Excessive neutrophils and neutrophil extracellular traps contribute to acute lung injury of influenza pneumonitis

Teluguakula Narasaraju, Edwin Yang, Ramar Perumal Samy, Huey Hian Ng, Wee Peng Poh, Audrey-Ann Liew, Meng Chee Phoon, Nico van Rooijen, Vincent T Chow, Teluguakula Narasaraju, Edwin Yang, Ramar Perumal Samy, Huey Hian Ng, Wee Peng Poh, Audrey-Ann Liew, Meng Chee Phoon, Nico van Rooijen, Vincent T Chow

Abstract

Complications of acute respiratory distress syndrome (ARDS) are common among critically ill patients infected with highly pathogenic influenza viruses. Macrophages and neutrophils constitute the majority of cells recruited into infected lungs, and are associated with immunopathology in influenza pneumonia. We examined pathological manifestations in models of macrophage- or neutrophil-depleted mice challenged with sublethal doses of influenza A virus H1N1 strain PR8. Infected mice depleted of macrophages displayed excessive neutrophilic infiltration, alveolar damage, and increased viral load, later progressing into ARDS-like pathological signs with diffuse alveolar damage, pulmonary edema, hemorrhage, and hypoxemia. In contrast, neutrophil-depleted animals showed mild pathology in lungs. The brochoalveolar lavage fluid of infected macrophage-depleted mice exhibited elevated protein content, T1-α, thrombomodulin, matrix metalloproteinase-9, and myeloperoxidase activities indicating augmented alveolar-capillary damage, compared to neutrophil-depleted animals. We provide evidence for the formation of neutrophil extracellular traps (NETs), entangled with alveoli in areas of tissue injury, suggesting their potential link with lung damage. When co-incubated with infected alveolar epithelial cells in vitro, neutrophils from infected lungs strongly induced NETs generation, and augmented endothelial damage. NETs induction was abrogated by anti-myeloperoxidase antibody and an inhibitor of superoxide dismutase, thus implying that NETs generation is induced by redox enzymes in influenza pneumonia. These findings support the pathogenic effects of excessive neutrophils in acute lung injury of influenza pneumonia by instigating alveolar-capillary damage.

Copyright © 2011 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1
Figure 1
Effects of macrophage or neutrophil depletions on animal weight, leukocyte recruitment, and virus titer. Depletion of macrophages was achieved by intranasal administration of clodronate-liposome (50 μL/dose/mouse) at days −4 and −1 before virus challenge. For depletion of neutrophils or polymorphonuclear leukocytes (PMN), animals were injected intraperitoneally with anti-Ly6G monoclonal antibody (1A8) at 200 μg/dose/mouse a day before infection and every 48 hours thereafter. Animals were challenged with a sublethal dose of PR8 virus (100 PFU) through intranasal inoculation. A: Changes in body weight of BALB/c mice after infection. Animal weights were recorded daily for 10 dpi and expressed as means ± SE. At 10 dpi, the CL-I group showed more than 30% weight loss, whereas the 1A8-I and INF groups displayed 20% and 10% weight loss, respectively (n = 10). B: Differential cell counts of BALF measured by modified Giemsa staining. Neutrophil-dominant infiltration was observed in the CL-I and INF groups. Macrophage numbers were significantly decreased in the CL-I group on 5 dpi. The 1A8-I group showed significantly decreased neutrophil numbers but increased macrophage numbers. Results are expressed as means ± SE, with n = 5 per group. C: Determination of virus titers in mouse lung homogenates at 5 and 10 dpi. MDCK cells were infected with serial 10-fold dilutions of lung homogenates from uninfected or virus-infected animals. The numbers on the y axis represent virus titers expressed as 10y 50% tissue culture infectious dose (TCID50) per gram of total protein (means ± SE), with n = 5. CL, clodronate-liposome–treated; INF, infected; CL-I, clodronate-liposome–treated, infected; Con, normal allantoic fluid–inoculated; 1A8-I: anti-Ly6G monoclonal antibody–treated, infected group. *P < 0.05 versus the CL-I group; †P < 0.05 versus the INF and 1A8-I groups.
