Heterotypic interactions enabled by polarized neutrophil microdomains mediate thromboinflammatory injury

Andrés Hidalgo, Jungshan Chang, Jung-Eun Jang, Anna J Peired, Elaine Y Chiang, Paul S Frenette, Andrés Hidalgo, Jungshan Chang, Jung-Eun Jang, Anna J Peired, Elaine Y Chiang, Paul S Frenette

Abstract

Selectins and their ligands mediate leukocyte rolling, allowing interactions with chemokines that lead to integrin activation and arrest. Here we show that E-selectin is crucial for generating a secondary wave of activating signals, transduced specifically by E-selectin ligand-1, that induces polarized, activated alpha(M)beta(2) integrin clusters at the leading edge of crawling neutrophils, allowing capture of circulating erythrocytes or platelets. In a humanized mouse model of sickle cell disease, the capture of erythrocytes by alpha(M)beta(2) microdomains leads to acute lethal vascular occlusions. In a model of transfusion-related acute lung injury, polarized neutrophils capture circulating platelets, resulting in the generation of oxidative species that produce vascular damage and lung injury. Inactivation of E-selectin or alpha(M)beta(2) prevents tissue injury in both inflammatory models, suggesting broad implications of this paradigm in thromboinflammatory diseases. These results indicate that endothelial selectins can influence neutrophil behavior beyond its canonical rolling step through delayed, organ-damaging, polarized activation.

