The Role of CD47 in Neutrophil Transmigration
Yuan Liu,Didier Merlin,Stephanie L. Burst,Mildred Pochet,James Madara,Charles A. Parkos
DOI: https://doi.org/10.1074/jbc.m104138200
2001-01-01
Abstract:CD47, a cell surface glycoprotein, plays an important role in modulating neutrophil (PMN) migration across endothelial and epithelial monolayers. Here we show that anti-CD47 monoclonal antibodies (mAbs) delay PMN migration across collagen-coated filters or T84 epithelial monolayers toward the chemoattractant formylmethionylleucylphenylalanine (fMLP). Despite delayed transmigration by anti-CD47 mAbs, the numbers of PMN migrating across in either condition were the same as in the presence of control non-inhibitory mAbs. Cell surface labeling and immunoprecipitation demonstrated upregulation of CD47 to the PMN cell surface with kinetics similar to those of the transmigration response. Subcellular fractionation studies revealed redistribution of CD47 from intracellular compartments that co-sediment with secondary granules to plasma membrane-containing fractions after fMLP stimulation. Experiments performed to investigate potential signaling pathways revealed that inhibition of tyrosine phosphorylation with genistein reversed the anti-CD47-mediated PMN migration delay, whereas inhibition of phosphatidylinositol 3-kinase only partially reversed anti-CD47 effects that correlated with a rapid increase in PMN cell surface CD47. Analysis of the contribution of epithelial-expressed CD47 to PMN transmigration revealed that PMN migration across CD47-deficient epithelial monolayers (CaCO2) was significantly increased after stable transfection with CD47. These results suggest that cell surface CD47 and downstream tyrosine phosphorylation signaling events regulate, in part, the rate of PMN migration during the inflammatory response. CD47, a cell surface glycoprotein, plays an important role in modulating neutrophil (PMN) migration across endothelial and epithelial monolayers. Here we show that anti-CD47 monoclonal antibodies (mAbs) delay PMN migration across collagen-coated filters or T84 epithelial monolayers toward the chemoattractant formylmethionylleucylphenylalanine (fMLP). Despite delayed transmigration by anti-CD47 mAbs, the numbers of PMN migrating across in either condition were the same as in the presence of control non-inhibitory mAbs. Cell surface labeling and immunoprecipitation demonstrated upregulation of CD47 to the PMN cell surface with kinetics similar to those of the transmigration response. Subcellular fractionation studies revealed redistribution of CD47 from intracellular compartments that co-sediment with secondary granules to plasma membrane-containing fractions after fMLP stimulation. Experiments performed to investigate potential signaling pathways revealed that inhibition of tyrosine phosphorylation with genistein reversed the anti-CD47-mediated PMN migration delay, whereas inhibition of phosphatidylinositol 3-kinase only partially reversed anti-CD47 effects that correlated with a rapid increase in PMN cell surface CD47. Analysis of the contribution of epithelial-expressed CD47 to PMN transmigration revealed that PMN migration across CD47-deficient epithelial monolayers (CaCO2) was significantly increased after stable transfection with CD47. These results suggest that cell surface CD47 and downstream tyrosine phosphorylation signaling events regulate, in part, the rate of PMN migration during the inflammatory response. polymorphonuclear leukocyte immunoglobulin variable domain myeloperoxidase Hanks' balanced salt buffer devoid of Ca2+ and Mg2+ formylmethionylleucylphenylalanine phosphatidylinositol 3-kinase monoclonal antibody Dulbecco's modified Eagle's medium junction adhesion molecule thrombospondin-1 fluorescence-activated cell sorting protein kinase C mitogen-activated protein kinase/extracellular signal-regulated kinase kinase fluorescein isothiocyanate polymerase chain reaction alkaline phosphatase In many inflammatory conditions, large numbers of PMN1migrate across vascular endothelium, cell matrix, and mucosal surfaces in response to chemotactic signals. Under these conditions, PMN release active oxygen species, proteolytic enzymes, and inflammatory mediators that result in tissue injury. Thus, it is intuitive that large scale PMN migration and accumulation in tissues correlates with disease activity and patient symptoms (1Stockley R.A. Am. J. Med. 1995; 99: 8S-13SAbstract Full Text PDF PubMed Scopus (47) Google Scholar,2Kumar N.B. Nostrant T.T. Appelman H.D. Am. J. Surg. Pathol. 1982; 6: 523-529Crossref PubMed Scopus (142) Google Scholar). The process of PMN movement from the vasculature, through the interstitial matrix, and across mucosal surfaces requires sequential interactions between neutrophil surface molecules and counter-receptors present on tissues or cell surfaces (3Parkos C.A. Bioessays. 1997; 19: 865-873Crossref PubMed Scopus (67) Google Scholar, 4Lawrence M.B. Springer T.A. Cell. 1991; 65: 859-873Abstract Full Text PDF PubMed Scopus (1887) Google Scholar, 5Muller W.A. Weigl S.A. Deng X. Phillips D.M. J. Exp. Med. 1993; 178: 449-460Crossref PubMed Scopus (990) Google Scholar, 6Spertini O. Kansas G.S. Munro J.M. Griffin J.D. Tedder T.F. Nature. 1991; 349: 691-694Crossref PubMed Scopus (235) Google Scholar). Previous studies have shown that the leukocyte β2 integrin CD11bCD18 is essential in this process, especially in early interactions with epithelial and endothelial cells during the transmigration response (3Parkos C.A. Bioessays. 1997; 19: 865-873Crossref PubMed Scopus (67) Google Scholar,7Diamond M.S. Alon R. Parkos C.A. Quinn M.T. Springer T.A. J. Cell Biol. 1995; 130: 1473-1482Crossref PubMed Scopus (255) Google Scholar, 8von Andrian U.H. Chambers J.D. McEvoy L.M. Bargatze R.F. Arfors K.E. Butcher E.C. Proc. Natl. Acad. Sci. U. S. A. 1991; 88: 7538-7542Crossref PubMed Scopus (901) Google Scholar). Although PMN migration across endothelial monolayers is only partially blocked by anti-CD11bCD18 antibodies, there is near complete inhibition of PMN migration across epithelial monolayers by the same antibodies (9Parkos C.A. Delp C. Arnaout M.A. Madara J.L. J. Clin. Invest. 1991; 88: 1605-1612Crossref PubMed Scopus (292) Google Scholar). The importance of β2 integrins in PMN transmigration is manifested clinically by patients with leukocyte adhesion deficiency who lack β2 integrins, resulting in a failure of PMN to migrate out of the vasculature and leaving high levels of circulating PMN (10Anderson D.C. Schmalstieg F.C. Arnaout M.A. Kohl S. Tosi M.F. Dana N. Buffone G.J. Hughes B.J. Brinkley B.R. Dickey W.D. et al.J. Clin. Invest. 1984; 74: 536-551Crossref PubMed Scopus (159) Google Scholar, 11Dana N. Todd R.F. d. Pitt J. Springer T.A. Arnaout M.A. J. Clin. Investig. 1984; 73: 153-159Crossref PubMed Scopus (111) Google Scholar). A second protein that has been shown to play an important role in PMN transmigration is CD47 (12Parkos C.A. Colgan S.P. Liang T.W. Nusrat A. Bacarra A.E. Carnes D.K. Madara J.L. J. Cell Biol. 1996; 132: 437-450Crossref PubMed Scopus (183) Google Scholar). PMN migration across both epithelial and endothelial monolayers can be inhibited by anti-CD47 antibodies (3Parkos C.A. Bioessays. 