An alternatively spliced variant of CXCR3 mediates the inhibition of endothelial cell growth induced by IP-10, Mig, and I-TAC, and acts as functional receptor for platelet factor 4

Laura Lasagni, Michela Francalanci, Francesco Annunziato, Elena Lazzeri, Stefano Giannini, Lorenzo Cosmi, Costanza Sagrinati, Benedetta Mazzinghi, Claudio Orlando, Enrico Maggi, Fabio Marra, Sergio Romagnani, Mario Serio, Paola Romagnani, Laura Lasagni, Michela Francalanci, Francesco Annunziato, Elena Lazzeri, Stefano Giannini, Lorenzo Cosmi, Costanza Sagrinati, Benedetta Mazzinghi, Claudio Orlando, Enrico Maggi, Fabio Marra, Sergio Romagnani, Mario Serio, Paola Romagnani

Abstract

The chemokines CXCL9/Mig, CXCL10/IP-10, and CXCL11/I-TAC regulate lymphocyte chemotaxis, mediate vascular pericyte proliferation, and act as angiostatic agents, thus inhibiting tumor growth. These multiple activities are apparently mediated by a unique G protein-coupled receptor, termed CXCR3. The chemokine CXCL4/PF4 shares several activities with CXCL9, CXCL10, and CXCL11, including a powerful angiostatic effect, but its specific receptor is still unknown. Here, we describe a distinct, previously unrecognized receptor named CXCR3-B, derived from an alternative splicing of the CXCR3 gene that mediates the angiostatic activity of CXCR3 ligands and also acts as functional receptor for CXCL4. Human microvascular endothelial cell line-1 (HMEC-1), transfected with either the known CXCR3 (renamed CXCR3-A) or CXCR3-B, bound CXCL9, CXCL10, and CXCL11, whereas CXCL4 showed high affinity only for CXCR3-B. Overexpression of CXCR3-A induced an increase of survival, whereas overexpression of CXCR3-B dramatically reduced DNA synthesis and up-regulated apoptotic HMEC-1 death through activation of distinct signal transduction pathways. Remarkably, primary cultures of human microvascular endothelial cells, whose growth is inhibited by CXCL9, CXCL10, CXCL11, and CXCL4, expressed CXCR3-B, but not CXCR3-A. Finally, monoclonal antibodies raised to selectively recognize CXCR3-B reacted with endothelial cells from neoplastic tissues, providing evidence that CXCR3-B is also expressed in vivo and may account for the angiostatic effects of CXC chemokines.