Figures

Figure 1. Heterotypic interactions of RBC and…
Figure 1. Heterotypic interactions of RBC and platelets with leukocyte microdomains are induced during inflammation
(a) Heterotypic interactions between leukocytes and platelets in a TNF-α- stimulated C57BL/6 mouse. Fluorescently conjugated antibodies to L-selectin (blue) and CD41 (red) allow identification of the trailing edge of crawling leukocytes and platelets, respectively. The short arrow indicates an interacting RBC, and the asterisks show interacting platelets (green for those mediated by the trailing edge, white for the leading edge). The large arrow points to the direction of blood flow. Scale bar = 10 µm. (b) Frequency of interactions between RBCs and leukocytes in venules of mice where surgical trauma or trauma plus TNF-α administration preceded imaging. n = 5 mice. ***, p<0.001, Mann-Whitney test. (c) Frequency of interactions between leukocytes and platelets. n=5–6 mice. ***, p<0.001, Mann-Whitney test. (d) Contribution of the leading and trailing edges in RBC interactions after TNF-α treatment. n = 4 mice; ‡, p<0.005. (e) Contribution of leukocyte microdomains to platelet interactions in wild-type mice, treated or not with TNF-α. n = 5 mice per group; ‡, p<0.005 compared to Trailing group, paired t-test; NS, not significant.
Figure 2. RBC and platelet interactions depend…
Figure 2. RBC and platelet interactions depend on E-selectin and its ligand ESL-1
(a) Number of RBC captures per adherent leukocyte in wild–type, Selp−/− and Sele−/− mice. n = 32–40 venules from 5–7 mice per group. *, p<0.05 compared to WT, **, p<0.01 compared to the other groups; Kruskal-Wallis test with Dunn’s multigroup comparison. (b) Reduced number of platelet captures per adherent leukocyte in Sele−/− mice. n = 32–42 venules from at least 5 mice per group. ***, p<0.0001. (c) Contribution of leukocyte microdomains to platelet interactions in TNF-α-treated mice. n = 5 mice per group. **, p<0.01, Mann-Whitney test. ‡, p=0.003 and NS, not significant compared to Trailing groups, paired t-test. (d) RBC-leukocyte interactions in wild-type, Selplg−/− and Cd44−/− mice. n = 31–42 venules from 5–7 mice per group. *, p<0.05, Kruskal-Wallis test with Dunn’s multigroup comparison. (e) RBC captures by leukocytes transduced with control (Scrmbl) or shESL-1 lentiviral vectors. n = 4–6 mice per group. ***, p<0.0001 compared to GFP− cells from the same lentiviral group, paired t-test. (f) nRBC interactions in mice pretreated with inhibitors to Src kinases (PP2), p38 MAPK (SB203580) or Syk (Piceatannol), or vehicle control (DMSO). n = 33–44 venules from 6 mice per group. *, p<0.05 compared to all other groups, Kruskal-Wallis test with Dunn’s multigroup comparison.
Figure 3. Heterotypic interactions with RBC and…
Figure 3. Heterotypic interactions with RBC and platelets are mediated by the leukocyte integrin αMβ2
(a) Micrographs of adherent leukocytes obtained by MFIM analyses of TNF-α-treated C57BL/6 mice injected with labeled antibodies against L-selectin (red) and anti-αM (green). αMβ2 integrin expression is homogenously distributed, including where an RBC (arrowhead) is shown interacting at the leading edge of an adherent leukocyte. Scale bar = 10 µm. (b) RBC-WBC interactions in Itgam−/− mice compared to wild-type (WT) controls. n = 41–42 venules from 6–7 mice per group. **, p<0.001, Mann-Whitney test. (c) Platelet–WBC interactions in Itgam−/− mice compared to WT. n = 35–36 venules from 5–6 mice per group. **, p<0.0001, Mann-Whitney test. (d) Contribution of leukocyte microdomains to platelet captures in TNF-α-treated Itgam−/− mice. n = 5 mice. ‡, p=0.008, paired t-test compared to Trailing values.
Figure 4. E-selectin and ESL-1 modulate regional…
Figure 4. E-selectin and ESL-1 modulate regional αMβ2 activity on adherent leukocytes in vivo