1997; 19: 865-873Crossref PubMed Scopus (67) Google Scholar, 12Parkos C.A. Colgan S.P. Liang T.W. Nusrat A. Bacarra A.E. Carnes D.K. Madara J.L. J. Cell Biol. 1996; 132: 437-450Crossref PubMed Scopus (183) Google Scholar, 13Cooper D. Lindberg F.P. Gamble J.R. Brown E.J. Vadas M.A. Proc. Natl. Acad. Sci. U. S. A. 1995; 92: 3978-3982Crossref PubMed Scopus (185) Google Scholar). CD47 was originally defined as a tumor-specific marker for ovarian carcinoma but is now known to be widely expressed on all hematopoietic cells and most other cell types (14Mawby W.J. Holmes C.H. Anstee D.J. Spring F.A. Tanner M.J. Biochem. J. 1994; 304: 525-530Crossref PubMed Scopus (114) Google Scholar, 15Lindberg F.P. Gresham H.D. Schwarz E. Brown E.J. J. Cell Biol. 1993; 123: 485-496Crossref PubMed Scopus (307) Google Scholar, 16Reinhold M.I. Lindberg F.P. Plas D. Reynolds S. Peters M.G. Brown E.J. J. Cell Sci. 1995; 108: 3419-3425Crossref PubMed Google Scholar). The primary structure of human and mouse CD47 predicts the protein containing an N-terminal extracellular IgV domain, a membrane-spanning domain with multiple transmembrane α helices, and an intracellular loop (15Lindberg F.P. Gresham H.D. Schwarz E. Brown E.J. J. Cell Biol. 1993; 123: 485-496Crossref PubMed Scopus (307) Google Scholar). In addition to regulating PMN transmigration, other studies have implicated CD47 in multiple cellular functions. In particular, antibodies to CD47 have been shown to interfere with αvβ3-mediated cell functions (15Lindberg F.P. Gresham H.D. Schwarz E. Brown E.J. J. Cell Biol. 1993; 123: 485-496Crossref PubMed Scopus (307) Google Scholar), as well as to block endothelial Ca2+ influxes during adhesion to fibronectin- or vitronectin-coated surfaces (17Schwartz M.A. Brown E.J. Fazeli B. J. Biol. Chem. 1993; 268: 19931-19934Abstract Full Text PDF PubMed Google Scholar). CD47 has been shown to bind to the C terminus of thrombospondin-1 (18Gao A.G. Lindberg F.P. Finn M.B. Blystone S.D. Brown E.J. Frazier W.A. J. Biol. Chem. 1996; 271: 21-24Abstract Full Text Full Text PDF PubMed Scopus (332) Google Scholar), therefore suggesting a role in platelet activation. Anti-CD47 antibodies have also been used to implicate CD47 in T cell activation (19Waclavicek M. Majdic O. Stulnig T. Berger M. Baumruker T. Knapp W. Pickl W.F. J. Immunol. 1997; 159: 5345-5354PubMed Google Scholar, 20Reinhold M.I. Lindberg F.P. Kersh G.J. Allen P.M. Brown E.J. J. Exp. Med. 1997; 185: 1-11Crossref PubMed Scopus (140) Google Scholar), T and B cell apoptosis (21Pettersen R.D. Hestdal K. Olafsen M.K. Lie S.O. Lindberg F.P. J. Immunol. 1999; 162: 7031-7040PubMed Google Scholar, 22Mateo V. Lagneaux L. Bron D. Biron G. Armant M. Delespesse G. Sarfati M. Nat. Med. 1999; 5: 1277-1284Crossref PubMed Scopus (198) Google Scholar), and stroma-supported erythropoiesis (23Furusawa T. Yanai N. Hara T. Miyajima A. Obinata M. J. Biochem. 1998; 123: 101-106Crossref PubMed Scopus (23) Google Scholar). Recently, CD47 has been shown to bind to P84 (also termed SIRPα, Bit, SHPS-1, and MFR) (24Jiang P. Lagenaur C.F. Narayanan V. J. Biol. Chem. 1999; 274: 559-562Abstract Full Text Full Text PDF PubMed Scopus (275) Google Scholar, 25Seiffert M. Cant C. Chen Z. Rappold I. Brugger W. Kanz L. Brown E.J. Ullrich A. Buhring H.J. Blood. 1999; 94: 3633-3643Crossref PubMed Google Scholar, 26Vernon-Wilson E.F. Kee W.J. Willis A.C. Barclay A.N. Simmons D.L. Brown M.H. Eur. J. Immunol. 2000; 30: 2130-2137PubMed Google Scholar), and this interaction has been broadly implicated in the regulation of memory formation, macrophage multinucleation, B cell aggregation, and red blood cell self-recognition in mice (27Babic I. Schallhorn A. Lindberg F.P. Jirik F.R. J. Immunol. 2000; 164: 3652-3658Crossref PubMed Scopus (47) Google Scholar, 28Oldenborg P.A. Zheleznyak A. Fang Y.F. Lagenaur C.F. Gresham H.D. Lindberg F.P. Science. 2000; 288: 2051-2054Crossref PubMed Scopus (1313) Google Scholar). Despite the diverse array of functional studies on CD47, the biochemical/molecular mechanism(s) of CD47 function remain undefined. Although we have previously observed anti-CD47 antibody-mediated inhibition of PMN transmigration (12Parkos C.A. Colgan S.P. Liang T.W. Nusrat A. Bacarra A.E. Carnes D.K. Madara J.L. J. Cell Biol. 1996; 132: 437-450Crossref PubMed Scopus (183) Google Scholar), the molecular basis of this effect is not clear. The objective of the current study was to investigate the mechanism of CD47 function in PMN migration across epithelial cell monolayers and collagen-coated filters. Using a time-course transmigration assay, we demonstrate that fMLP-driven PMN migration across epithelial monolayers and collagen-coated filters is significantly delayed after cell surface CD47 is blocked by functionally inhibitory anti-CD47 mAbs. However, despite delayed transmigration caused by anti-CD47 mAbs, the number of PMN that eventually migrate is the same as that observed with isotype-matched binding controls. Cell surface labeling and immunoprecipitation experiments demonstrate increased CD47 on the PMN cell surface after stimulation with fMLP and after transmigration. PMN subcellular fractionation experiments revealed that increased cell surface CD47 is derived from intracellular membranes that co-sediment with the secondary (specific) granules. Experiments performed to investigate potential signaling pathways revealed that inhibition of PI3K partially reversed anti-CD47 mediated inhibition of migration while also directly enhancing the rate of PMN migration in a fashion that paralleled a rapid increase of PMN cell surface CD47. Additional inhibitor studies revealed that tyrosine phosphorylation inhibition by genistein reversed anti-CD47 effect in a near complete fashion. In further support of the notion that cell surface CD47 positively regulates the rate of migration, we found that transfection of CD47 into deficient epithelial cells (CaCO2) results in enhanced PMN transepithelial migration. Taken together, these results suggest that translocation of CD47 to the cell surface and tyrosine kinase-mediated signaling events are key components that determine the rate of PMN migration in an inflammatory response. T84 cells (passages 60–80) were grown in a 1:1 mixture of Dulbecco's modified Eagle's medium (DMEM) and Ham's F-12 medium supplemented with 15 mm HEPES buffer (pH 7.5), 14 mm NaHCO3, 40 µg/ml penicillin, 8 µg/ml ampicillin, 90 µg/ml streptomycin, and 6% newborn calf serum. Cells were subcultured or harvested every 6–8 days with 0.1% trypsin and 1.0 mm EDTA in Ca2+ and Mg2+-free phosphate-buffered saline. For transmigration experiments, T84 cells were grown on collagen-coated, permeable polycarbonate filters (5-µm pore size) with a surface area of 0.33 cm2 (Costar, Cambridge, MA) as previously described (12Parkos C.A. Colgan S.P. Liang T.W. Nusrat A. Bacarra A.E. Carnes D.K. Madara J.L. J. Cell Biol. 1996; 132: 437-450Crossref PubMed Scopus (183) Google Scholar). CaCO2 cells were cultured in DMEM supplemented with 10% fetal bovine serum. Transfected CaCO2 cells (CaCO2-pCB6 and CaCO2-CD47) were maintained in the same medium containing 1 mg/ml G418. PMN were isolated from whole blood of normal human volunteers as previously described (12Parkos C.A. Colgan S.P. Liang T.W. Nusrat A. Bacarra A.E. Carnes D.K. Madara J.L. J. Cell Biol. 1996; 132: 437-450Crossref PubMed Scopus (183) Google Scholar). Isolated PMN were resuspended in modified Hanks' balanced salt solution devoid of calcium or magnesium (HBSS(−)) (4 °C) at a concentration of 5 × 107 cells/ml and used within 4 h of isolation. C5D5, a functionally inhibitory mAb (IgG1) was produced in our laboratory as previously reported (12Parkos C.A. Colgan S.P. Liang T.W. Nusrat A. Bacarra A.E. Carnes D.K. Madara J.L. J. Cell Biol. 1996; 132: 437-450Crossref PubMed Scopus (183) Google Scholar). B6H12.2, another functionally inhibitory anti-CD47 mAb (IgG1) (13Cooper D. Lindberg F.P. Gamble J.R. Brown E.J. Vadas M.A. Proc. Natl. Acad. Sci. U. S. A. 1995; 92: 3978-3982Crossref PubMed Scopus (185) Google Scholar) was purified from supernatants of hybridoma cells obtained from the American Tissue Culture Co. CD47 mAbs PF2.1, PF3.1, and PF10.2 were produced in our laboratory by immunizing female balb/C mice with CD47 immunopurified from 50 g of human spleen cells using previously described methods (12Parkos C.A. Colgan S.P. Liang T.W. Nusrat A. Bacarra A.E. Carnes D.K. Madara J.L. J. Cell Biol. 1996; 132: 437-450Crossref PubMed Scopus (183) Google Scholar). Splenocytes from immunized mice were fused with P3U1 myeloma cells and cultured in selection media containing DBA-2 feeder thymocytes. Hybridoma supernatants were screened for reactivity with purified CD47 by enzyme-linked immunosorbent assay in concert with cell surface staining by immunofluorescence and flow cytometry on T84 epithelial cells and PMN. Hybridomas were then subcloned by limiting dilution, weaned from selection media, and injected into the peritoneal cavities of balb/C mice for ascites fluid production. Antibodies were purified using protein-A-Sepharose affinity chromatography and dialyzed against 150 mm NaCl, 10 mm HEPES, pH 7.4. Monoclonal antibody PF3.1 was coupled to CNBr-activated Sepharose at 2–3 mg/ml described by the manufacturer (Amersham Pharmacia Biotech) and used for immunoprecipitation of CD47. A polyclonal antibody (R12989) was also produced by immunizing rabbits with synthetic peptide SNQKTIQPPRNN, corresponding to a region of the putative C terminus of CD47 (15Lindberg F.P. Gresham H.D. Schwarz E. Brown E.J. J. Cell Biol. 1993; 123: 485-496Crossref PubMed Scopus (307) Google Scholar). As a non-inhibitory binding control, mAb J3F.1 (mouse IgG1) reactive with human junction adhesion molecule (JAM) was used (29Liu Y. Nusrat A. Schnell F.J. Reaves T.A. Walsh S. Pochet M. Parkos C.A. J. Cell Sci. 2000; 113: 2363-2374Crossref PubMed Google Scholar). Similar to CD47, JAM is a transmembrane IgV superfamily member expressed on the surfaces of both PMN and epithelial cells (29Liu Y. Nusrat A. Schnell F.J. Reaves T.A. Walsh S. Pochet M. Parkos C.A. J. Cell Sci. 2000; 113: 2363-2374Crossref PubMed Google Scholar). As a positive functionally inhibitory control, anti-CD11b mAb CBMR1/29, was used (30Balsam L.B. Liang T.W. Parkos C.A. J. Immunol. 1998; 160: 5058-5065PubMed Google Scholar). Anti-β3 integrin mAbs 15, 33, 27, and LIBs-1 were kind gifts from Dr. Mark Ginsberg. LM609 was kindly provided by Dr. David Cherish. An additional anti-β3 mAb AP1 was purchased from GTI Inc. A functionally blocking mAb to β1 integrin, mAb 13, was kindly provided by Dr. Kenneth Yamada. Purified human thrombospondin-1 (TS1) and anti-TS1 polyclonal antibody R1 were kindly provided by Dr. Jack Lawler (31Sipes J.M. Krutzsch H.C. Lawler J. Roberts D.D. J. Biol. Chem. 1999; 274: 22755-22762Abstract Full Text Full Text PDF PubMed Scopus (55) Google Scholar). Rabbit polyclonal antibody against human CD11b R7928A was raised by immunizing rabbit with a peptide (DMMSEGGPPGAEPQ) corresponding to the C terminus of CD11b. Peptide 4N1K (KRFYVVMWKK) corresponding to the C-terminal cell binding domain of thrombospondin, which has previously been shown to bind to CD47-transfected cells (18Gao A.G. Lindberg F.P. Finn M.B. Blystone S.D. Brown E.J. Frazier W.A. J. Biol. Chem. 1996; 271: 21-24Abstract Full Text Full Text PDF PubMed Scopus (332) Google Scholar, 32Wang X.Q. Frazier W.A. Mol. Biol. Cell. 1998; 9: 865-874Crossref PubMed Scopus (140) Google Scholar), was synthesized and high performance liquid chromatography-purified to >90% by the Emory Microchemical Facility. PMN transepithelial migration experiments were performed using epithelial monolayers as previously described (12Parkos C.A. Colgan S.P. Liang T.W. Nusrat A. Bacarra A.E. Carnes D.K. Madara J.L. J. Cell Biol. 1996; 132: 437-450Crossref PubMed Scopus (183) Google Scholar). Briefly, confluent T84 or CaCO2 monolayers were washed twice with HBSS (20 °C). PMN (1 × 106) in 150 µl of HBSS with or without antibody was added to the upper chamber of the monolayer setup. Transmigration was initiated by adding 1 ml of 1 µm fMLP (in HBSS) into the lower chamber followed by incubation at 37 °C. PMN migration across monolayers into the fMLP-containing lower chambers was quantified by myeloperoxidase (MPO) assay as previously described (12Parkos C.A. Colgan S.P. Liang T.W. Nusrat A. Bacarra A.E. Carnes D.K. Madara J.L. J. Cell Biol. 1996; 132: 437-450Crossref PubMed Scopus (183) Google Scholar). To assay the time course of PMN transmigration, the upper chambers of Transwell devices containing PMN and monolayers were gently moved to new wells containing 1 ml of 1 µm fMLP in 24-well plates at every 30 min as depicted in Fig. 1. PMN migration into the lower chambers at the different time periods was then analyzed by MPO assay. To assay the PMN associated with epithelial monolayers, transmigration was stopped at different time points, and monolayers were washed with HBSS and lysed with 0.5% Triton X-100. Monolayer lysates were then assayed for myeloperoxidase. Apical to basolateral PMN transepithelial migration was performed similarly except that twice as many PMN (2 × 106) were added onto the upper chamber. Additional transmigration experiments were performed using collagen-coated filters devoid of epithelial cells. For these experiments, 20 µl of a PMN suspension containing 1 × 106 cells was stimulated to transmigrate from the upper chamber to the lower chamber containing 0.1 µm fMLP. A number of signal transduction pathway inhibitors were tested for the ability to reverse the effects of anti-CD47 mAbs on PMN transmigration. For these experiments, PMN were pre-treated for 15–60 min with the following agents prior to use in transfilter migration assays: 50–100 nm wortmannin or 20–100 µm LY294002 (PI3K inhibitor (Sigma Chemical Co.)), 0.5 µm straurosporine (PKC and other kinases inhibitor (Biomol)), 10–200 µg/ml genistein (tyrosine phosphorylation inhibitor) and its analog daidzein (both from Biomol), 0.1 µm AG126 (tyrosine phosphorylation inhibitor (Biomol)), 40 µm PP1 (Src family tyrosine kinase inhibitor (Biomol)), 20 µm SB203580 (mitogen-activated protein kinase inhibitor (Biomol)), 20 µm PD-98059 (MEK inhibitor (Biomol)), 10 µmU73122 (phospholipase C inhibitor (Biomol)), 1 mm propranolol (phospholipase D inhibitor (Biomol)), 1 µm GF109203X (PKC inhibitor (Biomol)), 20 µm AG490 (Jak kinase inhibitor (Biomol)), 20 µm H-8 (PKA/G inhibitor (Biomol)), 10 µm H-89 (PKA inhibitor (Biomol)), etc. Control migration was done in the presence of the same dilution of vehicle (Me2SO) in HBSS. For G-protein pathway inhibition experiments, PMN were pre-treated with 200–2000 ng/ml pertussis toxin (Calbiochem) for 2 h at 37 °C before use in migration assays. Surface CD47 labeling was performed by incubation of 100 µl of non-permeabilized PMN (5 × 106 cells) with anti-CD47 mAb C5D5 (20 µg/ml) for 1 h at 4 °C in HBSS after blocking nonspecific binding with 0.5% bovine serum albumin. After washing, PMN were incubated with FITC-conjugated goat anti-mouse secondary antibody (1:200 dilution in HBSS) (PharMingen) for 30 min at 4 °C followed by wash before flow cytometric analysis. CD47 labeling in this condition was compared to that obtained with normal mouse IgG1. To assay PMN surface CD47 expression during transmigration, PMN that