Figures

Figure 1.
Figure 1.
Identification, molecular characterization, and tissue distribution of CXCR3-B. (a) Structure of the human CXCR3 gene. Upper case, the exons; lower case, the intron; arrow, the CXCR3-B transcription start; underline, the CXCR3-B translation start site (ATG); light shading, the donor (AG/gt, common for both CXCR3-A and CXCR3-B) and acceptor (ag/GG, CXCR3-B; AG/GT, CXCR3-A) splice sites. The nested primers used for 5′ and 3′ RACE are boxed and the direction is pointed by the arrow. The 182-bp probe (recognizing CXCR3-B) and the 401-bp probe (recognizing both CXCR3-A and CXCR3-B) used for Northern blot analysis are evidenced in yellow and light green, respectively. (b) RACE performed on primary cultures of HMVEC. Lane 1, DNA ladder Marker VIII; lane 2, 5′ RACE product; lane 3, 3′ RACE product; lane 4, DNA ladder 1 Kb plus. (c) Schematic representation illustrating the generation of CXCR3-A and CXCR3-B mRNA through alternative splicing. (d) Multiple tissue Northern blot hybridization with the 182-bp probe (top) or the 401-bp probe (bottom). These sequence data are available from GenBank/EMBL/DDBJ under accession no. AF469635.
Figure 2.
Figure 2.
CXCR3-A and CXCR3-B expression by HMEC-1 transfectants and their binding affinity to cognate ligands. (a) Flow cytometric analysis of surface CXCR3-A. (b) Flow cytometric analysis of surface CXCR3-B. (c) CXCL10, CXCL9, and CXCL11 displacement of 125I-labeled CXCL10 bound to CXCR3-A transfectants. (d) CXCL10, CXCL9, and CXCL11 displacement of 125I-labeled CXCL10 bound to CXCR3-B transfectants. (e) Lack of significant displacement by cold CXCL4 of 125I-labeled CXCL10 bound to CXCR3-A transfectants. (f) CXCL4 displacement of 125I-labeled CXCL10 bound to CXCR3-B transfectants. (g) Lack of significant binding of CXCL4 to CXCR3-A transfectants. (h) High affinity binding of CXCL4 to CXCR3-B transfectants. Figures show mean displacement curves generated by analysis with the Allfit software obtained in eight separate experiments.
Figure 3.
Figure 3.
Functional properties of CXCR3-A- and CXCR3-B-transfected cells. (a) Apoptosis in CXCR3-B transfectants as assessed by detection with flow cytometry of the percentage of cells with a sub-G1 DNA content or of cells showing DNA fragmentation by using the TUNEL technique (inset). Results are from one representative of four experiments. (b) In vitro vessel formation capability of CXCR3-B transfectants as assessed by the Matrigel assay. Data are from one representative of four independent experiments. (c) Apoptosis in CXCR3-A–transfected cells (for details see Fig. 4 a). (d) In vitro vessel formation capability of CXCR3-A transfectants (for details see Fig. 4 b). (e) Detection of CXCL11 mRNA expression by HMEC-1 as assessed by real-time quantitative RT-PCR. Columns represent mean values (±SD) of four separate experiments. (f) Flow cytometric analysis on HMEC-1 of CXCL9, CXCL10, and CXCL11 protein. Black line, isotype control mAb; green line, CXCL10; light blue, CXCL9; pink line, CXCL11. Data are from one representative of five independent experiments. (g) Opposite effects of neutralizing anti-CXCL11 mAb on the proliferation of CXCR3-A (open bar) and CXCR3-B (solid bar) transfectants. Results represent mean values (±SD) of the percentage of proliferation over isotype controls obtained in four separate experiments. (h) Opposite effects of CXCL10 on the proliferation of CXCR3-A (left bars) and CXCR3-B (right bars) transfectants. (i) Effects of CXCL4 on the proliferation of CXCR3-A (left bars) and CXCR3-B (right bars) transfectants. Results represent mean values (±SD) of the percentage of proliferation over controls (unstimulated cells) from four separate experiments *, P

Figure 4.

Activation of distinct signal transduction…

Figure 4.

Activation of distinct signal transduction pathways in CXCR3-A and CXCR3-B transfectants. (a) Increased…

Figure 4.
Activation of distinct signal transduction pathways in CXCR3-A and CXCR3-B transfectants. (a) Increased proliferative activity in CXCR3-A transfectants is strongly reduced by PTX both in basal conditions and after treatment with 500 nM CXCL10. (b) PTX has no effect on the proliferation of CXCR3-B transfectants both in basal conditions and after treatment with 4 μM CXCL10. Cells were incubated for 60 h with 1 μg/ml PTX and thymidine incorporation was assessed in the last 12 h. Columns represent mean values (±SD) of three separate experiments. *, P ++ mobilization in CXCR3-A transfectants. Results are from one representative of four experiments. (d) Effect of CXCL10, CXCL9, CXCL11, and CXCL4 (2 μM) on basal cAMP production and of CXCL10 and CXCL4 (2 μM) on forskolin (1 μM)-stimulated cAMP production in CXCR3-B transfectants. Columns represent mean values (±SD) of six separate experiments. *, P < 0.05; **, P < 0.001. (e) Absence of p21Cip1/Waf1 and p53 regulation by CXCL4 in CXCR3-A transfectants as assessed by real-time quantitative RT-PCR. (f) Up-regulation in CXCR3-B transfectants of p21Cip1/Waf1, but not of p53, mRNA levels by increasing concentrations of CXCL4 as assessed by real-time quantitative RT-PCR.

Figure 5.

Cellular expression of CXCR3-A (solid…

Figure 5.