(a) Frequency of fluosphere (beads) binding by F4/80− and F4/80+ adherent leukocytes. Data obtained from analyses of 84 fluosphere captures from 4 mice. ***, p<0.001, t-test. (b) Time-lapse micrographs of in vivo fluosphere capture by two representative adherent leukocytes (asterisks). Leading edges are outlined with dotted lines and L-selectin clusters are in red. Sequence of capture of fluospheres at the times indicated. A movie depicting capturing events is shown in Supplementary Video 3. The histogram (right panel) shows the quantitation of the fluospheres captured by the leading and trailing edges from 98 events identified in 7 mice; ‡, p=0.0005. Scale bar = 10 µm. (c) Binding of albumin-coated fluospheres to leukocytes in wild-type and Itgam−/− mice. Each dot represents the average number of fluospheres bound per leukocyte in individual venules. n = 40 venules from 4–5 mice per group. **, p<0.001, Mann-Whitney test. (d) Binding of albumin-coated fluospheres to leukocytes in wild-type, Selp−/− and Sele−/− mice. n = 33–43 venules from 4–7 mice per group. *, p<0.05 compared to the other groups; Kruskal-Wallis test. (e) Binding of fluospheres in wild-type, Selplg−/− and Cd44−/− mice. n = 30–47 venules from 4–6 mice per group. (f) Fluosphere capture in leukocytes transduced with shESL-1 or scrambled sequence lentiviral vectors. n = 5 mice per group. ***, p<0.0001 compared to GFP− cells from the same lentiviral group, paired t-test.
Figure 5. Antibody-induced lung injury requires platelet-leukocyte…
Figure 5. Antibody-induced lung injury requires platelet-leukocyte interactions and is blocked by antibodies to E-selectin and αMβ2
(a) Protein accumulation in BAL fluids after anti-H2b or H2d antibody administration. αPlt, platelet-depleted mice; n = 7–9 mice. **, p<0.01. (b) Platelet counts from mice in (a). n = 9–10 mice; ***, p<0.001. (c) Left panels are representative micrographs of platelet-leukocyte interactions before (pre-H2d) and after (post-H2d) anti-H2d administration (L-selectin, blue; CD41, red). Arrow, direction of flow. Scale bar = 10 µm. Right panel, frequency of platelet-leukocyte interactions in control or anti-E-selectin-treated (9A9) mice. §, p<0.01 and NS, not significant, paired t-test. #, p<0.05, unpaired t-test. (d) Contribution of microdomains to interactions in the rIgG group. n = 5 mice; *, p=0.01. (e) Representative micrographs depicting vascular permeability after anti-H2d or anti-H2b treatment (FITC-dextran, blue). Scale bar = 100 µm. (f) FITC-dextran extravasation in mice treated with rat IgG, anti-E-selectin (9A9) or anti-αMβ2 (M1/70) antibodies. n = 20–32 venules. **, p<0.01; ***, p<0.001. (g) FITC-dextran extravasation in platelet-depleted (αPlt) or control mice after anti-H2d administration. n = 16–20 venules. §, p = 0.002. (h) Effect of P-selectin (RB40.34), E-selectin (9A9) and αMβ2 (M1/70) inhibition in lung injury. n = 7–13 mice. *, p<0.05; **, p<0.001. (i) Percentage of ROS-positive adherent leukocytes in control (Ctrl), platelet-depleted (αPlt) or anti-E-selectin antibody-treated (9A9) mice; n = 30–53 venules. *, p<0.05; ***, p<0.001 compared to all other groups. (j) Protein content in BAL fluids of mice treated with saline (Ctrl) or n-acetyl-cysteine (N-AcC). n = 5–14 mice. *, p < 0.05, unpaired t-test.
Figure 6. Vaso-occlusion in sickle cell disease…
Figure 6. Vaso-occlusion in sickle cell disease requires E-selectin-mediated activation of αMβ2
(a) Individual contributions of P- and E-selectins in sRBC capture by adherent leukocytes using genetically deficient mice or function-blocking antibodies. n = 9–17 mice. §, p< 0.01 compared to time 0–90 in the same group; *, p<0.05; **, p<0.01; ***, p<0.01 compared to the SCD group at the same time point; ¶, p<0.05 compared to the SCD Selp−/− group at the same time point; Kruskal-Wallis test. (b) Analysis of blood flow rates from the experiments shown in (a), at the 181–270 time point. SS, sickle cell; n = 9–16 mice; *, p < 0.01, one-way ANOVA. (c) Representative micrographs showing albumin-coated fluospheres bound to adherent leukocytes in hemizygous (SA) or homozygous (SS) SCD mice. Scale bar = 10 µm. (d) Binding of albumin-coated fluospheres to leukocytes in SA and SS chimeras with wild-type endothelium or Sele−/− endothelium. Each circle represents the average number of fluospheres bound per leukocyte for an individual venule, and bars represent mean values. n = 24–34 venules from 3–5 mice per group. **, p<0.001 compared to the two other groups; Kruskal-Wallis test. (e) Experimental scheme to assess the role of αMβ2 in vaso-occlusion. (f) αMβ2 blockade reduced the number of sRBC interactions with leukocytes. n = 6 mice per group; *, p = 0.001. (g) Kaplan-Meier survival curves after TNF-α treatment in control and M1/70 treated groups. n = 6 mice per group. Log-rank test, p< 0.002.