remained in the upper chambers or had migrated into the lower chambers of Transwell setups were cooled on ice after various migration time periods followed by antibody labeling and flow cytometry. Cell surface CD47 on PMN was also assessed after PMN were stimulated with fMLP. In these experiments, 5 × 106 PMN in 2 ml of HBSS were allowed to settle in 24-well or 6-well non-tissue culture plates at room temperature for 5 min. fMLP was then added (50 nm) followed by incubation at 37 °C for various time periods. Cells were then chilled on ice and labeling performed as above. PMN (1.5 × 107/ml), in suspension or adherent to 6-well plates, were stimulated with fMLP followed by cell surface biotinylation with 0.5 mg of sulfo-NHS-Biotin (Pierce) in 1 ml of HBSS for 1 h on ice. Excess free Biotin was washed off and quenched with 100 mmNH4Cl. Cells were then solubilized with lysis buffer containing 150 mm NaCl, 2 mmCaCl2, 2 mm Mg Cl2, 1% Triton, 2 µg/ml aprotinin, 1 mm phenylmethylsulfonyl fluoride, 2 µg/ml leupeptin, and 1 µg/ml pepstatin. Cell lysates (200–400 µg of total protein) were precleared with control IgG before incubation with 10 µl of PF3.1-Sepharose (conjugated at 2 mg of antibody/ml of Sepharose) for 3 h (4 °C). Washed immunoprecipitates were boiled in 100 µl of sample buffer and subjected to SDS-polyacrylamide gel electrophoresis followed by transfer to nitrocellulose. Nonspecific binding was blocked by 5% nonfat dry milk in TTBS (50 mm Tris. HCl, 200 nm NaCl, 0.05% Tween 20, pH 7.4). Biotin-labeled cell surface CD47 was detected with streptavidin-peroxidase followed by enhanced chemiluminescence (ECL (Amersham Pharmacia Biotech)). Total CD47 was detected by blotting with polyclonal rabbit anti-CD47 antibody R12989 (1:200) and was visualized with peroxidase-conjugated goat anti-rabbit secondary and ECL. Freshly isolated, unstimulated, and fMLP-stimulated PMN (2 × 108/per condition) were resuspended in 5 ml of nitrogen cavitation buffer (0.34m sucrose, 10 mm Hepes, 1 mm EDTA, 0.1 mm MgCl2, 1 mmNa2ATP, pH 7.4) followed by lysis by nitrogen cavitation (15 min, 400 p.s.i.) (4 °C). The lysates were centrifuged (1000 × g) and supernatants were then subjected to isopycnic sucrose density gradient fractionation on linear 20–55% sucrose gradients in a Beckman SW-28 swinging bucket rotor (100,000 × g; 3 h, 4 °C) exactly as previously described (33Parkos C.A. Cochrane C.G. Schmitt M. Jesaitis A.J. J. Biol. Chem. 1985; 260: 6541-6547Abstract Full Text PDF PubMed Google Scholar). 1.5-ml fractions were collected from each gradient and analyzed for sucrose density, protein, plasma membrane (alkaline phosphatase), primary granules (myeloperoxidase), and secondary granules (lactoferrin) as previously described (33Parkos C.A. Cochrane C.G. Schmitt M. Jesaitis A.J. J. Biol. Chem. 1985; 260: 6541-6547Abstract Full Text PDF PubMed Google Scholar). The entire protein coding region of CD47 was PCR-amplified from reverse-transcribed human colonic cDNA with primers C3 (sense) (5′-atgataagcttcctgcatgtggcccctggtagcg) and C6 (antisense) (5′-agcttctagactactctccaaatcggagtccat) located at the 5′-untranslated repeat and 3′-untranslated repeat, respectively. Amplified PCR product was digested with HindIII/XbaI and cloned into pCB6, which had been predigested with the same restriction endonucleases. CaCO2 cells were transfected with pCB6-CD47 or pCB6 (control transfection) using Lipofectin reagent as described by the manufacturer (Life Technologies, Inc.). Stable transfectants were selected by culturing in DMEM containing 1.2 mg/ml Geneticin (Life Technologies, Inc.). Cell surface expression of CD47 was confirmed by immunofluorescence labeling and FACS analysis. The putative extracellular domain of human CD47, including the signal peptide, was PCR-amplified by primers C3(5′-atgataagcttcctgcatgtggcccctggtagcgg) and C4 (5′-ttgattagatctatttggagaaaaccatga) from reverse-transcribed T84 cDNA. The DNA fragment was cloned into the mammalian expression vector AP-tag2 (34Cheng H.J. Flanagan J.G. Cell. 1994; 79: 157-168Abstract Full Text PDF PubMed Scopus (328) Google Scholar), which contains the catalytic domain of human placental alkaline phosphatase (AP) at the C terminus, throughBglII and HindIII restriction sites. After transformation, at least three independent clones were isolated and the entire protein-coding region was confirmed by DNA sequencing. Transient transfections of COS-7 were performed using a standard DEAE-dextran method (35Liu Y. The Mechanism of GAG Autoproteolytic Activation and Enzyme Catalysis.Ph.D. dissertation. University of South Alabama, Mobile, AL1997Google Scholar). Protein expression and secretion was monitored by assaying AP activity in cell culture medium (34Cheng H.J. Flanagan J.G. Cell. 1994; 79: 157-168Abstract Full Text PDF PubMed Scopus (328) Google Scholar). Expressed CD47-AP fusion protein in medium was affinity-purified by PF3.1-Sepharose and eluted with 10 mm TEA/HCl buffer containing 100 mmNaCl, pH 10.5, followed by neutralization, concentration, and dialysis. Previously, we demonstrated that anti-CD47 mAb C5D5 inhibited PMN migration across T84 monolayers (12Parkos C.A. Colgan S.P. Liang T.W. Nusrat A. Bacarra A.E. Carnes D.K. Madara J.L. J. Cell Biol. 1996; 132: 437-450Crossref PubMed Scopus (183) Google Scholar). However, we noticed that the efficiency of inhibition by C5D5 or another anti-CD47 antibody, B6H12.2 (13Cooper D. Lindberg F.P. Gamble J.R. Brown E.J. Vadas M.A. Proc. Natl. Acad. Sci. U. S. A. 1995; 92: 3978-3982Crossref PubMed Scopus (185) Google Scholar), was variable in the standard transmigration assays typically consisting of a single time point (normally 2 h). We surmised that the variable inhibition observed might be the result of differences in PMN motility after CD47 inhibition. To address this and further investigate the mechanism of CD47-mediated migration, we modified the in vitro transepithelial migration assay in which PMN migration was assessed at several time points (Fig. 1). Using this assay setup, we tested the effect of inhibitory anti-CD47 mAbs C5D5 and B6H12.2 on the time course of PMN migration across T84 monolayers. As controls, parallel experiments were performed with binding, non-inhibitory isotype-matched anti-JAM mAb J3F.1 (29Liu Y. Nusrat A. Schnell F.J. Reaves T.A. Walsh S. Pochet M. Parkos C.A. J. Cell Sci. 2000; 113: 2363-2374Crossref PubMed Google Scholar) and an inhibitory control antibody reactive with CD11b CD18 (CBRM1/29 (30Balsam L.B. Liang T.W. Parkos C.A. J. Immunol. 1998; 160: 5058-5065PubMed Google Scholar)). As shown in Fig. 2A, in the presence of control non-inhibitory mAbs, less than 10% of the total applied PMN migrated across T84 monolayers during the first 30 min (5.8 ± 0.6% for no antibody and 6.1 ± 2.0% for J3F.1). Massive PMN migration across T84 monolayers occurred during the next 30-min period (2nd 30′) with ∼40% of the applied PMN migrating into the lower chambers (43.6 ± 3.7% for no antibody and 38.7 ± 4.4% for J3F.1). In the third 30-min period (3rd 30′), PMN migration into the lower chamber decreased to 14.6 ± 2.9% for no antibody and 16.8 ± 5.2% for J3F.1. In subsequent time periods, PMN migration across the monolayers continued to decrease to <10% in the fourth 30 min and to <5% in the fifth and sixth 30-min periods, respectively. From Fig. 2 A, total migration of PMN into the lower chamber can be plotted as a function of migration time, which is shown in Fig. 2 B. As can be seen, under non-in
What problem does this paper attempt to address?