Cellular expression of CXCR3-A (solid bars) and CXCR3-B (hatched bars) and their opposite…

Figure 5.
Cellular expression of CXCR3-A (solid bars) and CXCR3-B (hatched bars) and their opposite effects on the proliferation of different cell types. (a) CXCR3-A and CXCR3-B expression by peripheral blood–activated T cells, HMC, and HMVEC as assessed by real-time quantitative RT-PCR. Columns represent mean values (±SD) of eight separate experiments. (b) Up-regulation of HMC growth by CXCL10 and its inhibition by an anti-CXCR3 mAb. (c) Inhibitory effect of CXCL4 and of CXCL10 on the proliferation of HMVEC. The inhibitory effects of both CXCL4 and CXCL10 are reverted by an anti-CXCR3 mAb. (d) Selective expression of CXCR3-B mRNA by the ACHN cell line as detected by quantitative RT-PCR. (e) Surface expression of CXCR3 on the membrane of ACHN cells as detected by FACS® analysis performed with the 49801.111 mAb. (f) Demonstration of a single type of surface receptor in ACHN cells with an IC50 for CXCL10 and CXCL4 comparable to that observed for CXCR3-B. (g) Inhibitory effect of CXCL4 and of CXCL10 on the proliferation of ACHN cells. The inhibitory effects of both CXCL4 and CXCL10 are reverted by an anti-CXCR3 mAb. (h) Effect of CXCL10 (2 μM) on basal cAMP production and of CXCL10 (2 μM) on forskolin (1 μM)-stimulated cAMP production in CXCR3-B transfectants. Columns represent mean values (±SD) of six separate experiments. *, P < 0.05; **, P < 0.001. (i) Up-regulation in CXCR3-B transfectants of p21Cip1/Waf1, but not of p53, mRNA levels by increasing concentrations of CXCL4 as assessed by real-time quantitative RT-PCR. Symbols represent mean values (±SD) of four separate experiments. *, P < 0.01. ▪, CXCL4 plus 20 μg/ml isotype mAb; □, CXCL4 plus 20 μg/ml anti-CXCR3 mAb; •, CXCL10 plus 15 μg/ml isotype mAb; ○, CXCL10 plus 15 μg/ml anti-CXCR3 mAb (clone 49801.111).

Figure 6.

Detection of CXCR3-B protein expression…

Figure 6.

Detection of CXCR3-B protein expression by different types of cell cultures and by…

