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

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 succ...

Full description

Bibliographic Details
Main Authors: Johannes B. Herrmann, Marcel Muenstermann, Lea Strobel, Alexandra Schubert-Unkmeir, Trent M. Woodruff, Scott D. Gray-Owen, Andreas Klos, Kay O. Johswich, Rino Rappuoli
Format: Article
Language:English
Published: American Society for Microbiology 2018-01-01
Series:mBio
Online Access:http://mbio.asm.org/cgi/content/full/9/1/e01755-17
id doaj-e787bbe51fa4490788ecc110f7fb57e5
record_format Article
spelling doaj-e787bbe51fa4490788ecc110f7fb57e52021-07-02T04:17:10ZengAmerican Society for MicrobiologymBio2150-75112018-01-0191e01755-1710.1128/mBio.01755-17Complement C5a Receptor 1 Exacerbates the Pathophysiology of N. meningitidis Sepsis and Is a Potential Target for Disease TreatmentJohannes B. HerrmannMarcel MuenstermannLea StrobelAlexandra Schubert-UnkmeirTrent M. WoodruffScott D. Gray-OwenAndreas KlosKay O. JohswichRino RappuoliSepsis 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.http://mbio.asm.org/cgi/content/full/9/1/e01755-17
collection DOAJ
language English
format Article
sources DOAJ
author Johannes B. Herrmann
Marcel Muenstermann
Lea Strobel
Alexandra Schubert-Unkmeir
Trent M. Woodruff
Scott D. Gray-Owen
Andreas Klos
Kay O. Johswich
Rino Rappuoli
spellingShingle Johannes B. Herrmann
Marcel Muenstermann
Lea Strobel
Alexandra Schubert-Unkmeir
Trent M. Woodruff
Scott D. Gray-Owen
Andreas Klos
Kay O. Johswich
Rino Rappuoli
Complement C5a Receptor 1 Exacerbates the Pathophysiology of N. meningitidis Sepsis and Is a Potential Target for Disease Treatment
mBio
author_facet Johannes B. Herrmann
Marcel Muenstermann
Lea Strobel
Alexandra Schubert-Unkmeir
Trent M. Woodruff
Scott D. Gray-Owen
Andreas Klos
Kay O. Johswich
Rino Rappuoli
author_sort Johannes B. Herrmann
title Complement C5a Receptor 1 Exacerbates the Pathophysiology of N. meningitidis Sepsis and Is a Potential Target for Disease Treatment
title_short Complement C5a Receptor 1 Exacerbates the Pathophysiology of N. meningitidis Sepsis and Is a Potential Target for Disease Treatment
title_full Complement C5a Receptor 1 Exacerbates the Pathophysiology of N. meningitidis Sepsis and Is a Potential Target for Disease Treatment
title_fullStr Complement C5a Receptor 1 Exacerbates the Pathophysiology of N. meningitidis Sepsis and Is a Potential Target for Disease Treatment
title_full_unstemmed Complement C5a Receptor 1 Exacerbates the Pathophysiology of N. meningitidis Sepsis and Is a Potential Target for Disease Treatment
title_sort complement c5a receptor 1 exacerbates the pathophysiology of n. meningitidis sepsis and is a potential target for disease treatment
publisher American Society for Microbiology
series mBio
issn 2150-7511
publishDate 2018-01-01
description 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.
url http://mbio.asm.org/cgi/content/full/9/1/e01755-17
work_keys_str_mv AT johannesbherrmann complementc5areceptor1exacerbatesthepathophysiologyofnmeningitidissepsisandisapotentialtargetfordiseasetreatment
AT marcelmuenstermann complementc5areceptor1exacerbatesthepathophysiologyofnmeningitidissepsisandisapotentialtargetfordiseasetreatment
AT leastrobel complementc5areceptor1exacerbatesthepathophysiologyofnmeningitidissepsisandisapotentialtargetfordiseasetreatment
AT alexandraschubertunkmeir complementc5areceptor1exacerbatesthepathophysiologyofnmeningitidissepsisandisapotentialtargetfordiseasetreatment
AT trentmwoodruff complementc5areceptor1exacerbatesthepathophysiologyofnmeningitidissepsisandisapotentialtargetfordiseasetreatment
AT scottdgrayowen complementc5areceptor1exacerbatesthepathophysiologyofnmeningitidissepsisandisapotentialtargetfordiseasetreatment
AT andreasklos complementc5areceptor1exacerbatesthepathophysiologyofnmeningitidissepsisandisapotentialtargetfordiseasetreatment
AT kayojohswich complementc5areceptor1exacerbatesthepathophysiologyofnmeningitidissepsisandisapotentialtargetfordiseasetreatment
AT rinorappuoli complementc5areceptor1exacerbatesthepathophysiologyofnmeningitidissepsisandisapotentialtargetfordiseasetreatment
_version_ 1721340521451880448