876:201–209.

    Nishiyori A, Minami M, Ohtani Y et al. Localization of fractalkine and CX3CR1 mRNAs in rat brain: does fractalkine play a role in signaling from neuron to microglia? FEBS Lett 1998;429:167–172.

    Garcia-Zepeda EA, Rothenberg ME, Ownbey RT et al. Human eotaxin is a specific chemoattractant for eosinophil cells and provides a new mechanism to explain tissue eosinophilia. Nat Med 1996;2:449–456.

    Korting HC, Schindler S, Hartinger A et al. MTT-assay and neutral red release (NRR)-assay: relative role in the prediction of the irritancy potential of surfactants. Life Sci 1994;55:533–540.

    Fields TA, Casey PJ. Signalling functions and biochemical properties of pertussis toxin-resistant G-proteins. Biochem J 1997;321:561–571.

    Baggiolini M. Chemokines and leukocyte traffic. Nature 1998;392:565–568.

    Melchers F, Rolink AG, Schaniel C. The role of chemokines in regulating cell migration during humoral immune responses. Cell 1999;99:351–354.

    Murdoch C, Finn A. Chemokine receptors and their role in inflammation and infectious diseases. Blood 2000;95:3032–3043.

    Bazan JF, Bacon KB, Hardiman G et al. A new class of membrane-bound chemokine with a CX3C motif. Nature 1997;385:640–644.

    Kelner GS, Kennedy J, Bacon KB et al. Lymphotactin: a cytokine that represents a new class of chemokine. Science 1994;266:1395–1399.

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

    Luster AD. Chemokines—chemotactic cytokines that mediate inflammation. N Engl J Med 1998;338:436–445.

    Wu D, LaRosa GJ, Simon MI. G protein-coupled signal transduction pathways for interleukin-8. Science 1993;261:101–103.

    al Aoukaty A, Schall TJ, Maghazachi AA. Differential coupling of CC chemokine receptors to multiple heterotrimeric G proteins in human interleukin-2-activated natural killer cells. Blood 1996;87:4255–4260.

    Arai H, Charo IF. Differential regulation of G-protein-mediated signaling by chemokine receptors. J Biol Chem 1996;271:21814–21819.

    Hesselgesser J, Liang M, Hoxie J et al. Identification and characterization of the CXCR4 chemokine receptor in human T cell lines: ligand binding, biological activity, and HIV-1 infectivity. J Immunol 1998;160:877–883.

    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;95:9448–9453.

    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.

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

    Hesselgesser J, Halks-Miller M, DelVecchio V et al. CD4-independent association between HIV-1 gp120 and CXCR4: functional chemokine receptors are expressed in human neurons. Curr Biol 1997;7:112–121.

    Feil C, Augustin HG. Endothelial cells differentially express functional CXC-chemokine receptor-4 (CXCR-4/fusin) under the control of autocrine activity and exogenous cytokines. Biochem Biophys Res Commun 1998;247:38–45.

    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.

    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.

    Harrison JK, Jiang Y, Chen S et al. Role for neuronally derived fractalkine in mediating interactions between neurons and CX3CR1-expressing microglia. Proc Natl Acad Sci USA 1998;95:10896–10901.

    Maciejewski-Lenoir D, Chen SZ, Feng LL et al. Characterization of fractalkine in rat brain cells: migratory and activation signals for CX3CR-1-expressing microglia. J Immunol 1999;163:1628–1635.

    Pan Y, Lloyd C, Zhou H et al. Neurotactin, a membrane-anchored chemokine upregulated in brain inflammation. Nature 1997;387:611–617.

    Imai T, Hieshima K, Haskell C et al. Identification and molecular characterization of fractalkine receptor CX3CR1, which mediates both leukocyte migration and adhesion. Cell 1997;91:521–530.

    Sanchez-Ramos JR. Neural cells derived from adult bone marrow and umbilical cord blood. J Neurosci Res 2002;69:880–893.

    Kopen GC, Prockop DJ, Phinney DG et al. Marrow stromal cells migrate throughout forebrain and cerebellum, and they differentiate into astrocytes after injection into neonatal mouse brains. Proc Natl Acad Sci USA 1999;96:10711–10716.

    Ying QL, Nichols J, Evans EP et al. Changing potency by spontaneous fusion. Nature 2002;416:545–548.

    Terada N, Hamazaki T, Oka M et al. Bone marrow cells adopt the phenotype of other cells by spontaneous cell fusion. Nature 2002;416:542–545.

    Chen X, Li Y, Wang L et al. Ischemic rat brain extracts induce human marrow stromal cell growth factor production. Neuropathology 2002;22:275–279.

    Chen X, Katakowski M, Li Y et al. Human bone marrow stromal cell cultures conditioned by traumatic brain tissue extracts: growth factor production. J Neurosci Res 2002;69:687–691.

    Li Y, Chen J, Chen XG et al. Human marrow stromal cell therapy for stroke in rat: neurotrophins and functional recovery. Neurology 2002;59:514–523.(Jun Feng Ji, Bei Ping He,)