Figure 6.
Detection of CXCR3-B protein expression by different types of cell cultures and by endothelial cells of human neoplastic tissues by CXCR3-B–specific mAbs. (a) Absence of reactivity in mock transfectants stained with an anti-CXCR3-B mAb. ×100. (b) Intense staining of CXCR3-B transfectants with an anti–CXCR3-B mAb. ×100. (c) Absence of reactivity in CXCR3-A transfectants stained with an anti–CXCR3-B mAb. ×100. (d) Absence of reactivity in primary cultures of HMC stained with an anti–CXCR3-B mAb. ×100. (e) Positive staining of primary cultures of HMVEC as well as the ACHN cell line (f) with an anti–CXCR3-B mAb. ×100. (g) Absence of reactivity in normal human renal tissue stained with PL1 anti–CXCR3-B mAb. ×10. (h) Staining with the same mAb of endothelial cells in a specimen of renal cell carcinoma. (i) Staining of both endothelial and tumor cells with the anti-CXCR3 mAb 49801.111 tested in an adjacent section. (j) Absence of reactivity in an adjacent section stained with an isotype-matched control mAb. (k and l) Reactivity of endothelial cells from the same renal carcinoma specimen before and after adsorption of PL1 mAb with the peptide used for mouse immunization. (m) Reactivity with PL1 mAb of endothelial cells from a group of vessels in a renal cell carcinoma specimen as detected at a higher power magnification. ×250. (n) Staining of an adjacent section with PL2 anti–CXCR3-B mAb. (o) Double label immunohistochemistry for CXCR3-B (red) and vWf (blue-gray), showing costaining (brown). (p) Absence of CXCR3-B reactivity in normal human lung tissue stained with PL1 anti–CXCR3-B mAb. (q) Staining with the same mAb of endothelial cells in a specimen of NSCLC. (r) Staining of both endothelial cells and other cell types with the anti-CXCR3 mAb 49801.111 in an adjacent section.
Figure 4.
Figure 4.
Activation of distinct signal transduction pathways in CXCR3-A and CXCR3-B transfectants. (a) Increased proliferative activity in CXCR3-A transfectants is strongly reduced by PTX both in basal conditions and after treatment with 500 nM CXCL10. (b) PTX has no effect on the proliferation of CXCR3-B transfectants both in basal conditions and after treatment with 4 μM CXCL10. Cells were incubated for 60 h with 1 μg/ml PTX and thymidine incorporation was assessed in the last 12 h. Columns represent mean values (±SD) of three separate experiments. *, P ++ mobilization in CXCR3-A transfectants. Results are from one representative of four experiments. (d) Effect of CXCL10, CXCL9, CXCL11, and CXCL4 (2 μM) on basal cAMP production and of CXCL10 and CXCL4 (2 μM) on forskolin (1 μM)-stimulated cAMP production in CXCR3-B transfectants. Columns represent mean values (±SD) of six separate experiments. *, P < 0.05; **, P < 0.001. (e) Absence of p21Cip1/Waf1 and p53 regulation by CXCL4 in CXCR3-A transfectants as assessed by real-time quantitative RT-PCR. (f) Up-regulation in CXCR3-B transfectants of p21Cip1/Waf1, but not of p53, mRNA levels by increasing concentrations of CXCL4 as assessed by real-time quantitative RT-PCR.
Figure 5.
Figure 5.
Cellular expression of CXCR3-A (solid bars) and CXCR3-B (hatched bars) and their opposite effects on the proliferation of different cell types. (a) CXCR3-A and CXCR3-B expression by peripheral blood–activated T cells, HMC, and HMVEC as assessed by real-time quantitative RT-PCR. Columns represent mean values (±SD) of eight separate experiments. (b) Up-regulation of HMC growth by CXCL10 and its inhibition by an anti-CXCR3 mAb. (c) Inhibitory effect of CXCL4 and of CXCL10 on the proliferation of HMVEC. The inhibitory effects of both CXCL4 and CXCL10 are reverted by an anti-CXCR3 mAb. (d) Selective expression of CXCR3-B mRNA by the ACHN cell line as detected by quantitative RT-PCR. (e) Surface expression of CXCR3 on the membrane of ACHN cells as detected by FACS® analysis performed with the 49801.111 mAb. (f) Demonstration of a single type of surface receptor in ACHN cells with an IC50 for CXCL10 and CXCL4 comparable to that observed for CXCR3-B. (g) Inhibitory effect of CXCL4 and of CXCL10 on the proliferation of ACHN cells. The inhibitory effects of both CXCL4 and CXCL10 are reverted by an anti-CXCR3 mAb. (h) Effect of CXCL10 (2 μM) on basal cAMP production and of CXCL10 (2 μM) on forskolin (1 μM)-stimulated cAMP production in CXCR3-B transfectants. Columns represent mean values (±SD) of six separate experiments. *, P < 0.05; **, P < 0.001. (i) Up-regulation in CXCR3-B transfectants of p21Cip1/Waf1, but not of p53, mRNA levels by increasing concentrations of CXCL4 as assessed by real-time quantitative RT-PCR. Symbols represent mean values (±SD) of four separate experiments. *, P < 0.01. ▪, CXCL4 plus 20 μg/ml isotype mAb; □, CXCL4 plus 20 μg/ml anti-CXCR3 mAb; •, CXCL10 plus 15 μg/ml isotype mAb; ○, CXCL10 plus 15 μg/ml anti-CXCR3 mAb (clone 49801.111).
Figure 6.
Figure 6.
Detection of CXCR3-B protein expression by different types of cell cultures and by endothelial cells of human neoplastic tissues by CXCR3-B–specific mAbs. (a) Absence of reactivity in mock transfectants stained with an anti-CXCR3-B mAb. ×100. (b) Intense staining of CXCR3-B transfectants with an anti–CXCR3-B mAb. ×100. (c) Absence of reactivity in CXCR3-A transfectants stained with an anti–CXCR3-B mAb. ×100. (d) Absence of reactivity in primary cultures of HMC stained with an anti–CXCR3-B mAb. ×100. (e) Positive staining of primary cultures of HMVEC as well as the ACHN cell line (f) with an anti–CXCR3-B mAb. ×100. (g) Absence of reactivity in normal human renal tissue stained with PL1 anti–CXCR3-B mAb. ×10. (h) Staining with the same mAb of endothelial cells in a specimen of renal cell carcinoma. (i) Staining of both endothelial and tumor cells with the anti-CXCR3 mAb 49801.111 tested in an adjacent section. (j) Absence of reactivity in an adjacent section stained with an isotype-matched control mAb. (k and l) Reactivity of endothelial cells from the same renal carcinoma specimen before and after adsorption of PL1 mAb with the peptide used for mouse immunization. (m) Reactivity with PL1 mAb of endothelial cells from a group of vessels in a renal cell carcinoma specimen as detected at a higher power magnification. ×250. (n) Staining of an adjacent section with PL2 anti–CXCR3-B mAb. (o) Double label immunohistochemistry for CXCR3-B (red) and vWf (blue-gray), showing costaining (brown). (p) Absence of CXCR3-B reactivity in normal human lung tissue stained with PL1 anti–CXCR3-B mAb. (q) Staining with the same mAb of endothelial cells in a specimen of NSCLC. (r) Staining of both endothelial cells and other cell types with the anti-CXCR3 mAb 49801.111 in an adjacent section.