References

    1. Bullard DC, et al. Infectious susceptibility and severe deficiency of leukocyte rolling and recruitment in E-selectin and P-selectin double mutant mice. J Exp Med. 1996;183:2329–2336.
    1. Frenette PS, Mayadas TN, H R, Hynes RO, Wagner DD. Susceptibility to infection and altered hematopoiesis in mice deficient in both P-and E-selectins. Cell. 1996;84:563–574.
    1. Labow MA, et al. Characterization of E-selectin-deficient mice: Demonstration of overlapping function of the endothelial selectins. Immunity. 1994;1:709–720.
    1. Hidalgo A, Peired AJ, Wild MK, Vestweber D, Frenette PS. Complete identification of E-selectin ligands on neutrophils reveals distinct functions of PSGL-1, ESL-1, and CD44. Immunity. 2007;26:477–489.
    1. Zarbock A, Lowell CA, Ley K. Spleen tyrosine kinase Syk is necessary for E-selectin-induced alpha(L)beta(2) integrin-mediated rolling on intercellular adhesion molecule-1. Immunity. 2007;26:773–783.
    1. Steegmaier M, et al. The E-selectin-ligand ESL-1 is a variant of a receptor for fibroblast growth factor. Nature. 1995;373:615–620.
    1. Lo SK, et al. Endothelial-leukocyte adhesion molecule 1 stimulates the adhesive activity of leukocyte integrin CR3 (CD11b/CD18, Mac-1, alpha m beta 2) on human neutrophils. J Exp Med. 1991;173:1493–1500.
    1. Simon SI, Hu Y, Vestweber D, Smith CW. Neutrophil tethering on E-selectin activates beta 2 integrin binding to ICAM-1 through a mitogen-activated protein kinase signal transduction pathway. J Immunol. 2000;164:4348–4358.
    1. Gahmberg CG, Tolvanen M, Kotovuori P. Leukocyte adhesion--structure and function of human leukocyte beta2-integrins and their cellular ligands. Eur J Biochem. 1997;245:215–232.
    1. Beller DI, Springer TA, Schreiber RD. Anti-Mac-1 selectively inhibits the mouse and human type three complement receptor. J Exp Med. 1982;156:1000–1009.
    1. Simon DI, et al. Platelet glycoprotein ibalpha is a counterreceptor for the leukocyte integrin Mac-1 (CD11b/CD18) J Exp Med. 2000;192:193–204.
    1. Barreiro O, de la Fuente H, Mittelbrunn M, Sanchez-Madrid F. Functional insights on the polarized redistribution of leukocyte integrins and their ligands during leukocyte migration and immune interactions. Immunol Rev. 2007;218:147–164.
    1. Tohyama Y, et al. The critical cytoplasmic regions of the alphaL/beta2 integrin in Rap1-induced adhesion and migration. Mol Biol Cell. 2003;14:2570–2582.
    1. Ridley AJ, et al. Cell migration: integrating signals from front to back. Science. 2003;302:1704–1709.
    1. Abboud M, Laver J, Blau CA. Granulocytosis causing sickle-cell crisis. Lancet. 1998;351:959.
    1. Hirahashi J, et al. Mac-1 signaling via Src-family and Syk kinases results in elastase-dependent thrombohemorrhagic vasculopathy. Immunity. 2006;25:271–283.
    1. Looney MR, Su X, Van Ziffle JA, Lowell CA, Matthay MA. Neutrophils and their Fc gamma receptors are essential in a mouse model of transfusion-related acute lung injury. J Clin Invest. 2006;116:1615–1623.
    1. Frenette PS, Atweh GF. Sickle cell disease: old discoveries, new concepts, and future promise. J Clin Invest. 2007;117:850–858.
    1. Stuart MJ, Nagel RL. Sickle-cell disease. Lancet. 2004;364:1343–1360.
    1. Chang J, Shi PA, Chiang EY, Frenette PS. Intravenous immunoglobulins reverse acute vaso-occlusive crises in sickle cell mice through rapid inhibition of neutrophil adhesion. Blood. 2008;111:915–923.
    1. Turhan A, Weiss LA, Mohandas N, Coller BS, Frenette PS. Primary role for adherent leukocytes in sickle cell vascular occlusion: a new paradigm. Proc Natl Acad Sci U S A. 2002;99:3047–3051.
    1. Holness L, Knippen MA, Simmons L, Lachenbruch PA. Fatalities caused by TRALI. Transfus Med Rev. 2004;18:184–188.
    1. Silliman CC, Ambruso DR, Boshkov LK. Transfusion-related acute lung injury. Blood. 2005;105:2266–2273.
    1. Falati S, Gross P, Merrill-Skoloff G, Furie BC, Furie B. Real-time in vivo imaging of platelets, tissue factor and fibrin during arterial thrombus formation in the mouse. Nat Med. 2002;8:1175–1181.
    1. Chiang EY, Hidalgo A, Chang J, Frenette PS. Imaging receptor microdomains on leukocyte subsets in live mice. Nat Methods. 2007;4:219–222.