Figure 2
Figure 2
Effects of macrophage or neutrophil depletion on histopathological changes and NETs induction in lungs of mice challenged with sublethal influenza virus infection. A: Animals administered clodronate-liposome displayed normal architecture of airway (BR) and alveolar epithelia (AV) with minimal neutrophilic infiltration. B: Infected mice showed mild peribronchial infiltration and damage of bronchiolar epithelium at 5 dpi. C and D: Macrophage-depleted infected mice at 5 dpi exhibited severe alveolar damage and extensive neutrophil infiltration (arrows). E and F: Neutrophil-depleted infected animals at 5 dpi displayed mild bronchiolitis and infiltration of macrophages in alveolar spaces (arrows), but neutrophils were rarely seen. G: Lungs from the INF group at 10 dpi exhibited mild alveolitis and bronchiolar epithelial regeneration with mild peribronchial inflammation. H: Neutrophil-depleted infected animals at 10 dpi revealed mild alveolar damage. IK: Histopathological changes in lungs of mice from the CL-I group on 10 dpi. I: Lung sections show prominent pathological signs of diffuse alveolar damage with thickened interstitium, and alveolar spaces filled with protein exudates (asterisks). J: Extensive NETs formation at the terminal bronchioles opening into alveoli (white arrows). K: Bundles of NETs with endothelial damage (asterisk) were observed in large blood vessels. Magnifications: ×200 (A, B, C, E, G, H, I), and ×1000 (D, F, J, K). Scale bars: 50 μm. Semiquantitative histopathology scoring was performed (n = 5), with the scores shown in Table 1.
Figure 3
Figure 3
Assessment of acute lung injury by determining alveolar–capillary damage and release of MMP-9 into the alveolar air spaces. A: Damage of the thin microvascular barrier was determined using Western blot analyses of BALF samples for T1-α and thrombomodulin, which are membrane proteins present on alveolar type I pneumocytes and capillary endothelium. Lanes depict BALF samples from five groups: uninfected control (lanes 1 and 6); CL (lanes 2 and 7); INF (lanes 3 and 8); CL-I (lanes 4 and 9); and 1A8-I (lanes 5 and 10). T1-α and thrombomodulin levels were significantly elevated in the BALF of the CL-I group at both 5 and 10 dpi (lanes 4 and 9) compared to other groups. To determine the impact of neutrophil accumulation on lung injury, pro- and active MMP-9 levels were also assayed using Western blot analysis. Significantly increased MMP-9 levels were observed in CL-I compared to other groups, thus alluding to the contribution of MMP-9 to basement membrane degradation and promoting tissue damage. B and C: Densitometric analyses of Western blot bands, each expressed as fold change versus the control group. Data are represented as means ± SE, with n = 5 per group. *P < 0.05 versus other groups.
Figure 4
Figure 4
Effects of macrophage or neutrophil depletion on protein leakage into the air spaces and on MPO enzyme activity. A: Significant increase in BALF protein content was observed in the CL-I group compared to other groups, indicating alveolar capillary damage. B and C: MPO activities measured in both BALF samples and lung homogenates from the CL-I group were significantly increased compared to the other groups, correlating with intense neutrophil recruitment. Data are represented as means ± SE, with n = 5. *P < 0.05 versus other groups.
Figure 5
Figure 5
In vivo evidence for NETs formation after lethal challenge of influenza virus in mice. Paraffin-embedded lung tissues from mice challenged with lethal doses of influenza A/PR/8 H1N1 virus (500 PFU) were stained with hematoxylin-eosin or by immunofluorescence. A: Histopathology of lungs showing tissue consolidation and alveolar destruction. B: Extensive induction of NETs was detected within the alveoli (white arrows). C: Bundles of NETs were also observed in large blood vessels showing damaged endothelium (arrows). D and E: Nuclei stained with DAPI showing alveoli (white arrows) or occlusion in the bronchiole (defined by the white line). F and G: NETs were identified by close localization of DNA (blue) with histone H2B (F, green), and neutrophil granule marker MMP-9 (G, green). White arrows indicate NETs formation. Scale bars: 20 μm (A); 50 μm (BG).