-
Soluble CD40 Ligand Stimulates CD40-Dependent Activation of the Β2 Integrin Mac-1 and Protein Kinase C Zeda (pkcζ) in Neutrophils: Implications for Neutrophil-Platelet Interactions and Neutrophil Oxidative Burst
Rong Jin,Shiyong Yu,Zifang Song,Xiaolei Zhu,Cuiping Wang,Jinchuan Yan,Fusheng Wu,Anil Nanda,D. Neil Granger,Guohong Li
DOI: https://doi.org/10.1371/journal.pone.0064631
IF: 3.7
2013-01-01
PLoS ONE
Abstract:Recent work has revealed an essential involvement of soluble CD40L (sCD40L) in inflammation and vascular disease. Activated platelets are the major source of sCD40L, which has been implicated in platelet and leukocyte activation, although its exact functional impact on leukocyte-platelet interactions and the underlying mechanisms remain undefined. We aimed to determine the impact and the mechanisms of sCD40L on neutrophils. We studied neutrophil interactions with activated, surface-adherent platelets as a model for leukocyte recruitment to the sites of injury. Our data show that CD40L contributes to neutrophil firm adhesion to and transmigration across activated surface-adherent platelets, possibly through two potential mechanisms. One involves the direct interaction of ligand-receptor (CD40L-CD40), i.e., platelet surface CD40L interaction with neutrophil CD40; another involves an indirect mechanism, i.e. soluble CD40L stimulates activation of the leukocyte-specific β2 integrin Mac-1 in neutrophils and thereby further promotes neutrophil adhesion and migration. Activation of the integrin Mac-1 is known to be critical for mediating neutrophil adhesion and migration. sCD40L activated Mac-1 in neutrophils and enhanced neutrophil-platelet interactions in wild-type neutrophils, but failed to elicit such responses in CD40-deficient neutrophils. Furthermore, our data show that the protein kinase C zeta (PKCζ) is critically required for sCD40L-induced Mac-1 activation and neutrophil adhesive function. sCD40L strongly stimulated the focal clustering of Mac-1 (CD11b) and the colocalization of Mac-1 with PKCζ in wild-type neutrophils, but had minimal effect in CD40-deficient neutrophils. Blocking PKCζ completely inhibited sCD40L-induced neutrophil firm adhesion. Moreover, sCD40L strongly stimulates neutrophil oxidative burst via CD40-dependent activation of PI3K/NF-KB, but independent of Mac-1 and PKCζ. These findings may contribute to a better understanding of the underlying mechanisms by which sCD40L/CD40 pathway contributes to inflammation and vascular diseases.
-
CD47 (Cluster of Differentiation 47)
Sukhbir Kaur,Jeffrey S Isenberg,David D Roberts
Abstract:CD47, also known as integrin-associated protein, is a constitutively and ubiquitously expressed transmembrane receptor. CD47 is conserved across amniotes including mammals, reptiles, and birds. Expression is increased in many cancers and, in non-malignant cells, by stress and with aging. The up-regulation of CD47 expression is generally epigenetic, whereas gene amplification occurs with low frequency in some cancers. CD47 is a high affinity signaling receptor for the secreted protein thrombospondin-1 (THBS1) and the counter-receptor for signal regulatory protein-α (SIRPA, SIRPα) and SIRPγ (SIRPG). CD47 interaction with SIRPα serves as a marker of self to innate immune cells and thereby protects cancer cells from phagocytic clearance. Consequently, higher CD47 correlates with a poor prognosis in some cancers, and therapeutic blockade can suppress tumor growth by enhancing innate antitumor immunity. CD47 expressed on cytotoxic T cells, dendritic cells, and NK cells mediates inhibitory THBS1 signaling that further limits antitumor immunity. CD47 laterally associates with several integrins and thereby regulates cell adhesion and migration. CD47 has additional lateral binding partners in specific cell types, and ligation of CD47 in some cases modulates their function. THBS1-CD47 signaling in non-malignant cells inhibits nitric oxide/cGMP, calcium, and VEGF signaling, mitochondrial homeostasis, stem cell maintenance, protective autophagy, and DNA damage response, and promotes NADPH oxidase activity. CD47 signaling is a physiological regulator of platelet activation, angiogenesis and blood flow. THBS1/CD47 signaling is frequently dysregulated in chronic diseases.
-
CD11B/CD18 SIALYLATION REGULATES NEUTROPHIL TRAFFICKING AND INFLAMMATORY FUNCTION IN THE INTESTINE
Jennifer Brazil,Veronica Azcutia,Matthias Kelm,Richard Cummings,Charles Parkos
DOI: https://doi.org/10.1093/ibd/izae020.138
2024-01-25
Inflammatory Bowel Diseases
Abstract:Abstract Pathogen-triggered neutrophil (PMN) recruitment is critical for innate immunity, but excessive PMN influx and associated mucosal tissue damage is implicated in the pathogenesis of numerous human inflammatory disorders including inflammatory bowel disease (IBD). Critically in both ulcerative colitis and Crohn’s disease, disease flares requiring medical intervention are associated with migration of large numbers of PMNs across colonic epithelium resulting in mucosal injury/ulceration. Recent studies have shown that specific PMN glycans can be targeted to reduce transepithelial migration (TEpM) the critical final step in PMN intestinal trafficking. However the role of Sialic acid (Sia), the most abundant PMN expressed terminal glycan, in regulating PMN migration and inflammatory function in the gut is not well understood. Using sophisticated models of human and murine PMN intestinal migration, we show that blocking sialidase-mediated removal of alpha 2-3 linked sialic acid from CD11b/CD18 inhibits PMN TEpM in vitro and in vivo.Measurement of cytochrome C reduction revealed that inhibition of sialidases reduced bacterial peptide (fMLF) stimulated superoxide release by human and murine PMN. Flow cytometry analyses revealed that sialidase inhibition decreased fMLF mediated PMN degranulation and fMLF mediated CD11b/CD18 conformational activation. Highlighting the functional importance of individual Sia residues, inhibition of alpha 2-3 sialidase activity with 3’ Sialyllactose (3’SL) was found to inhibit PMN intestinal trafficking in vitro and in vivo and reduce PMN degranulation responses. Immunoblotting studies showed that sialidase inhibition deactivates spleen tyrosine kinase (Syk) and p38 MAP kinase, key intracellular mediators that signal downstream of PMN CD11b/CD18. Finally, inhibition of sialidase activity reduced adhesion of PMN to ICAM-1 (a major physiological substrate of CD11b/CD18) demonstrating that endogenous PMN sialidases regulate binding interactions between migrating PMN and intestinal epithelial receptors. Taken together data show that upon activation PMN sialidases remove alpha 2-3 Sia from CD11b/CD18 which leads to integrin activation and subsequent outside in signaling mediated by Syk to drive PMN inflammatory effector functions including migration, degranulation, and superoxide release. As such targeting CD11b/CD18 sialylation represents a promising new therapeutic strategy for reducing dysregulated PMN influx and associated mucosal tissue damage in IBD and other inflammatory disorders.