References

    1. Zlotnik, A., and O. Yoshie. 2000. Chemokines: a new classification system and their role in immunity. Immunity. 12:121–127.
    1. Rossi, D., and A. Zlotnik. 2000. The biology of chemokines and their receptors. Annu. Rev. Immunol. 18:217–242.
    1. Grone, H.J., C.D. Cohen, E. Grone, C. Schmidt, M. Kretzler, D. Schlondorff, and P.J. Nelson. 2002. Spatial and temporally restricted expression of chemokines and chemokine receptors in the developing human kidney. J. Am. Soc. Nephrol. 13:957–967.
    1. Belperio, J.A., M.P. Keane, D.A. Arenberg, C.L. Addison, J.E. Ehlert, M.D. Burdick, and R.M. Strieter. 2000. CXC chemokines in angiogenesis. J. Leukoc. Biol. 68:1–8.
    1. Homey, B., A. Müller, and A. Zlotnik. 2002. Chemokines: agents for the immunotherapy of cancer? Nat. Rev. Immunol. 2:175–184.
    1. Strieter, R.M., J.A. Belperio, and M.P. Keane. 2002. CXC chemokines in angiogenesis related to pulmonary fibrosis. Chest. 122:298S–301S.
    1. Arenberg, D.A., S.L. Kunkel, P.J. Polverini, S.B. Morris, M.D. Burdick, M.C. Glass, D.T. Taub, M.D. Iannettoni, R.I. Whyte, and R.M. Strieter. 1996. Interferon-γ–inducible protein 10 (IP-10) is an angiostatic factor that inhibits human non-small cell lung cancer (NSCLC) tumorigenesis and spontaneous metastases. J. Exp. Med. 184:981–992.
    1. Sgadari, C., J.M. Farber, A.L. Angiolillo, F. Liao, J. Teruya-Feldstein, P.R. Burd, L. Yao, G. Gupta, C. Kanegane, and G. Tosato. 1997. Mig, the monokine induced by interferon-gamma, promotes tumor necrosis in vivo. Blood. 89:2635–2643.
    1. Tanaka, T., Y. Manome, P. Wen, D.W. Kufe, and H.A. Fine. 1997. Viral vector-mediated transduction of a modified platelet factor 4 cDNA inhibits angiogenesis and tumor growth. Nat. Med. 3:437–442.
    1. Maione, T.E., G.S. Gray, J. Petro, A.J. Hunt, A.L. Donner, S.I. Bauer, H.F. Carson, and R.J. Sharpe. 1990. Inhibition of angiogenesis by recombinant human platelet factor-4 and related peptides. Science. 247:77–79.
    1. Romagnani, P., F. Annunziato, L. Lasagni, E. Lazzeri, C. Beltrame, M. Francalanci, M. Uguccioni, G. Galli, L. Cosmi, L. Maurenzig, et al. 2001. Cell cycle-dependent expression of CXC chemokine receptor 3 by endothelial cells mediates angiostatic activity. J. Clin. Invest. 107:53–63.
    1. Salcedo, R., J.H. Resau, D. Halverson, E.A. Hudson, M. Dambach, D. Powell, K. Wasserman, and J.J. Oppenheim. 2000. Differential expression and responsiveness of chemokine receptors (CXCR1-3) by human microvascular endothelial cells and umbilical vein endothelial cells. FASEB J. 14:2055–2064.
    1. Arenberg, D.A., A. Zlotnick, S.R. Strom, M.D. Burdick, and R.M. Strieter. 1999. The murine CC chemokine, 6C-kine, inhibits tumor growth and angiogenesis in a human lung cancer SCID mouse model. Cancer Immunol. Immunother. 49:587–592.
    1. Luster, A.D., S.M. Greenberg, and P. Leder. 1995. The IP-10 chemokine binds to a specific cell surface heparan sulfate site shared with platelet factor 4 and inhibits endothelial cell proliferation. J. Exp. Med. 182:219–231.