Figure 6
Figure 6
In vitro NETs induction by co-incubation of neutrophils with infected alveolar epithelial cells, and the role of redox enzymes. Neutrophils isolated from lungs were incubated with influenza virus–infected alveolar epithelial cells (at a multiplicity of infection of 20 for 5 hours) in the presence of inhibitors of specific enzymes, including 10 μg/mL anti-MPO, 20 μmol/L diphenyleneiodonium chloride (DPI, inhibitor of NADPH oxidase), 2 mmol/L diethyldithiocarbamate (DETC, inhibitor of SOD), or 2 μmol/L H2O2. After 150 minutes of incubation, the DNA release was visualized by fluorescence microscopy with DAPI staining. A and B: Induction of NETs when neutrophils were co-incubated with infected alveolar epithelial cells (white arrows). C: Double staining with anti-influenza antibody showing infected epithelial cells embroiled by NETs (white arrows). Influenza virus was stained with anti-rabbit polyclonal antibody and secondary anti-rabbit Alexa Fluor 546 (orange). D: NETs stained with anti-MPO (arrowheads), FITC-phalloidin (for F-actin) and DAPI (arrow) to detect the MPO localization within NETs. E: Neutrophils co-incubated with uninfected alveolar epithelial cells revealed minimal NETs generation in the absence of viral stimulation. F: Neutrophils incubated in the presence of anti-MPO antibody resulted in decreased NETs induction. G: Neutrophils alone showing intact nuclei. H: Quantitative assessment of the percentage of NETs-forming cells revealed significant NETs induction following co-incubation of neutrophils (PMN) with infected alveolar epithelial cells (IEP), but not with uninfected control epithelial cells (CONEP). NETs induction was significantly diminished in the presence of anti-MPO and DETC, but not by DPI or anti–ENA-78 (anti–CXCL-5). The presence of H2O2 also significantly induced NETs formation. Values are means ± SE of three independent experiments. *P < 0.05 versus PMN+IEP group.
Figure 7
Figure 7
NETs-induced endothelial cell damage. Neutrophils (PMN) were incubated with HUVECs (ENDO) in the presence or absence of 2 or 5 μmol/L of H2O2. Endothelial damage was assessed by propidium iodide staining followed by immunofluorescence analysis. A: Neutrophils alone do not induce endothelial damage, but the presence of H2O2 significantly induced NETs to cause significant endothelial damage as indicated by increased propidium iodide–positive cells. Statistical significance at *P < 0.05 versus other groups. B: Western blot analysis for release of thrombomodulin into the culture supernatant. Lanes 1 and 2: Supernatants from neutrophil-endothelium cocultures without H2O2 or in the presence of (lane 3) 2 μmol/L H2O2 or (lane 4) 5 μmol/L H2O2. Supernatants of HUVECs alone in the presence of (lane 5) 2 μmol/L H2O2 or (lane 6) 5 μmol/L H2O2. The data are expressed as means ± SE of three independent experiments.

References

    1. Beigel J.H., Farrar J., Han A.M., Hayden F.G., Hyer R., de Jong M.D., Lochindarat S., Nguyen T.K., Nguyen T.H., Tran T.H., Nicoll A., Touch S., Yuen K.Y., Writing Committee of the World Health Organization (WHO) Consultation on Human Influenza A/H5 Avian influenza A (H5N1) infection in humans. N Engl J Med. 2005;353:1374–1385.
    1. Bauer T.T., Ewig S., Rodloff A.C., Müller E.E. Acute respiratory distress syndrome and pneumonia: a comprehensive review of clinical data. Clin Infect Dis. 2006;43:748–756.
    1. Yu H., Gao Z., Feng Z., Shu Y., Xiang N., Zhou L., Huai Y., Feng L., Peng Z., Li Z., Xu C., Li J., Hu C., Li Q., Xu X., Liu X., Liu Z., Xu L., Chen Y., Luo H., Wei L., Zhang X., Xin J., Guo J., Wang Q., Yuan Z., Zhou L., Zhang K., Zhang W., Yang J., Zhong X., Xia S., Li L., Cheng J., Ma E., He P., Lee S.S., Wang Y., Uyeki T.M., Yang W. Clinical characteristics of 26 human cases of highly pathogenic avian influenza A (H5N1) virus infection in China. PLoS One. 2008;3:e2985.
    1. Kawachi S., Luong S.T., Shigematsu M., Furuya H., Phung T.T., Phan P.H., Nunoi H., Nguyen L.T., Suzuki K. Risk parameters of fulminant acute respiratory distress syndrome and avian influenza (H5N1) infection in Vietnamese children. J Infect Dis. 2009;200:510–515.
