Complement C5a Receptor 1 Exacerbates the Pathophysiology of N. meningitidis Sepsis and Is a Potential Target for Disease Treatment

Johannes B Herrmann, Marcel Muenstermann, Lea Strobel, Alexandra Schubert-Unkmeir, Trent M Woodruff, Scott D Gray-Owen, Andreas Klos, Kay O Johswich, Johannes B Herrmann, Marcel Muenstermann, Lea Strobel, Alexandra Schubert-Unkmeir, Trent M Woodruff, Scott D Gray-Owen, Andreas Klos, Kay O Johswich

Abstract

Sepsis caused by Neisseria meningitidis (meningococcus) is a rapidly progressing, life-threatening disease. Because its initial symptoms are rather unspecific, medical attention is often sought too late, i.e., when the systemic inflammatory response is already unleashed. This in turn limits the success of antibiotic treatment. The complement system is generally accepted as the most important innate immune determinant against invasive meningococcal disease since it protects the host through the bactericidal membrane attack complex. However, complement activation concomitantly liberates the C5a peptide, and it remains unclear whether this potent anaphylatoxin contributes to protection and/or drives the rapidly progressing immunopathogenesis associated with meningococcal disease. Here, we dissected the specific contribution of C5a receptor 1 (C5aR1), the canonical receptor for C5a, using a mouse model of meningococcal sepsis. Mice lacking C3 or C5 displayed susceptibility that was enhanced by >1,000-fold or 100-fold, respectively, consistent with the contribution of these components to protection. In clear contrast, C5ar1-/- mice resisted invasive meningococcal infection and cleared N. meningitidis more rapidly than wild-type (WT) animals. This favorable outcome stemmed from an ameliorated inflammatory cytokine response to N. meningitidis in C5ar1-/- mice in both in vivo and ex vivo whole-blood infections. In addition, inhibition of C5aR1 signaling without interference with the complement bactericidal activity reduced the inflammatory response also in human whole blood. Enticingly, pharmacologic C5aR1 blockade enhanced mouse survival and lowered meningococcal burden even when the treatment was administered after sepsis induction. Together, our findings demonstrate that C5aR1 drives the pathophysiology associated with meningococcal sepsis and provides a promising target for adjunctive therapy.IMPORTANCE The devastating consequences of N. meningitidis sepsis arise due to the rapidly arising and self-propagating inflammatory response that mobilizes antibacterial defenses but also drives the immunopathology associated with meningococcemia. The complement cascade provides innate broad-spectrum protection against infection by directly damaging the envelope of pathogenic microbes through the membrane attack complex and triggers an inflammatory response via the C5a peptide and its receptor C5aR1 aimed at mobilizing cellular effectors of immunity. Here, we consider the potential of separating the bactericidal activities of the complement cascade from its immune activating function to improve outcome of N. meningitidis sepsis. Our findings demonstrate that the specific genetic or pharmacological disruption of C5aR1 rapidly ameliorates disease by suppressing the pathogenic inflammatory response and, surprisingly, allows faster clearance of the bacterial infection. This outcome provides a clear demonstration of the therapeutic benefit of the use of C5aR1-specific inhibitors to improve the outcome of invasive meningococcal disease.

Keywords: C5aR1; Neisseria meningitidis; anaphylatoxins; complement system; inflammation; invasive disease; mouse model; neutrophils; sepsis; whole-blood model.

Copyright © 2018 Herrmann et al.

Figures

FIG 1
FIG 1
Role of complement in murine model of N. meningitidis sepsis. (A) Survival curves (top panels) and bacteremia (lower panels) of WT, C3−/−, and Hc°/° mice intraperitoneally infected with N. meningitidis strain MC58 inocula as indicated above the graphs. LOD, limit of detection; ns, not significant; *, P < 0.05; **, P < 0.01 (in Mantel-Cox test). (B) In vivo C3b deposition onto N. meningitidis (Nme) in infected mice. Blood smears of the infected mice described in the panel A legend were analyzed by immunofluorescence microscopy at ×60 magnification after staining with DAPI, rabbit anti-N. meningitidis (Alexa 488 channel), and anti-C3 (Cy3 channel). About 500 N. meningitidis cells per sample were identified by green fluorescence, and the fraction of C3-positive N. meningitidis cells was expressed as the percentage of all analyzed N. meningitidis cells. Each dot represents results from one animal. Representative immunofluorescence (IF) microscopy images are shown in Fig. S3. ns, not significant; *, P < 0.05 (in one-way analysis of variance [ANOVA] with Dunnett’s post hoc test). (C) Ex vivo C3b deposition from mouse lepirudin plasma on N. meningitidis MC58 as analyzed by whole-cell ELISA. ns, not significant; ***, P < 0.005 (in one-way ANOVA with Dunnett’s post hoc test). (D) Ex vivo uptake of N. meningitidis by neutrophils of WT, C3−/−, and Hc°/° mice. Lepirudinized whole-mouse blood was infected with 107 CFU/ml of acapsulate N. meningitidis expressing GFP (MC58Δcsb-GFP), and the mean fluorescence intensity (MFI) of neutrophils (PMN; gated as Ly6Ghi) was analyzed by flow cytometry. The graph shows means ± standard deviations of the means (SEM) of results from three independent experiments. *, P < 0.05 (in one-way ANOVA with Bonferroni’s post hoc test). ns, not significant.