    1. Strieter, R.M., P.J. Polverini, S.L. Kunkel, D.A. Arenberg, M.D. Burdick, J. Kasper, J. Dzuiba, J. Van Damme, A. Walz, D. Marriott, et al. 1995. The functional role of the ELR motif in CXC chemokine-mediated angiogenesis. J. Biol. Chem. 270:27348–27357.
    1. Aronica, S.M., C. Mantel, R. Gonin, M.S. Marshall, A. Sarris, S. Cooper, N. Hague, X.F. Zhang, and H.E. Broxmeyer. 1995. Interferon-inducible protein 10 and macrophage inflammatory protein-1 alpha inhibit growth factor stimulation of Raf-1 kinase activity and protein synthesis in a human growth factor-dependent hematopoietic cell line. J. Biol. Chem. 270:21998–22007.
    1. Bonecchi, R., G. Bianchi, P.P. Bordignon, D. D'Ambrosio, R. Lang, A. Borsetti, S. Sozzani, P. Allavena, P.A. Gray, A. Mantovani, et al. 1998. Differential expression of chemokine receptors and chemotactic responsiveness of type 1 T helper cells (Th1s) and Th2s. J. Exp. Med. 187:129–134.
    1. Sallusto, F., D. Lenig, C.R. Mackay, and A. Lanzavecchia. 1998. Flexible programs of chemokine receptor expression on human polarized T helper 1 and 2 lymphocytes. J. Exp. Med. 187:875–883.
    1. Loetscher, M., B. Gerber, P. Loetscher, S.A. Jones, L. Piali, I. Clark-Lewis, M. Baggiolini, and B. Moser. 1996. Chemokine receptor specific for IP-10 and Mig: structure, function, and expression in activated T lymphocytes. J. Exp. Med. 184:963–969.
    1. Janatpour, M.J., S. Hudak, M. Sathe, J.D. Sedgwick, and L.M. McEvoy. 2001. Tumor necrosis factor–dependent segmental control of MIG expression by high endothelial venules in inflamed lymph nodes regulates monocyte recruitment. J. Exp. Med. 194:1375–1384.
    1. Penna, G., S. Sozzani, and L. Adorini. 2001. Cutting edge: selective usage of chemokine receptors by plasmacytoid dendritic cells. J. Immunol. 167:1862–1866.
    1. Romagnani, P., C. Beltrame, F. Annunziato, L. Lasagni, M. Luconi, G. Galli, L. Cosmi, E. Maggi, M. Salvadori, C. Pupilli, et al. 1999. Role for interactions between IP-10/Mig and their receptor (CXCR3) in proliferative glomerulonephritis. J. Am. Soc. Nephrol. 10:2518–2526.
    1. Romagnani, P., E. Lazzeri, L. Lasagni, C. Mavilia, C. Beltrame, M. Francalanci, M. Rotondi, F. Annunziato, L. Maurenzig, L. Cosmi, et al. 2002. IP-10 and Mig production by glomerular cells in human proliferative glomerulonephritis and regulation by nitric oxide. J. Am. Soc. Nephrol. 13:53–64.
    1. Bonacchi, A., P. Romagnani, R.G. Romanelli, E. Efsen, F. Annunziato, L. Lasagni, M. Francalanci, M. Serio, G. Laffi, M. Pinzani, et al. 2001. Signal transduction by the chemokine receptor CXCR3: activation of Ras/ERK, Src, and phosphatidylinositol 3-kinase/Akt controls cell migration and proliferation in human vascular pericytes. J. Biol. Chem. 276:9945–9954.
    1. Pupilli, C., L. Lasagni, P. Romagnani, F. Bellini, M. Mannelli, N. Misciglia, C. Mavilia, U. Vellei, D. Villari, and M. Serio. 1999. Angiotensin II stimulates the synthesis and secretion of vascular permeability factor/vascular endothelial growth factor in human mesangial cells. J. Am. Soc. Nephrol. 10:245–255.
    1. Romagnani, P., M. Rotondi, E. Lazzeri, L. Lasagni, M. Francalanci, A. Buonamano, S. Milani, P. Vitti, L. Chiovato, M. Tonacchera, et al. 2002. Expression of IP-10/CXCL10 and MIG/CXCL9 in the thyroid and increased levels of IP-10/CXCL10 in the serum of patients with recent-onset Graves' disease. Am. J. Pathol. 161:195–206.