    1. Miller R.R., Markewitz B.A., Rolfs R.T., Brown S.M., Dascomb K.K., Grissom C.K., Friedrichs M.D., Mayer J., Hirshberg E.L., Conklin J., Paine R., Dean N.C. Clinical findings and demographic factors associated with ICU admission in Utah due to novel 2009 influenza A(H1N1) infection. Chest. 2010;137:752–758.
    1. Lew T.W., Kwek T.K., Tai D., Earnest A., Loo S., Singh K., Kwan K.M., Chan Y., Yim C.F., Bek S.L., Kor A.C., Yap W.S., Chelliah Y.R., Lai Y.C., Goh S.K. Acute respiratory distress syndrome in critically ill patients with severe acute respiratory syndrome. JAMA. 2003;290:374–380.
    1. Kobasa D., Jones S.M., Shinya K., Kash J.C., Copps J., Ebihara H., Hatta Y., Kim J.H., Halfmann P., Hatta M., Feldmann F., Alimonti J.B., Fernando L., Li Y., Katze M.G., Feldmann H., Kawaoka Y. Aberrant innate immune response in lethal infection of macaques with the 1918 influenza virus. Nature. 2007;445:319–323.
    1. Xu T., Qiao J., Zhao L., Wang G., He G., Li K., Tian Y., Gao M., Wang J., Wang H., Dong C. Acute respiratory distress syndrome induced by avian influenza A (H5N1) virus in mice. Am J Respir Crit Care Med. 2006;174:1011–1017.
    1. Deng G., Bi J., Kong F., Li X., Xu Q., Dong J., Zhang M., Zhao L., Luan Z., Lv N., Qiao J. Acute respiratory distress syndrome induced by H9N2 virus in mice. Arch Virol. 2010;155:187–195.
    1. Brown K.A., Brain S.D., Pearson J.D., Edgeworth J.D., Lewis S.M., Treacher D.F. Neutrophils in development of multiple organ failure in sepsis. Lancet. 2006;368:157–169.
    1. Kolls J.K. Oxidative stress in sepsis: a redox redux. J Clin Invest. 2006;116:860–863.
    1. Kodama T., Yukioka H., Kato T., Kato N., Hato F., Kitagawa S. Neutrophil elastase as a predicting factor for development of acute lung injury. Intern Med. 2007;46:699–704.
    1. Zmijewski J.W., Lorne E., Zhao X., Tsuruta Y., Sha Y., Liu G., Siegal G.P., Abraham E. Mitochondrial respiratory complex I regulates neutrophil activation and severity of lung injury. Am J Respir Crit Care Med. 2008;178:168–179.
    1. Zhao X., Zmijewski J.W., Lorne E., Liu G., Park Y.J., Tsuruta Y., Abraham E. Activation of AMPK attenuates neutrophil proinflammatory activity and decreases the severity of acute lung injury. Am J Physiol Lung Cell Mol Physiol. 2008;295:L497–L504.
    1. El Kebir D., Jozsef L., Pan W., Wang L., Petasis N.A., Serhan C.N., Filep J.G. 15-epi-lipoxin A4 inhibits myeloperoxidase signaling and enhances resolution of acute lung injury. Am J Respir Crit Care Med. 2009;180:311–319.
    1. Bdeir K., Higazi A.A., Kulikovskaya I., Christofidou-Solomidou M., Vinogradov S.A., Allen T.C., Idell S., Linzmeier R., Ganz T., Cines D.B. Neutrophil alpha-defensins cause lung injury by disrupting the capillary-epithelial barrier. Am J Respir Crit Care Med. 2010;181:935–946.
    1. Perez-Padilla R., de la Rosa-Zamboni D., Ponce de Leon S., Hernandez M., Quiñones-Falconi F., Bautista E., Ramirez-Venegas A., Rojas-Serrano J., Ormsby C.E., Corrales A., Higuera A., Mondragon E., Cordova-Villalobos J.A., INER Working Group on Influenza Pneumonia and respiratory failure from swine-origin influenza A (H1N1) in Mexico. N Engl J Med. 2009;361:680–689.
    1. Mauad T., Hajjar L.A., Callegari G.D., da Silva L.F., Schout D., Galas F.R., Alves V.A., Malheiros D.M., Auler J.O., Ferreira A.F., Borsato M.R., Bezerra S.M., Gutierrez P.S., Caldini E.T., Pasqualucci C.A., Dolhnikoff M., Saldiva P.H. Lung pathology in fatal novel human influenza A (H1N1) infection. Am J Respir Crit Care Med. 2010;181:72–79.