FIG 2
FIG 2
Anaphylatoxin release during N. meningitidis sepsis. (A and C) Plasma levels of C3a and C5a, respectively, in WT mice at 12 h postinfection with 105 CFU of N. meningitidis strain MC58 as measured by ELISA. Plotted are means ± SEM. *, P < 0.05; **, P < 0.01 (in unpaired, two-tailed Student’s t test). (B and D) Correlation of C3a and C5a concentrations, respectively, with bacterial burden of infected mice. (E and F) Dose response of plasma C5a liberation in ex vivo infection of lepirudin anticoagulated whole-mouse blood (n = 3 independent samples) and whole human blood (n = 15 individual donors), respectively, with N. meningitidis MC58 (means ± SEM). *, P < 0.05; ***, <0.001; ****, <0.0001 (in one-way ANOVA with Dunnett’s post hoc test using PBS as a comparator).
FIG 3
FIG 3
In vivo N. meningitidis sepsis in WT versus C5ar1−/− mice. (A) Surface expression of C5aR1 as measured by flow cytometry on neutrophils (gated as Ly6Ghi) in WT mouse lepirudin-treated whole blood after 1 h of infection with 107 CFU of N. meningitidis MC58 versus uninfected control (ctrl.) and C5ar1−/− neutrophils as a staining control. Plotted is the mean fluorescence intensity (MFI) of results from three independent experiments. *, P < 0.05 (in paired matched observations per mouse; two-tailed Student’s t test). (B) Survival curves of WT and C5aR1−/− mice after intraperitoneal infection with 105 CFU of N. meningitidis MC58. ****, P < 0.0001 (in Mantel-Cox test). (C) N. meningitidis counts in blood of infected mice at indicated time points. The 18/24 h data comprise 18-h values from mice not surviving until 24 h plus 24-h values from the mice surviving until then. ns, not significant; **, P < 0.001; ***, P < 0.0001 (in unpaired, two-tailed Mann-Whitney test). (D) Plasma levels of inflammatory mediators at 12 h after intraperitoneal infection with 105 CFU N. meningitidis MC58 of WT versus C5ar1−/− mice (means ± SEM; n = 5 per genotype). ns, not significant; **, P < 0.01 (in unpaired, two-tailed Student’s t test).