gastroenterology & hepatology
-
The Heparan Sulfate Proteoglycan Form of Epithelial CD44v3 Serves as a CD11b/CD18 Counter-receptor during Polymorphonuclear Leukocyte Transepithelial Migration
Ke Zen,Dan-Qing Liu,Li-Min Li,Celia X-J Chen,Ya-Lan Guo,Bihn Ha,Xi Chen,Chen-Yu Zhang,Yuan Liu
DOI: https://doi.org/10.1074/jbc.M807805200
IF: 5.485
2009-01-01
Journal of Biological Chemistry
Abstract:Leukocyte beta(2)-integrin CD11b/CD18 mediates the firm adhesion and subsequent transepithelial migration of polymorphonuclear leukocytes, but the identity of its counter-receptor(s) on epithelia remains elusive. Here we identified a monoclonal antibody, clone C3H7, which strongly bound to the basolateral membranes of epithelial cells and inhibited both the adhesion of epithelial cells to immobilized CD11b/CD8 and the transepithelial migration of PMNs in a physiologically relevant basolateral-to-apical direction. C3H7 antigen expression in epithelial monolayers was significantly increased by treatment with pro-inflammatory cytokine interferon-gamma or a combination of interferon-gamma and tumor necrosis factor-alpha. Up-regulation of C3H7 antigen was also observed in the epithelium of inflamed human colon tissues. Microsequencing and Western blotting of the purified antigen showed it to be CD44 variant 3 (CD44v3), a similar to 160-kDa membrane glycoprotein. Further studies demonstrated that this epithelial CD44v3 specifically binds to CD11b/CD18 through its heparan sulfate moieties. In summary, our study demonstrates for the first time that the heparan sulfate proteoglycan form of epithelial CD44v3 plays a critical role in facilitating PMN recruitment during inflammatory episodes via directly binding to CD11b/CD18.
-
Cleavage of the CD11b Extracellular Domain by the Leukocyte Serprocidins is Critical for Neutrophil Detachment During Chemotaxis (173.7)
Ke Zen,Yalan Guo,Zhen Bian,Yuan Liu
DOI: https://doi.org/10.4049/jimmunol.188.supp.173.7
2012-01-01
The Journal of Immunology
Abstract:Abstract The β(2)-integrin CD11b/CD18 mediates the firm adhesion of neutrophils (PMNs) to epithelial monolayers, a key step in PMN transepithelial migration. To complete the transmigration process, adherent PMNs must detach from epithelial surfaces to move forward. The mechanism that governs the detachment of adherent PMNs, however, is not clear. Here, we present evidence that cleavage of the CD11b extracellular domain containing the ligand-binding I-domain by 3 structural and functional related serine proteases (elastase, proteinase-3 and cathepsin G) serves as a novel mechanism for PMN detachment after the initial cell adhesion. Kinetic studies showed that the cleavage of CD11b is positively correlated with PMN detachment and subsequent transmigration. Moreover, the results demonstrated that elastase, proteinase-3 and cathepsin G all cleaved the purified, functionally active form of CD11b in a pattern similar to the CD11b shedding that occurs during PMN transmigration. Their cleavage sites on purified CD11b were located at (761)Thr-Ala(762) (elastase/proteinase-3) and (760)Phe-Thr(761) (cathepsin G), respectively. CD11b cleavage and PMN detachment and chemotaxis, were impaired in elastase/cathepsin G-deficient Beige mice; this defect could be restored by the addition of extracellular elastase. By illustrating CD11b shedding by elastase, proteinase-3 and cathepsin G as a novel mechanism for PMN detachment, our study provides novel therapeutic targets for controlling inflammation.
-
CD47 Masks Pro-Phagocytic Ligands in Cis on Tumor Cells to Suppress Antitumor Immunity
Zhenghai Tang,Ming-Chao Zhong,Jin Qian,Cristian Camilo Galindo,Dominique Davidson,Jiaxin Li,Yunlong Zhao,Enfu Hui,André Veillette
DOI: https://doi.org/10.1038/s41590-023-01671-2
IF: 30.5
2023-01-01
Nature Immunology
Abstract:Cancer cells often overexpress CD47, which triggers the inhibitory receptor SIRPα expressed on macrophages, to elude phagocytosis and antitumor immunity. Pharmacological blockade of CD47 or SIRPα is showing promise as anticancer therapy, although CD47 blockade has been associated with hematological toxicities that may reflect its broad expression pattern on normal cells. Here we found that, in addition to triggering SIRPα, CD47 suppressed phagocytosis by a SIRPα-independent mechanism. This mechanism prevented phagocytosis initiated by the pro-phagocytic ligand, SLAMF7, on tumor cells, due to a cis interaction between CD47 and SLAMF7. The CD47–SLAMF7 interaction was disrupted by CD47 blockade and by a first-in-class agonist SLAMF7 antibody, but not by SIRPα blockade, thereby promoting antitumor immunity. Hence, CD47 suppresses phagocytosis not only by engaging SIRPα, but also by masking cell-intrinsic pro-phagocytic ligands on tumor cells and knowledge of this mechanism may influence the decision between CD47 blockade or SIRPα blockade for therapeutic purposes.
-
Prognostic risk stratification derived from individual patient level data for men with advanced penile squamous cell carcinoma receiving first-line systemic therapy.
G. Pond,G. Di Lorenzo,A. Necchi,B. Eigl,M. Kolinsky,R. Chacko,T. Dorff,L. Harshman,M. Milowsky,R. J. Lee,M. Galsky,P. Federico,G. Bolger,M. Deshazo,A. Mehta
DOI: https://doi.org/10.1016/j.urolonc.2013.10.007
2014-05-01
Abstract:
-
Cell autonomous functions of CD47 in regulating cellular plasticity and metabolic plasticity
Ruhi Polara,Raja Ganesan,Stuart M Pitson,Nirmal Robinson
DOI: https://doi.org/10.1038/s41418-024-01347-w
Abstract:CD47 is a ubiquitously expressed cell surface receptor, which is widely known for preventing macrophage-mediated phagocytosis by interacting with signal regulatory protein α (SIRPα) on the surface of macrophages. In addition to its role in phagocytosis, emerging studies have reported numerous noncanonical functions of CD47 that include regulation of various cellular processes such as proliferation, migration, apoptosis, differentiation, stress responses, and metabolism. Despite lacking an extensive cytoplasmic signaling domain, CD47 binds to several cytoplasmic proteins, particularly upon engaging with its secreted matricellular ligand, thrombospondin 1. Indeed, the regulatory functions of CD47 are greatly influenced by its interacting partners. These interactions are often cell- and context-specific, adding a further level of complexity. This review addresses the downstream cell-intrinsic signaling pathways regulated by CD47 in various cell types and environments. Some of the key pathways modulated by this receptor include the PI3K/AKT, MAPK/ERK, and nitric oxide signaling pathways, as well as those implicated in glucose, lipid, and mitochondrial metabolism. These pathways play vital roles in maintaining tissue homeostasis, highlighting the importance of understanding the phagocytosis-independent functions of CD47. Given that CD47 expression is dysregulated in a variety of cancers, improving our understanding of the cell-intrinsic signals regulated by this molecule will help advance the development of CD47-targeted therapies.
-
Reversible encephalopathy and axonal neuropathy in Parkinson's disease during duodopa therapy
Helen Ling,Kanjana Unnwongse,R. Bhidayasiri
DOI: https://doi.org/10.1002/mds.22807
IF: 9.698
2009-11-15
Movement Disorders
Abstract:eep brain stimulation in acute management of status Deep brain
stimulation in acute management of status dystonicus.
-
How Adhesion Molecule Patterns Change While Neutrophils Traffic through the Lung during Inflammation
Franziska M. Konrad,Julia Wohlert,Jutta Gamper-Tsigaras,Kristian-Christos Ngamsri,Jörg Reutershan
DOI: https://doi.org/10.1155/2019/1208086
2019-02-28
Mediators of Inflammation
Abstract:In acute pulmonary inflammation, polymorphonuclear cells (PMNs) pass a transendothelial barrier from the circulation into the lung interstitium followed by a transepithelial migration into the alveolar space. These migration steps are regulated differentially by a concept of adhesion molecules and remain—despite decades of research—incompletely understood. Current knowledge of changes in the expression pattern of adhesion molecules mainly derives from in vitro studies or from studies in extrapulmonary organ systems, where regulation of adhesion molecules differs significantly. In a murine model of lung inflammation, we determined the expression pattern of nine relevant neutrophilic adhesion molecules on their way through the different compartments of the lung. We used a flow cytometry-based technique that allowed describing spatial distribution of the adhesion molecules expressed on PMNs during their migration through the lung in detail. For example, the highest expression of CD29 was found in the intravascular compartment, highlighting its impact on the initial adhesion to the endothelium. CD47 showed its peak of expression on the later phase of transendothelial migration, whereas CD11b and CD54 expression peaked interstitial. A pivotal role for transepithelial migration was found for the adhesion molecule CD172a. Thereby, expression may correlate with functional impact for specific migration steps. In vitro studies further confirmed our in vivo findings. In conclusion, we are the first to determine the changes in expression patterns of relevant adhesion molecules on their migration through the different compartments of the lung. These findings may help to further understand the regulation of neutrophil trafficking in the lung.
immunology,cell biology
-
Retinal hamartoma, acquired retinoschisis, and retinal hole.