    1. Djibre M., Berkane N., Salengro A., Ferrand E., Denis M., Chalumeau-Lemoine L., Parrot A., Mayaud C., Fartoukh M. Non-invasive management of acute respiratory distress syndrome related to Influenza A (H1N1) virus pneumonia in a pregnant woman. Intensive Care Med. 2010;36:373–374.
    1. Yokoyama T., Tsushima K., Ushiki A., Kobayashi N., Urushihata K., Koizumi T., Kubo K. Acute lung injury with alveolar hemorrhage due to a novel swine-origin influenza A (H1N1) virus. Intern Med. 2010;49:427–430.
    1. Kobasa D., Takada A., Shinya K., Hatta M., Halfmann P., Theriault S., Suzuki H., Nishimura H., Mitamura K., Sugaya N., Usui T., Murata T., Maeda Y., Watanabe S., Suresh M., Suzuki T., Suzuki Y., Feldmann H., Kawaoka Y. Enhanced virulence of influenza A viruses with the haemagglutinin of the 1918 pandemic virus. Nature. 2004;431:703–707.
    1. Tumpey T.M., Basler C.F., Aguilar P.V., Zeng H., Solorzano A., Swayne D.E., Cox N.J., Katz J.M., Taubenberger J.K., Palese P., Garcia-Sastre A. Characterization of the reconstructed 1918 Spanish influenza pandemic virus. Science. 2005;310:77–80.
    1. Narasaraju T., Sim M.K., Ng H.H., Phoon M.C., Shanker N., Lal S.K., Chow V.T. Adaptation of human influenza H3N2 virus in a mouse pneumonitis model: insights into viral virulence, tissue tropism and host pathogenesis. Microbes Infect. 2009;11:2–11.
    1. Sakai S., Kawamata H., Mantani N., Kogure T., Shimada Y., Terasawa K., Sakai T., Imanishi N., Ochiai H. Therapeutic effect of anti-macrophage inflammatory protein 2 antibody on influenza virus-induced pneumonia in mice. J Virol. 2000;74:2472–2476.
    1. Crowe C.R., Chen K., Pociask D.A., Alcorn J.F., Krivich C., Enelow R.I., Ross T.M., Witztum J.L., Kolls J.K. Critical role of IL-17RA in immunopathology of influenza infection. J Immunol. 2009;183:5301–5310.
    1. Perrone L.A., Szretter K.J., Katz J.M., Mizgerd J.P., Tumpey T.M. Mice lacking both TNF and IL-1 receptors exhibit reduced lung inflammation and delay in onset of death following infection with a highly virulent H5N1 virus. J Infect Dis. 2010;202:1161–1170.
    1. Tate M.D., Brooks A.G., Reading P.C. The role of neutrophils in the upper and lower respiratory tract during influenza virus infection of mice. Respir Res. 2008;9:57.
    1. Tate M.D., Deng Y.M., Jones J.E., Anderson G.P., Brooks A.G., Reading P.C. Neutrophils ameliorate lung injury and the development of severe disease during influenza infection. J Immunol. 2009;183:7441–7450.
    1. Brinkmann V., Reichard U., Goosmann C., Fauler B., Uhlemann Y., Weiss D.S., Weinrauch Y., Zychlinsky A. Neutrophil extracellular traps kill bacteria. Science. 2004;303:1532–1535.
    1. Clark S.R., Ma A.C., Tavener S.A., McDonald B., Goodarzi Z., Kelly M.M., Patel K.D., Chakrabarti S., McAvoy E., Sinclair G.D., Keys E.M., Allen-Vercoe E., Devinney R., Doig C.J., Green F.H., Kubes P. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med. 2007;13:463–469.
    1. Perrone L.A., Plowden J.K., García-Sastre A., Katz J.M., Tumpey T.M. H5N1 and 1918 pandemic influenza virus infection results in early and excessive infiltration of macrophages and neutrophils in the lungs of mice. PLoS Pathog. 2008;4:e1000115.
    1. Curran C.S., Demick K.P., Mansfield J.M. Lactoferrin activates macrophages via TLR4-dependent and -independent signalling pathways. Cell Immunol. 2006;242:23–30.