FIG 4
FIG 4
Role of phagocytes in N. meningitidis sepsis in WT and C5ar1−/− mice. (A) Immunofluorescence microscopy (×60 magnification) of tissue sections of lung at indicated time points after intraperitoneal infection with 105 CFU N. meningitidis MC58. Blue, nuclei; red, neutrophil elastase; green, N. meningitidis (arrowheads). Background data in the green channel stem from erythrocytes, indicating the position of blood vessels. Insets are enlargements of points of interest from the same image. (B) Oxidative burst of Ly6Ghi neutrophils (n = 3) assayed by DHR123 assay in lepirudin-treated whole blood infected with 105 or 107 CFU per ml of N. meningitidis MC58. The positive control was PMA (100 nM). ns, not significant; **, P < 0.01 (in unpaired, two-tailed Student’s t test). (C) Neutrophil degranulation measured as the difference in levels of CD11b surface expression between infected (107 CFU/ml) and uninfected lepirudin-treated whole-blood samples from WT and C5ar1−/− mice after 1 h of incubation. Neutrophils were gated as Ly6Ghi cells and CD11b stained with clone M1/70. *, P < 0.05 (in unpaired, two-tailed Student’s t test). (D) Uptake of N. meningitidis MC58Δcsb-GFP by Ly6Ghi neutrophils in ex vivo infection of lepirudin-treated whole blood (means ± SEM of the geometric mean of GFP fluorescence; n = 3). ns, not significant, *, P < 0.05 (in one-way ANOVA with Bonferroni’s post hoc test). (E) Ex vivo N. meningitidis survival at different inocula in lepirudin-treated whole blood of WT and C5ar1−/− mice (means of CFU per milliliter ± SEM; n = 3). As a positive control for N. meningitidis killing, 1 µg/ml of anti-serogroup B mouse monoclonal antibody mAb735 was added. (F and H) Survival of n = 7 to 8 WT and C5ar1−/− mice, respectively, infected with 104 CFU of N. meningitidis MC58 after depletion of monocytes/macrophages (clodronate liposomes) or neutrophils (RB6-8C5) or the control (PBS). The experiment was conducted in a blind manner for depletion treatment. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001 (in Mantel-Cox analysis relative to control). (G and I) N. meningitidis counts in blood of mice in panels G and I. ns, not significant; *, P < 0.05; **, P < 0.01; ****, P < 0.0001 (in one-way ANOVA, applying Bonferrroni’s post hoc test).
FIG 5
FIG 5
Cytokine induction in in vivo N. meningitidis infection of WT and C5ar1−/− mice and in whole-blood model. (A and B) Levels of IL-6 and CXCL-1 in plasma and peritoneal lavage fluid (PL), respectively, from n = 8 mice per genotype after intraperitoneal administration of 5 × 108 CFU of heat-inactivated N. meningitidis MC58. Plotted are means ± SEM. ns, not significant; *, P < 0.05; **, P < 0.01 (in unpaired, two-tailed Student’s t test). (C) Bacterial counts over time in ex vivo infection of lepirudin-treated mouse whole blood with 106 CFU/ml N. meningitidis. Plotted are means ± SEM of results from n = 3 independent experiments. (D) CXCL-1 concentrations (means ± SEM) in plasma of whole-blood infection from the experiment described for panel C. *, P < 0.05; **, P < 0.01 (in unpaired, two-tailed Student’s t test).
FIG 6
FIG 6
Inhibition of inflammatory cytokine release and neutrophil responses by C5aR1 blockade in lepirudin-anticoagulated human whole-blood infection with N. meningitidis. (A) IL-8 in human whole blood infected with 106 CFU/ml N. meningitidis MC58 in the presence of C5aR1 antagonist (C5aRAs) PMX53 or W-54011 (means ± SEM; n = 5 donors). (B) IL-8 in human whole blood infected for the indicated durations with 106 CFU/ml N. meningitidis MC58 in the presence of PMX53 (10 µM) or W-54011 (300 nM) (means ± SEM; n = 15 donors). (C) Cytokines at 90 min of infection of human whole blood with 106 CFU N. meningitidis MC58 (means ± SEM; n = 16 donors). (D) Oxidative burst measured by DHR123 fluorescence in neutrophils during infection of whole human blood. (E) Neutrophil degranulation during whole-blood infection by surface localization of CD11b normalized to “no C5aRA.” (F) Phagocytosis of MC58-GFP by PMNs in whole blood as determined by flow cytometry and expressed as percentages of PMNs with an increase in the level of FL1-H above the level measured for the noninfected control. (G) N. meningitidis viability in blood of donors with or without C5aRAs. (D to F) Infection with 107 CFU/ml of N. meningitidis MC58; lines indicate medians. (D to G) PMX53 was used at 10 µM and W-54011 at 300 nM. (A–G) *, P < 0.05; **, P < 0.01; ***, P < 0.001 (in repeated-measure ANOVA; matched observations per individual donor).
FIG 7
FIG 7
Pharmacologic targeting of C5aR1 ameliorates in vivo N. meningitidis sepsis. (A) Survival of WT mice after intraperitoneal infection with 105 CFU of N. meningitidis strain MC58. Mice were randomized into three treatment cohorts with intraperitoneal injections every 6 to 12 h (see Materials and Methods) with 3 mg/kg with C5aR1-antagonist PMX205 starting either 12 h before infection (“pre”; n = 8) or 4 h after infection (“post”; n = 6) or received vehicle only (“control”; n = 8). The experiment was conducted in a blind manner with respect to treatment cohorts. *, P < 0.05; ***, P < 0.001 (in Mantel-Cox test). (B) Bacteremia in the mice from the experiment described for panel A. ns, not significant; *, P < 0.05; **, P < 0.01 (in Kruskal-Wallis test with Dunn’s post hoc test). (C) Clinical scores for mice from the experiment described for panel A over time. Plotted are means ± SEM. (D) Relative body weights (means ± SEM). (E) Levels of CXCL-1, IL-6, and TNF-α in tail vein blood samples from mice from the experiment described for panel A at 12 h. ns, not significant; *, P < 0.05 (in one-way ANOVA, applying Dunnett’s post hoc test with the control as the comparator).