A. Schachat,B. Glaser
DOI: https://doi.org/10.1016/S0002-9394(14)77975-9
1985-05-01
Abstract:
-
Neutrophils exhibit distinct migration phenotypes that are regulated by transendothelial migration
Amy B. Schwartz,Adithan Kadasamy,Juan C. del Alamo,Yi-Ting Yeh
DOI: https://doi.org/10.1101/2024.10.17.618860
2024-12-07
Abstract:The extravasation of polymorphonuclear neutrophils (PMNs) is a critical component of the innate immune response that involves transendothelial migration (TEM) and interstitial migration. TEM-mediated interactions between PMNs and vascular endothelial cells (VECs) trigger a cascade of biochemical and mechanobiological signals whose effects on interstitial migration are currently unclear. To address this question, we cultured human VECs on a fibronectin-treated transwell insert to model the endothelium and basement membrane, loaded PMN-like differentiated HL60 (dHL-60) cells in the upper chamber of the insert, and collected the PMNs that crossed the membrane-supported monolayer from the lower chamber. The 3D chemotactic migration of the TEM-conditioned PMNs through collagen matrices was then quantified. Data collected from over 50,000 trajectories showed two distinct migratory phenotypes, i.e., a high-persistence phenotype and a low-persistence phenotype. These phenotypes were conserved across treatment conditions, and their existence was confirmed in human primary PMNs. The high-persistence phenotype was characterized by more straight trajectories and faster migration speeds, whereas the low-persistence one exhibited more frequent sharp turns and loitering periods. A key finding of our study is that TEM induced a phenotypic shift in PMNs from high-persistence migration to low-persistence migration. Changes in the relative proportion of high-persistence and low-persistence populations correlated with GRK2 expression levels. Inhibiting GRK2 hindered the TEM-induced shift in migratory phenotype and impaired the phagocytic function of PMNs. Overall, our study suggests that TEM-mediated GRK2 signaling primes PMNs for a migration phenotype better suited for spatial exploration and inflammation resolution. These observations provide novel insight into the biophysical impacts of TEM that priming PMNs is essential to conduct sentinel functions.
Cell Biology
-
Structural–functional diversity of CD47 proteoforms
Ting Zhang,Feng Wang,Lu Xu,Yong-Guang Yang
DOI: https://doi.org/10.3389/fimmu.2024.1329562
IF: 7.3
2024-02-15
Frontiers in Immunology
Abstract:The ubiquitously expressed transmembrane glycoprotein CD47 participates in various important physiological cell functions, including phagocytosis, apoptosis, proliferation, adhesion, and migration, through interactions with its ligands, including the inhibitory receptor signal regulatory protein α (SIRPα), secreted glycoprotein thrombospondin-1 (TSP-1), and integrins. Elevated expression of CD47 is observed in a wide range of cancer cells as a mechanism for evading the immune system, blocking the interaction between the CD47 and SIRPα is the most advanced and promising therapeutic approach currently investigated in multiple clinical trials. The widely held view that a single type of CD47 protein acts through membrane interactions has been challenged by the discovery of a large cohort of CD47 proteins with cell-, tissue-, and temporal-specific expression and functional profiles. These profiles have been derived from a single gene through alternative splicing and post-translational modifications, such as glycosylation, pyroglutamate modification, glycosaminoglycan modification, and proteolytic cleavage and, to some extent, via specific CD47 clustering in aging and tumor cells and the regulation of its subcellular localization by a pre-translational modification, alternative cleavage and polyadenylation (APA). This review explores the origins and molecular properties of CD47 proteoforms and their roles under physiological and pathological conditions, mentioning the new methods to improve the response to the therapeutic inhibition of CD47–SIRPα immune checkpoints, contributing to the understanding of CD47 proteoform diversity and identification of novel clinical targets and immune-related therapeutic candidates.
immunology
-
Natural Killer Cell Recruitment and Activation Are Regulated by CD47 Expression in the Tumor Microenvironment
Pulak Ranjan Nath,Dipasmita Pal-Nath,Ajeet Mandal,Margaret C Cam,Anthony L Schwartz,David D Roberts,Margaret C. Cam
DOI: https://doi.org/10.1158/2326-6066.CIR-18-0367
IF: 10.1
2019-11-25
Cancer Immunology Research
Abstract:Elevated CD47 expression in some cancers is associated with decreased survival and limited clearance by phagocytes expressing the CD47 counterreceptor SIRPα. In contrast, elevated CD47 mRNA expression in human melanomas was associated with improved survival. Gene-expression data were analyzed to determine a potential mechanism for this apparent protective function and suggested that high CD47 expression increases recruitment of natural killer (NK) cells into the tumor microenvironment. The CD47 ligand thrombospondin-1 inhibited NK cell proliferation and CD69 expression in vitro . Cd47 –/– NK cells correspondingly displayed augmented effector phenotypes, indicating an inhibitory function of CD47 on NK cells. Treating human NK cells with a CD47 antibody that blocks thrombospondin-1 binding abrogated its inhibitory effect on NK cell proliferation. Similarly, treating wild-type mice with a CD47 antibody that blocks thrombospondin-1 binding delayed B16 melanoma growth, associating with increased NK cell recruitment and increased granzyme B and interferon- levels in intratumoral NK but not CD8 + T cells. However, B16 melanomas grew faster in Cd47 –/– than in wild-type mice. Melanoma-bearing Cd47 –/– mice exhibited decreased splenic NK cell numbers, with impaired effector protein expression and elevated exhaustion markers. Proapoptotic gene expression in Cd47 –/– NK cells was associated with stress-mediated increases in mitochondrial proton leak, reactive oxygen species, and apoptosis. Global gene-expression profiling in NK cells from tumor-bearing mice identified CD47-dependent transcriptional responses that regulate systemic NK activation and exhaustion. Therefore, CD47 positively and negatively regulates NK cell function, and therapeutic antibodies that block inhibitory CD47 signaling can enhance NK immune surveillance of melanomas.
oncology,immunology
-
Neutrophil CD16b Crosslinking Induces Lipid Raft-Mediated Activation of SHP-2 and Affects Cytokine Expression and Retarded Neutrophil Apoptosis
H. Yang,H. Jiang,Y. Song,D. J. Chen,X. J. Shen,J. H. Chen
DOI: https://doi.org/10.1016/j.yexcr.2017.11.009
IF: 4.145
2017-01-01
Experimental Cell Research
Abstract:Two different types of FcRs for IgG are constitutively expressed on the surface of human neutrophils, namely, FcγRIIA (CD32a) and FcγRIIIB (CD16b). Unlike FcγRIIA, FcγRIIIb is GPI anchored to the cell membrane and its signal transduction is still ambiguous. To further understand the signal transduction of CD16b, we compared neutrophil cytokine expression and apoptosis by the cross-linking of CD32a and CD16b respectively. We found that both CD32a and CD16b crosslinking can activate neutrophils, but did not exactly share cytokine expression profiles. On the other hand, CD16b cross-linking retarded neutrophil apoptosis while CD32a promoted it. By interrupting the lipid raft with methyl-β-cyclodextrin (MβCD) and inhibiting the ITAM-SYK pathway with an SYK inhibitor (piceatannol), we found reduced apoptosis was at least partially mediated by lipid raft structure, but not the ITAM-SYK pathway. Additionally, CD16b but not CD32a cross-linking triggered SHP-2 phosphorylation and led to its translocation into lipid rafts. SHP-2 phosphorylation and translocation were inhibited by MβCD. Moreover, pre-inhibition of SHP-2 by a specific inhibitor (SHP099) converted IL-10 and SOCS3 expression level and promoted neutrophil apoptosis after CD16b crosslinking. In conclusion, these results, for the first time, collectively indicate that SHP-2 is activated by CD16b crosslinking in neutrophils and functions as a component of the raft-mediated signaling pathway.