    1. Jaillon S., Peri G., Delneste Y., Frémaux I., Doni A., Moalli F., Garlanda C., Romani L., Gascan H., Bellocchio S., Bozza S., Cassatella M.A., Jeannin P., Mantovani A. The humoral pattern recognition receptor PTX3 is stored in neutrophil granules and localizes in extracellular traps. J Exp Med. 2007;204:793–804.
    1. Wartha F., Beiter K., Normark S., Henriques-Normark B. Neutrophil extracellular traps: casting the NET over pathogenesis. Curr Opin Microbiol. 2007;10:52–56.
    1. Kessenbrock K., Krumbholz M., Schönermarck U., Back W., Gross W.L., Werb Z., Grone H.J., Brinkmann V., Jenne D.E. Netting neutrophils in autoimmune small-vessel vasculitis. Nat Med. 2009;15:623–625.
    1. Fuchs T.A., Abed U., Goosmann C., Hurwitz R., Schulze I., Wahn V., Weinrauch Y., Brinkmann V., Zychlinsky A. A novel cell death program leads to neutrophil extracellular traps. J Cell Biol. 2007;176:231–241.
    1. Ermert D., Urban C.F., Laube B., Goosmann C., Zychlinsky A., Brinkmann V. Mouse neutrophil extracellular traps in microbial infections. J Innate Immun. 2009;1:181–193.
    1. Beiter K., Wartha F., Albiger B., Normark S., Zychlinsky A., Henriques-Normark B. An endonuclease allows Streptococcus pneumoniae to escape from neutrophil extracellular traps. Curr Biol. 2006;16:401–407.
    1. Wardini A.B., Guimarães-Costa A.B., Nascimento M.T., Nadaes N.R., Danelli M.D., Mazur C., Benjamim C.F., Saraiva E.M., Pinto-da-Silva L.H. Characterization of neutrophil extracellular traps in cats naturally-infected with feline leukemia virus. J Gen Virol. 2010;91:259–264.
    1. Van Rooijen N., Sanders A., van den Berg T.K. Apoptosis of macrophages induced by liposome-mediated intracellular delivery of clodronate and propamidine. J Immunol Methods. 1996;193:93–99.
    1. Van Rooijen N.V., Sanders A. Liposome mediated depletion of macrophages: mechanism of action, preparation of liposomes and applications. J Immunol Methods. 1994;174:83–93.
    1. Daley J.M., Thomay A.A., Connolly M.D., Reichner J.S., Albina J.E. Use of Ly6G-specific monoclonal antibody to deplete neutrophils in mice. J Leukoc Biol. 2008;83:64–70.
    1. Narasaraju T., Ng H.H., Phoon M.C., Chow V.T. MCP-1 antibody treatment enhances damage and impedes repair of the alveolar epithelium in influenza pneumonitis. Am J Respir Cell Mol Biol. 2010;42:732–743.
    1. Ferretti S., Bonneau O., Dubois G.R., Jones C.E., Trifilieff A. IL-17, produced by lymphocytes and neutrophils, is necessary for lipopolysaccharide-induced airway neutrophilia: iL-15 as a possible trigger. J Immunol. 2003;170:2106–2112.
    1. Wang S.Z., Forsyth K.D. The interaction of neutrophils with respiratory epithelial cells in viral infection. Respirology. 2000;5:1–10.
    1. Mugge A., Elwell J.H., Peterson T.E., Harrison D.G. Release of intact endothelium-derived relaxing factor depends on endothelial superoxide dismutase activity. Am J Physiol. 1991;260:C219–C225.
    1. Wind S., Beuerlein K., Eucker T., Muller H., Scheurer P., Armitage M.E., Ho H., Schmidt H.H.H.W., Wingler K. Comparative pharmacology of chemically distinct NADPH oxidase inhibitors. Br J Pharmacol. 2010;161:885–898.
    1. Gotte M., Bernfield M., Joussen A.M. Increased leukocyte-endothelial interactions in syndecan-1-deficient mice involve heparan sulfate-dependent and -independent steps. Curr Eye Res. 2005;30:417–422.
    1. Knobil K., Choi A.M., Weigand G.W., Jacoby D.B. Role of oxidants in influenza virus-induced gene expression. Am J Physiol. 1998;274:L134–L142.

Source: PubMed

3
Abonnieren