References

    1. Caugant DA, Maiden MC. 2009. Meningococcal carriage and disease—population biology and evolution. Vaccine 27(Suppl 2):B64–B70. doi:10.1016/j.vaccine.2009.04.061.
    1. Rosenstein NE, Perkins BA, Stephens DS, Popovic T, Hughes JM. 2001. Meningococcal disease. N Engl J Med 344:1378–1388. doi:10.1056/NEJM200105033441807.
    1. van Deuren M, Brandtzaeg P, van der Meer JW. 2000. Update on meningococcal disease with emphasis on pathogenesis and clinical management. Clin Microbiol Rev 13:144–166, table of contents. doi:10.1128/CMR.13.1.144-166.2000.
    1. Pathan N, Faust SN, Levin M. 2003. Pathophysiology of meningococcal meningitis and septicaemia. Arch Dis Child 88:601–607. doi:10.1136/adc.88.7.601.
    1. Nadel S. 2016. Treatment of meningococcal disease. J Adolesc Health 59:S21–S28. doi:10.1016/j.jadohealth.2016.04.013.
    1. Lewis LA, Ram S. 2014. Meningococcal disease and the complement system. Virulence 5:98–126. doi:10.4161/viru.26515.
    1. Figueroa JE, Densen P. 1991. Infectious diseases associated with complement deficiencies. Clin Microbiol Rev 4:359–395. doi:10.1128/CMR.4.3.359.
    1. Borrow R, Balmer P, Miller E. 2005. Meningococcal surrogates of protection—serum bactericidal antibody activity. Vaccine 23:2222–2227. doi:10.1016/j.vaccine.2005.01.051.
    1. Klos A, Tenner AJ, Johswich KO, Ager RR, Reis ES, Köhl J. 2009. The role of the anaphylatoxins in health and disease. Mol Immunol 46:2753–2766. doi:10.1016/j.molimm.2009.04.027.
    1. Ehrengruber MU, Geiser T, Deranleau DA. 1994. Activation of human neutrophils by C3a and C5A. Comparison of the effects on shape changes, chemotaxis, secretion, and respiratory burst. FEBS Lett 346:181–184. doi:10.1016/0014-5793(94)00463-3.
    1. Gerard C, Gerard NP. 1994. C5A anaphylatoxin and its seven transmembrane-segment receptor. Annu Rev Immunol 12:775–808. doi:10.1146/annurev.iy.12.040194.004015.
    1. Huey R, Hugli TE. 1985. Characterization of a C5a receptor on human polymorphonuclear leukocytes (PMN). J Immunol 135:2063–2068.
    1. Wetsel RA. 1995. Expression of the complement C5a anaphylatoxin receptor (C5aR) on non-myeloid cells. Immunol Lett 44:183–187. doi:10.1016/0165-2478(94)00212-A.
    1. Niederbichler AD, Hoesel LM, Westfall MV, Gao H, Ipaktchi KR, Sun L, Zetoune FS, Su GL, Arbabi S, Sarma JV, Wang SC, Hemmila MR, Ward PA. 2006. An essential role for complement C5a in the pathogenesis of septic cardiac dysfunction. J Exp Med 203:53–61. doi:10.1084/jem.20051207.
    1. Lacy M, Jones J, Whittemore SR, Haviland DL, Wetsel RA, Barnum SR. 1995. Expression of the receptors for the C5a anaphylatoxin, interleukin-8 and FMLP by human astrocytes and microglia. J Neuroimmunol 61:71–78. doi:10.1016/0165-5728(95)00075-D.
    1. Höpken UE, Lu B, Gerard NP, Gerard C. 1996. The C5a chemoattractant receptor mediates mucosal defence to infection. Nature 383:86–89. doi:10.1038/383086a0.