    1. Jenh, C.H., M.A. Cox, H. Kaminski, M. Zhang, H. Byrnes, J. Fine, D. Lundell, C.C. Chou, S.K. Narula, and P.J. Zavodny. 1999. Cutting edge: species specificity of the CC chemokine 6Ckine signaling through the CXC chemokine receptor CXCR3: human 6Ckine is not a ligand for the human or mouse CXCR3 receptors. J. Immunol. 162:3765–3769.
    1. Ades, E.W., F.J. Candal, R.A. Swerlick, V.G. George, S. Summers, D.C. Bosse, and T.J. Lawley. 1992. HMEC-1: establishment of an immortalized human microvascular endothelial cell line. J. Invest. Dermatol. 99:683–690.
    1. Gengrinovitch, S., S.M. Greenberg, T. Cohen, H. Gitay-Goren, P. Rockwell, T.E. Maione, B.Z. Levi, and G. Neufeld. 1995. Platelet factor-4 inhibits the mitogenic activity of VEGF121 and VEGF165 using several concurrent mechanisms. J. Biol. Chem. 270:15059–15065.
    1. Pugliese, G., F. Pricci, G. Romeo, F. Pugliese, P. Mene, S. Giannini, B. Cresci, G. Galli, C.M. Rotella, H. Vlassara, et al. 1997. Upregulation of mesangial growth factor and extracellular matrix synthesis by advanced glycation end products via a receptor-mediated mechanism. Diabetes. 46:1881–1887.
    1. De Lean, A., P.J. Munson, and D. Rodbard. 1978. Simultaneous analysis of families of sigmoidal curves: application to bioassay, radioligand assay, and physiological dose-response curves. Am. J. Physiol. 235:E97–E102.
    1. Van Riper, G., S. Siciliano, P.A. Fischer, R. Meurer, M.S. Springer, and H. Rosen. 1993. Characterization and species distribution of high affinity GTP-coupled receptors for human RANTES and monocyte chemoattractant protein 1. J. Exp. Med. 177:851–856.
    1. Chan, A.I., and P.C. Keng. 1987. Potentiation of radiation cytotoxicity by recombinant interferons, a phenomenon associated with increased blockage at the G2-M phase of the cell cycle. Cancer Res. 47:4338–4341.
    1. Romagnani, P., F. Annunziato, R. Manetti, C. Mavilia, L. Lasagni, C. Manuelli, G.B. Vannelli, V. Vanini, E. Maggi, C. Pupilli, et al. 1998. High CD30 ligand expression by epithelial cells and Hassal's corpuscles in the medulla of human thymus. Blood. 91:3323–3332.
    1. Files, J.C., T.W. Malpass, E.K. Yee, J.L. Ritchie, and L.A. Harker. 1981. Studies of human platelet α-release in vivo. Blood. 58:607–618.
    1. Perollet, C., Z.C. Han, C. Savona, J.B. Caen, and A. Bikfalvi. 1998. Platelet factor 4 modulates fibroblast growth factor 2 (FGF-2) activity and inhibits FGF-2 dimerization. Blood. 91:3289–3299.
    1. Sulpice, E., M. Bryckaert, J. Lacour, J.O. Contreres, and G. Tobelem. 2002. Platelet factor 4 inhibits FGF2-induced endothelial cell proliferation via the extracellular signal-regulated kinase pathway but not by the phosphatidylinositol 3-kinase pathway. Blood. 100:3087–3094.
    1. Gentilini, G., N.C. Kirschbaum, J.A. Augustine, R.H. Aster, and G.P. Visenti. 1999. Inhibition of human umbilical vein endothelial cell proliferation by the CXC chemokine, platelet factor 4 (PF-4), is associated with impaired downregulation of p21Cip1/WAF1. Blood. 93:25–33.