-
Naming speed as a clinical marker in predicting basic calculation skills in children with specific language impairment.
Tijs Kleemans,E. Segers,L. Verhoeven
DOI: https://doi.org/10.1016/j.ridd.2011.12.007
IF: 3
2012-05-01
Research in Developmental Disabilities
Abstract:
-
Tolerating CD47
Jeffrey S. Isenberg,Enrique Montero
DOI: https://doi.org/10.1002/ctm2.1584
IF: 8.554
2024-02-17
Clinical and Translational Medicine
Abstract:Connecting the dots on CD47 1. CD47 interacts with ligands along with known and to‐be‐determined (TBD) receptors in cis and trans. 2. CD47 is the center piece of a pleiotropic receptor‐multiligand interaction network. 3. CD47‐targeted interventions may disrupt homeostatic connectivity and have on‐target effects. 4. CD47 immunomodulation to improve health requires connecting the missing dots. Cluster of differentiation 47 (CD47) occupies the outer membrane of human cells, where it binds to soluble and cell surface receptors on the same and other cells, sculpting their topography and resulting in a pleiotropic receptor‐multiligand interaction network. It is a focus of drug development to temper and accentuate CD47‐driven immune cell liaisons, although consideration of on‐target CD47 effects remain neglected. And yet, a late clinical trial of a CD47‐blocking antibody was discontinued, existent trials were restrained, and development of CD47‐targeting agents halted by some pharmaceutical companies. At this point, if CD47 can be exploited for clinical advantage remains to be determined. Herein an airing is made of the seemingly conflicting actions of CD47 that reflect its position as a junction connecting receptors and signalling pathways that impact numerous human cell types. Prospects of CD47 boosting and blocking are considered along with potential therapeutic implications for autoimmune diseases and cancer.
oncology,medicine, research & experimental
-
Mechanistic Insights into Macrophages Regulation of Neutrophil Transendothelial Migration in Inflamed Mucosa
Xingsheng Ren,Jessica M. Urbanczyk,Ronen Sumagin
DOI: https://doi.org/10.1096/fasebj.2022.36.s1.r2090
2022-05-01
The FASEB Journal
Abstract:Rapid neutrophil (PMN) mobilization to sites of insult is critical for host defense and requires crossing of the vascular wall. PMN transendothelial migration (TEM) involves several well‐studied sequential adhesive interactions with vascular ECs, however what initiates or terminates this process is not well‐understood. Our findings identified a new mechanism where gut interstitial macrophages, which are rapidly recruited towards the vascular wall in response to inflammatory cues, were found to locally prime endothelial cells (ECs) responses to regulate PMN TEM. Using real‐time intravital microscopy (IVM) on lipopolysaccharides (LPS)‐inflamed intestines in anesthetized CX3CR1‐EGFP macrophage‐reporter mice, complemented by whole‐mount tissue imaging we demonstrate that macrophage presence was critical for the induction of PMN‐ECs adhesive interactions and subsequent PMN recruitment and accumulation in the intestinal mucosa. Anti CSFR‐1 antibody‐based macrophage depletion in the lamina propria and at the vessel wall significantly reduced PMN adhesion and TEM in inflamed intestines. We further observed that macrophages at the vessel wall localized specifically to regions of high ICAM‐1 intensity and their removal resulted in elimination of the ICAM‐1 “hot spots”, overall lowering the ECs ICAM‐1 expression. Mechanistically, using murine/human ECs‐macrophage and PMN co‐cultures we established that activated macrophages elevate PMN adhesion and TEM via TNFα‐dependent upregulation of ECs ICAM‐1. Antibody‐mediated neutralization of TNFα in macrophage co‐cultures with ECs and/or PMNs suppressed ICAM‐1 upregulation, decreasing PMN TEM. Further in vivo imaging studies of inflamed gut revealed high TNFα expression in macrophages and specific expression of TNFa receptor type II (TNFR II) but not type I in intestinal ECs. The use of bone marrow chimeras with TNFα knockout macrophages further confirmed the novel role of macrophages‐TNFα in regulating ECs adhesion molecule expression and PMN TEM. As such, our findings identify new, clinically relevant mechanism by which macrophages regulate PMN trafficking in inflamed mucosa.
cell biology,biochemistry & molecular biology,biology
-
CD31 signaling promotes the detachment at the uropod of extravasating neutrophils allowing their migration to sites of inflammation
Francesco Andreata,Marc Clément,Robert A Benson,Juliette Hadchouel,Emanuele Procopio,Guillaume Even,Julie Vorbe,Samira Benadda,Véronique Ollivier,Benoit Ho-Tin-Noe,Marie Le Borgne,Pasquale Maffia,Antonino Nicoletti,Giuseppina Caligiuri
DOI: https://doi.org/10.7554/eLife.84752
IF: 7.7
2023-08-07
eLife
Abstract:Effective neutrophil migration to sites of inflammation is crucial for host immunity. A coordinated cascade of steps allows intravascular leukocytes to counteract the shear stress, transmigrate through the endothelial layer, and move toward the extravascular, static environment. Those events are tightly orchestrated by integrins, but, while the molecular mechanisms leading to their activation have been characterized, the regulatory pathways promoting their detachment remain elusive. In light of this, it has long been known that platelet-endothelial cell adhesion molecule (Pecam1, also known as CD31) deficiency blocks leukocyte transmigration at the level of the outer vessel wall, yet the associated cellular defects are controversial. In this study, we combined an unbiased proteomic study with in vitro and in vivo single-cell tracking in mice to study the dynamics and role of CD31 during neutrophil migration. We found that CD31 localizes to the uropod of migrating neutrophils along with closed β2-integrin and is required for essential neutrophil actin/integrin polarization. Accordingly, the uropod of Pecam1-/- neutrophils is unable to detach from the extracellular matrix, while antagonizing integrin binding to extracellular matrix components rescues this in vivo migratory defect. Conversely, we showed that sustaining CD31 co-signaling actively favors uropod detachment and effective migration of extravasated neutrophils to sites of inflammation in vivo. Altogether, our results suggest that CD31 acts as a molecular rheostat controlling integrin-mediated adhesion at the uropod of egressed neutrophils, thereby triggering their detachment from the outer vessel wall to reach the inflammatory sites.
-
Human CD99L2 Regulates a Unique Step in Leukocyte Transmigration
Nakisha S Rutledge,Faith T Ogungbe,Richard L Watson,David P Sullivan,William A Muller
DOI: https://doi.org/10.4049/jimmunol.2101091
2022-09-01
Abstract:CD99-like 2 (CD99L2 [L2]) is a highly glycosylated 52-kDa type 1 membrane protein that is important for leukocyte transendothelial migration (TEM) in mice. Inhibiting L2 using function-blocking Ab significantly reduces the recruitment of leukocytes to sites of inflammation in vivo. Similarly, L2 knockout mice have an inherent defect in leukocyte transmigration into sites of inflammation. However, the role of L2 in inflammation has only been studied in mice. Furthermore, the mechanism by which it regulates TEM is not known. To study the relevance to human inflammation, we studied the role of L2 on primary human cells in vitro. Our data show that like PECAM and CD99, human L2 is constitutively expressed at the borders of endothelial cells and on the surface of leukocytes. Inhibiting L2 using Ab blockade or genetic knockdown significantly reduces transmigration of human neutrophils and monocytes across endothelial cells. Furthermore, our data also show that L2 regulates a specific, sequential step of TEM between PECAM and CD99, rather than operating in parallel or redundantly with these molecules. Similar to PECAM and CD99, L2 promotes transmigration by recruiting the lateral border recycling compartment to sites of TEM, specifically downstream of PECAM initiation. Collectively, our data identify a novel functional role for human L2 in TEM and elucidate a mechanism that is distinct from PECAM and CD99.