    1. Ward PA. 2004. The dark side of C5a in sepsis. Nat Rev Immunol 4:133–142. doi:10.1038/nri1269.
    1. Hill DJ, Griffiths NJ, Borodina E, Virji M. 2010. Cellular and molecular biology of Neisseria meningitidis colonization and invasive disease. Clin Sci (Lond) 118:547–564. doi:10.1042/CS20090513.
    1. Gorringe AR, Reddin KM, Funnell SG, Johansson L, Rytkönen A, Jonsson AB. 2005. Experimental disease models for the assessment of meningococcal vaccines. Vaccine 23:2214–2217. doi:10.1016/j.vaccine.2005.01.053.
    1. Oftung F, Lovik M, Andersen SR, Froholm LO, Bjune G. 1999. A mouse model utilising human transferrin to study protection against Neisseria meningitidis serogroup B induced by outer membrane vesicle vaccination. FEMS Immunol Med Microbiol 26:75–82. doi:10.1111/j.1574-695X.1999.tb01374.x.
    1. Melican K, Michea Veloso P, Martin T, Bruneval P, Duménil G. 2013. Adhesion of Neisseria meningitidis to dermal vessels leads to local vascular damage and purpura in a humanized mouse model. PLoS Pathog 9:e1003139. doi:10.1371/journal.ppat.1003139.
    1. Yi K, Stephens DS, Stojiljkovic I. 2003. Development and evaluation of an improved mouse model of meningococcal colonization. Infect Immun 71:1849–1855. doi:10.1128/IAI.71.4.1849-1855.2003.
    1. Johswich KO, McCaw SE, Strobel L, Frosch M, Gray-Owen SD. 2015. Sterilizing immunity elicited by Neisseria meningitidis carriage shows broader protection than predicted by serum antibody cross-reactivity in CEACAM1-humanized mice. Infect Immun 83:354–363. doi:10.1128/IAI.02495-14.
    1. Mollnes TE, Brekke OL, Fung M, Fure H, Christiansen D, Bergseth G, Videm V, Lappegård KT, Köhl J, Lambris JD. 2002. Essential role of the C5a receptor in E coli-induced oxidative burst and phagocytosis revealed by a novel lepirudin-based human whole blood model of inflammation. Blood 100:1869–1877.
    1. Oppermann M, Götze O. 1994. Plasma clearance of the human C5a anaphylatoxin by binding to leucocyte C5a receptors. Immunology 82:516–521.
    1. Unnewehr H, Rittirsch D, Sarma JV, Zetoune F, Flierl MA, Perl M, Denk S, Weiss M, Schneider ME, Monk PN, Neff T, Mihlan M, Barth H, Gebhard F, Ward PA, Huber-Lang M. 2013. Changes and regulation of the C5a receptor on neutrophils during septic shock in humans. J Immunol 190:4215–4225. doi:10.4049/jimmunol.1200534.
    1. Guo RF, Riedemann NC, Bernacki KD, Sarma VJ, Laudes IJ, Reuben JS, Younkin EM, Neff TA, Paulauskis JD, Zetoune FS, Ward PA. 2003. Neutrophil C5a receptor and the outcome in a rat model of sepsis. FASEB J 17:1889–1891. doi:10.1096/fj.03-0009fje.
    1. Bosmann M, Ward PA. 2012. Role of C3, C5 and anaphylatoxin receptors in acute lung injury and in sepsis. Adv Exp Med Biol 946:147–159. doi:10.1007/978-1-4614-0106-3_9.
    1. Grommes J, Soehnlein O. 2011. Contribution of neutrophils to acute lung injury. Mol Med 17:293–307. doi:10.2119/molmed.2010.00138.
    1. Staab EB, Sanderson SD, Wells SM, Poole JA. 2014. Treatment with the C5a receptor/CD88 antagonist PMX205 reduces inflammation in a murine model of allergic asthma. Int Immunopharmacol 21:293–300. doi:10.1016/j.intimp.2014.05.008.
    1. Jain U, Woodruff TM, Stadnyk AW. 2013. The C5a receptor antagonist PMX205 ameliorates experimentally induced colitis associated with increased IL-4 and IL-10. Br J Pharmacol 168:488–501. doi:10.1111/j.1476-5381.2012.02183.x.
    1. Fonseca MI, Ager RR, Chu SH, Yazan O, Sanderson SD, LaFerla FM, Taylor SM, Woodruff TM, Tenner AJ. 2009. Treatment with a C5aR antagonist decreases pathology and enhances behavioral performance in murine models of Alzheimer’s disease. J Immunol 183:1375–1383. doi:10.4049/jimmunol.0901005.