    1. Shiraha, H., A. Glading, K. Gupta, and A. Wells. 1999. IP-10 inhibits epidermal growth factor–induced motility by decreasing epidermal growth factor receptor–mediated calpain activity. J. Cell Biol. 146:243–254.
    1. Maione, T.E., G.S. Gray, A.J. Hunt, and R.J. Sharpe. 1991. Inhibition of tumor growth in mice by an analogue of platelet factor 4 that lacks affinity for heparin and retains potent angiostatic activity. Cancer Res. 51:2077–2083.
    1. Lecomte-Raclet, L., M. Alemany, A. Sequeira-Le Grand, J. Amiral, G. Quentin, A.M. Vissac, J.P. Caen, and Z.C. Han. 1998. New insights into the negative regulation of hematopoiesis by chemokine platelet factor 4 and related peptides. Blood. 91:2772–2780.
    1. Jouan, V., X. Canron, M. Alemany, J.P. Caen, G. Quentin, J. Plouet, and A. Bikfalvi. 1999. Inhibition of in vitro angiogenesis by platelet factor-4-derived peptides and mechanism of action. Blood. 94:984–993.
    1. Hansell, P., T.E. Maione, and P. Borgstrom. 1995. Selective binding of platelet factor 4 to regions of active angiogenesis in vivo. Am. J. Physiol. 269:H829–H836.
    1. Soto, H., W. Wang, R.M. Strieter, N.G. Copeland, D.J. Gilbert, N.A. Jenkins, J. Hedrick, and A. Zlotnik. 1998. The CC chemokine 6Ckine binds the CXC chemokine receptor CXCR3. Proc. Natl. Acad. Sci. USA. 95:8205–8210.
    1. Rappert, A., K. Biber, C. Nolte, M. Lipp, A. Schubel, B. Lu, N.P. Gerard, C. Gerard, H.W. Boddeke, and H. Kettenmann. 2002. Secondary lymphoid tissue chemokine (CCL21) activates CXCR3 to trigger a Cl-current and chemotaxis in murine microglia. J. Immunol. 168:3221–3226.
    1. Maghazachi, A.A., B.S. Skalhegg, B. Rolstad, and A. Al-Aoukaty. 1997. Interferon-inducible protein-10 and lymphotactin induce the chemotaxis and mobilization of intracellular calcium in natural killer cells through pertussis toxin-sensitive and -insensitive heterotrimeric G-proteins. FASEB J. 11:765–774.
    1. Marinissen, M.J., and J.S. Gutkind. 2001. G-protein-coupled receptors and signaling networks: emerging paradigms. Trends Pharmacol. Sci. 22:368–376.
    1. Kim, S., M. Bakre, H. Yin, and J.A. Varner. 2002. Inhibition of endothelial cell survival and angiogenesis by protein kinase A. J. Clin. Invest. 110:933–941.
    1. Ho, H.H., D. Du, and M.C. Gershengorn. 1999. The N terminus of Kaposi's sarcoma-associated herpesvirus G protein-coupled receptor is necessary for high affinity chemokine binding but not for constitutive activity. J. Biol. Chem. 274:31327–31332.
    1. Couty, J.P., E. Geras-Raaka, B.B. Weksler, and M.C. Gershengorn. 2001. Kaposi's sarcoma-associated herpesvirus G protein-coupled receptor signals through multiple pathways in endothelial cells. J. Biol. Chem. 276:33805–33811.
    1. Zhou, Y., S. Wang, B.G. Yue, A. Gobl, and K. Oberg. 2002. Effects of interferon alpha on the expression of p21cip1/waf1 and cell cycle distribution in carcinoid tumors. Cancer Invest. 20:348–356.
    1. Lee, T.H., L.Y. Chuang, and W.C. Hung. 2000. Induction of p21WAF1 expression via Sp1-binding sites by tamoxifen in estrogen receptor-negative lung cancer cells. Oncogene. 19:3766–3773.

Source: PubMed

3
Tilaa