    1. Totani L, et al. Src-family kinases mediate an outside-in signal necessary for beta2 integrins to achieve full activation and sustain firm adhesion of polymorphonuclear leucocytes tethered on E-selectin. Biochem J. 2006;396:89–98.
    1. Smith ML, Olson TS, Ley K. CXCR2- and E-selectin-induced neutrophil arrest during inflammation in vivo. J Exp Med. 2004;200:935–939.
    1. Simon SI, et al. L-selectin (CD62L) cross-linking signals neutrophil adhesive functions via the Mac-1 (CD11b/CD18) beta 2-integrin. J Immunol. 1995;155:1502–1514.
    1. Butcher EC. Leukocyte-endothelial cell recognition: three (or more) steps to specificity and diversity. Cell. 1991;67:1033–1036.
    1. Lawrence MB, Springer TA. Leukocytes roll on a selectin at physiologic flow rates: distinction from and prerequisite for adhesion through integrins. Cell. 1991;65:859–873.
    1. Barreiro O, et al. Dynamic interaction of VCAM-1 and ICAM-1 with moesin and ezrin in a novel endothelial docking structure for adherent leukocytes. J Cell Biol. 2002;157:1233–1245.
    1. Carman CV, Springer TA. A transmigratory cup in leukocyte diapedesis both through individual vascular endothelial cells and between them. J Cell Biol. 2004;167:377–388.
    1. Lorant DE, et al. Inflammatory roles of P-selectin. J Clin Invest. 1993;92:559–570.
    1. Evangelista V, et al. Platelet/polymorphonuclear leukocyte interaction: P-selectin triggers protein-tyrosine phosphorylation-dependent CD11b/CD18 adhesion: role of PSGL-1 as a signaling molecule. Blood. 1999;93:876–885.
    1. Wang HB, et al. P-selectin primes leukocyte integrin activation during inflammation. Nat Immunol. 2007;8:882–892.
    1. Shappell SB, et al. Mac-1 (CD11b/CD18) mediates adherence-dependent hydrogen peroxide production by human and canine neutrophils. J Immunol. 1990;144:2702–2711.
    1. Husemann J, Obstfeld A, Febbraio M, Kodama T, Silverstein SC. CD11b/CD18 mediates production of reactive oxygen species by mouse and human macrophages adherent to matrixes containing oxidized LDL. Arterioscler Thromb Vasc Biol. 2001;21:1301–1305.
    1. Zarbock A, Singbartl K, Ley K. Complete reversal of acid-induced acute lung injury by blocking of platelet-neutrophil aggregation. J Clin Invest. 2006;116:3211–3219.
    1. Doerschuk CM. Mechanisms of leukocyte sequestration in inflamed lungs. Microcirculation. 2001;8:71–88.
    1. Lutz HU, et al. Naturally occurring anti-band-3 antibodies and complement together mediate phagocytosis of oxidatively stressed human erythrocytes. Proc Natl Acad Sci U S A. 1987;84:7368–7372.
    1. Wang RH, Phillips G, Jr, Medof ME, Mold C. Activation of the alternative complement pathway by exposure of phosphatidylethanolamine and phosphatidylserine on erythrocytes from sickle cell disease patients. J Clin Invest. 1993;92:1326–1335.
    1. Zennadi R, et al. Role and regulation of sickle red cell interactions with other cells: ICAM-4 and other adhesion receptors. Transfus Clin Biol. 2008;15:23–28.
    1. Gaarder A, Jonsen J, Laland S, Hellem A, Owren PA. Adenosine diphosphate in red cells as a factor in the adhesiveness of human blood platelets. Nature. 1961;192:531–532.
    1. Santos MT, et al. Enhancement of platelet reactivity and modulation of eicosanoid production by intact erythrocytes. A new approach to platelet activation and recruitment. J Clin Invest. 1991;87:571–580.
    1. Coller BS. Leukocytosis and ischemic vascular disease morbidity and mortality: is it time to intervene? Arterioscler Thromb Vasc Biol. 2005;25:658–670.
    1. Goel MS, Diamond SL. Adhesion of normal erythrocytes at depressed venous shear rates to activated neutrophils, activated platelets, and fibrin polymerized from plasma. Blood. 2002;100:3797–3803.
    1. Wakefield TW, Myers DD, Henke PK. Mechanisms of venous thrombosis and resolution. Arterioscler Thromb Vasc Biol. 2008;28:387–391.
    1. Dagia NM, et al. G-CSF induces E-selectin ligand expression on human myeloid cells. Nat Med. 2006;12:1185–1190.
    1. Arimura K, et al. Acute lung Injury in a healthy donor during mobilization of peripheral blood stem cells using granulocyte-colony stimulating factor alone. Haematologica. 2005;90 ECR10.
    1. Paszty C, et al. Transgenic knockout mice with exclusively human sickle hemoglobin and sickle cell disease. Science. 1997;278:876–878.

Source: PubMed

3
S'abonner