    1. Lee JD, Kumar V, Fung JN, Ruitenberg MJ, Noakes PG, Woodruff TM. 2017. Pharmacological inhibition of complement C5a-C5a1 receptor signalling ameliorates disease pathology in the hSOD1G93A mouse model of amyotrophic lateral sclerosis. Br J Pharmacol 174:689–699. doi:10.1111/bph.13730.
    1. Estabrook MM, Zhou D, Apicella MA. 1998. Nonopsonic phagocytosis of group C Neisseria meningitidis by human neutrophils. Infect Immun 66:1028–1036.
    1. Heyderman RS, Ison CA, Peakman M, Levin M, Klein NJ. 1999. Neutrophil response to Neisseria meningitidis: inhibition of adhesion molecule expression and phagocytosis by recombinant bactericidal/permeability-increasing protein (rBPI21). J Infect Dis 179:1288–1292. doi:10.1086/314706.
    1. Plant L, Wan H, Jonsson AB. 2006. MyD88-dependent signaling affects the development of meningococcal sepsis by nonlipooligosaccharide ligands. Infect Immun 74:3538–3546. doi:10.1128/IAI.00128-06.
    1. Plant L, Wan H, Jonsson AB. 2007. Non-lipooligosaccharide-mediated signalling via Toll-like receptor 4 causes fatal meningococcal sepsis in a mouse model. Cell Microbiol 9:657–669. doi:10.1111/j.1462-5822.2006.00816.x.
    1. Mook-Kanamori BB, Brouwer MC, Geldhoff M, Ende Av, van de Beek D. 2014. Cerebrospinal fluid complement activation in patients with pneumococcal and meningococcal meningitis. J Infect 68:542–547. doi:10.1016/j.jinf.2013.12.016.
    1. Sprong T, Brandtzaeg P, Fung M, Pharo AM, Høiby EA, Michaelsen TE, Aase A, van der Meer JW, van Deuren M, Mollnes TE. 2003. Inhibition of C5a-induced inflammation with preserved C5b-9-mediated bactericidal activity in a human whole blood model of meningococcal sepsis. Blood 102:3702–3710. doi:10.1182/blood-2003-03-0703.
    1. Sônego F, Castanheira FV, Ferreira RG, Kanashiro A, Leite CA, Nascimento DC, Colón DF, Borges VDF, Alves-Filho JC, Cunha FQ. 2016. Paradoxical roles of the neutrophil in sepsis: protective and deleterious. Front Immunol 7:155. doi:10.3389/fimmu.2016.00155.
    1. Riedemann NC, Guo RF, Bernacki KD, Reuben JS, Laudes IJ, Neff TA, Gao H, Speyer C, Sarma VJ, Zetoune FS, Ward PA. 2003. Regulation by C5a of neutrophil activation during sepsis. Immunity 19:193–202. doi:10.1016/S1074-7613(03)00206-1.
    1. Demissie DE, Kaplan SL, Romero JR, Leake JA, Barson WJ, Halasa NB, Byington CL, Shetty AK, Tan TQ, Hoffman JA, Lin PL, Edwards KM, Mason EO Jr, Cooperstock MS. 2013. Altered neutrophil counts at diagnosis of invasive meningococcal infection in children. Pediatr Infect Dis J 32:1070–1072. doi:10.1097/INF.0b013e31829e31f1.
    1. Talà A, Monaco C, Nagorska K, Exley RM, Corbett A, Zychlinsky A, Alifano P, Tang CM. 2011. Glutamate utilization promotes meningococcal survival in vivo through avoidance of the neutrophil oxidative burst. Mol Microbiol 81:1330–1342. doi:10.1111/j.1365-2958.2011.07766.x.
    1. Wilks KE, Dunn KL, Farrant JL, Reddin KM, Gorringe AR, Langford PR, Kroll JS. 1998. Periplasmic superoxide dismutase in meningococcal pathogenicity. Infect Immun 66:213–217.
    1. Bindl L, Buderus S, Dahlem P, Demirakca S, Goldner M, Huth R, Kohl M, Krause M, Kühl P, Lasch P, Lewandowski K, Merz U, Moeller J, Mohamad Y, Peters M, Porz W, Vierzig A, Rüchard J, Scharf J, Varnholt V; ESPNIC ARDS Database Group . 2003. Gender-based differences in children with sepsis and ARDS: the ESPNIC ARDS Database Group. Intensive Care Med 29:1770–1773. doi:10.1007/s00134-003-1948-z.
    1. Deniset JF, Surewaard BG, Lee WY, Kubes P. 2017. Splenic Ly6Ghigh mature and Ly6Gint immature neutrophils contribute to eradication of S. pneumoniae. J Exp Med 214:1333–1350. doi:10.1084/jem.20161621.
    1. Platonov AE, Beloborodov VB, Vershinina IV. 1993. Meningococcal disease in patients with late complement component deficiency: studies in the U.S.S.R. Medicine 72:374–392. doi:10.1097/00005792-199311000-00002.
    1. Flexner S. 1913. The results of the serum treatment in thirteen hundred cases of epidemic meningitis. J Exp Med 17:553–576. doi:10.1084/jem.17.5.553.
    1. Swartz MN. 2004. Bacterial meningitis—a view of the past 90 years. N Engl J Med 351:1826–1828. doi:10.1056/NEJMp048246.
    1. Serruto D, Rappuoli R, Scarselli M, Gros P, van Strijp JA. 2010. Molecular mechanisms of complement evasion: learning from staphylococci and meningococci. Nat Rev Microbiol 8:393–399. doi:10.1038/nrmicro2366.
    1. Del Tordello E, Vacca I, Ram S, Rappuoli R, Serruto D. 2014. Neisseria meningitidis NalP cleaves human complement C3, facilitating degradation of C3b and survival in human serum. Proc Natl Acad Sci U S A 111:427–432. doi:10.1073/pnas.1321556111.
    1. Weiss J, Elsbach P, Shu C, Castillo J, Grinna L, Horwitz A, Theofan G. 1992. Human bactericidal/permeability-increasing protein and a recombinant NH2-terminal fragment cause killing of serum-resistant gram-negative bacteria in whole blood and inhibit tumor necrosis factor release induced by the bacteria. J Clin Invest 90:1122–1130. doi:10.1172/JCI115930.
    1. Teng NN, Kaplan HS, Hebert JM, Moore C, Douglas H, Wunderlich A, Braude AI. 1985. Protection against gram-negative bacteremia and endotoxemia with human monoclonal IgM antibodies. Proc Natl Acad Sci U S A 82:1790–1794. doi:10.1073/pnas.82.6.1790.
    1. Levin M, Quint PA, Goldstein B, Barton P, Bradley JS, Shemie SD, Yeh T, Kim SS, Cafaro DP, Scannon PJ, Giroir BP. 2000. Recombinant bactericidal/permeability-increasing protein (rBPI21) as adjunctive treatment for children with severe meningococcal sepsis: a randomised trial. rBPI21 Meningococcal Sepsis Study Group. Lancet 356:961–967. doi:10.1016/S0140-6736(00)02712-4.
    1. Derkx B, Wittes J, McCloskey R. 1999. Randomized, placebo-controlled trial of HA-1A, a human monoclonal antibody to endotoxin, in children with meningococcal septic shock. European Pediatric Meningococcal Septic Shock Trial Study Group. Clin Infect Dis 28:770–777. doi:10.1086/515184.
    1. Brouwer MC, McIntyre P, Prasad K, van de Beek D. 2015. Corticosteroids for acute bacterial meningitis. Cochrane Database Syst Rev 12:CD004405. doi:10.1002/14651858.CD004405.pub5.
    1. Tolaj I, Dreshaj S, Qehaja E, Tolaj J, Doda-Ejupi T, Mehmeti M. 2010. Dexamethasone as adjuvant therapy in the treatment of invasive meningococcal diseases. Med Arh 64:228–230.
    1. Johswich K, Martin M, Bleich A, Kracht M, Dittrich-Breiholz O, Gessner JE, Suerbaum S, Wende E, Rheinheimer C, Klos A. 2009. Role of the C5a receptor (C5aR) in acute and chronic dextran sulfate-induced models of inflammatory bowel disease. Inflamm Bowel Dis 15:1812–1823. doi:10.1002/ibd.21012.
    1. Brennan FH, Lee JD, Ruitenberg MJ, Woodruff TM. 2016. Therapeutic targeting of complement to modify disease course and improve outcomes in neurological conditions. Semin Immunol 28:292–308. doi:10.1016/j.smim.2016.03.015.
    1. Arumugam TV, Magnus T, Woodruff TM, Proctor LM, Shiels IA, Taylor SM. 2006. Complement mediators in ischemia-reperfusion injury. Clin Chim Acta 374:33–45. doi:10.1016/j.cca.2006.06.010.
    1. Riedemann NC, Guo RF, Ward PA. 2003. Novel strategies for the treatment of sepsis. Nat Med 9:517–524. doi:10.1038/nm0503-517.
    1. Woodruff TM, Pollitt S, Proctor LM, Stocks SZ, Manthey HD, Williams HM, Mahadevan IB, Shiels IA, Taylor SM. 2005. Increased potency of a novel complement factor 5a receptor antagonist in a rat model of inflammatory bowel disease. J Pharmacol Exp Ther 314:811–817. doi:10.1124/jpet.105.086835.
    1. Baelder R, Fuchs B, Bautsch W, Zwirner J, Köhl J, Hoymann HG, Glaab T, Erpenbeck V, Krug N, Braun A. 2005. Pharmacological targeting of anaphylatoxin receptors during the effector phase of allergic asthma suppresses airway hyperresponsiveness and airway inflammation. J Immunol 174:783–789. doi:10.4049/jimmunol.174.2.783.
    1. Strachan AJ, Woodruff TM, Haaima G, Fairlie DP, Taylor SM. 2000. A new small molecule C5a receptor antagonist inhibits the reverse-passive Arthus reaction and endotoxic shock in rats. J Immunol 164:6560–6565. doi:10.4049/jimmunol.164.12.6560.
    1. Flierl MA, Stahel PF, Rittirsch D, Huber-Lang M, Niederbichler AD, Hoesel LM, Touban BM, Morgan SJ, Smith WR, Ward PA, Ipaktchi K. 2009. Inhibition of complement C5a prevents breakdown of the blood-brain barrier and pituitary dysfunction in experimental sepsis. Crit Care 13:R12. doi:10.1186/cc7710.
    1. Riedemann NC, Guo RF, Neff TA, Laudes IJ, Keller KA, Sarma VJ, Markiewski MM, Mastellos D, Strey CW, Pierson CL, Lambris JD, Zetoune FS, Ward PA. 2002. Increased C5a receptor expression in sepsis. J Clin Invest 110:101–108. doi:10.1172/JCI15409.
    1. Rittirsch D, Flierl MA, Nadeau BA, Day DE, Huber-Lang M, Mackay CR, Zetoune FS, Gerard NP, Cianflone K, Köhl J, Gerard C, Sarma JV, Ward PA. 2008. Functional roles for C5a receptors in sepsis. Nat Med 14:551–557. doi:10.1038/nm1753.
    1. Huber-Lang MS, Sarma JV, McGuire SR, Lu KT, Guo RF, Padgaonkar VA, Younkin EM, Laudes IJ, Riedemann NC, Younger JG, Ward PA. 2001. Protective effects of anti-C5a peptide antibodies in experimental sepsis. FASEB J 15:568–570. doi:10.1096/fj.00-0653fje.
    1. Köhl J. 2006. Drug evaluation: the C5a receptor antagonist PMX-53. Curr Opin Mol Ther 8:529–538.
    1. Woodruff TM, Nandakumar KS, Tedesco F. 2011. Inhibiting the C5-C5a receptor axis. Mol Immunol 48:1631–1642. doi:10.1016/j.molimm.2011.04.014.
    1. McGuinness BT, Clarke IN, Lambden PR, Barlow AK, Poolman JT, Jones DM, Heckels JE. 1991. Point mutation in meningococcal por A gene associated with increased endemic disease. Lancet 337:514–517. doi:10.1016/0140-6736(91)91297-8.
    1. Lappann M, Haagensen JA, Claus H, Vogel U, Molin S. 2006. Meningococcal biofilm formation: structure, development and phenotypes in a standardized continuous flow system. Mol Microbiol 62:1292–1309. doi:10.1111/j.1365-2958.2006.05448.x.
    1. Steeghs L, den Hartog R, den Boer A, Zomer B, Roholl P, van der Ley P. 1998. Meningitis bacterium is viable without endotoxin. Nature 392:449–450. doi:10.1038/33046.
    1. Johswich KO, McCaw SE, Islam E, Sintsova A, Gu A, Shively JE, Gray-Owen SD. 2013. In vivo adaptation and persistence of Neisseria meningitidis within the nasopharyngeal mucosa. PLoS Pathog 9:e1003509. doi:10.1371/journal.ppat.1003509.

Source: PubMed

3
Suscribir