ArticlePDF Available

Kineret (R)/IL-1ra Blocks the IL-1/IL-8 Inflammatory Cascade during Recombinant Panton Valentine Leukocidin-Triggered Pneumonia but Not during S-aureus Infection

Authors:

Abstract and Figures

Objectives Community-acquired Staphylococcus aureus necrotizing pneumonia is a life-threatening disease. Panton Valentine Leukocidin (PVL) has been associated with necrotizing pneumonia. PVL triggers inflammasome activation in human macrophages leading to IL-1β release. IL-1β activates lung epithelial cells to release IL-8. This study aimed to assess the relevance of this inflammatory cascade in vivo and to test the potential of an IL-1 receptor antagonist (IL-1Ra/Kineret) to decrease inflammation-mediated lung injury. Methods We used the sequential instillation of Heat-killed S. aureus and PVL or S. aureus infection to trigger necrotizing pneumonia in rabbits. In these models, we investigated inflammation in the presence or absence of IL-1Ra/Kineret. Results We demonstrated that the presence of PVL was associated with IL-1β and IL-8 release in the lung. During PVL-mediated sterile pneumonia, Kineret/IL-1Ra reduced IL-8 production indicating the relevance of the PVL/IL-1/IL-8 cascade in vivo and the potential of Kineret/IL-1Ra to reduce lung inflammation. However, Kineret/IL-1Ra was ineffective in blocking IL-8 production during infection with S. aureus. Furthermore, treatment with Kineret increased the bacterial burden in the lung. Conclusions Our data demonstrate PVL-dependent inflammasome activation during S.aureus pneumonia, indicate that IL-1 signaling controls bacterial burden in the lung and suggest that therapy aimed at targeting this pathway might be deleterious during pneumonia.
Content may be subject to copyright.
Kineret
H
/IL-1ra Blocks the IL-1/IL-8 Inflammatory Cascade
during Recombinant Panton Valentine Leukocidin-
Triggered Pneumonia but Not during
S. aureus
Infection
Delphine Labrousse
1.
, Magali Perret
2,3,4,5.
, Davy Hayez
1
, Sonia Da Silva
1
,Ce
´dric Badiou
2,3,4,5
,
Florence Couzon
2,3,4,5
, Miche
`le Bes
2,3,4,5
, Pascal Chavanet
6
,Ge
´rard Lina
2,3,4,5
, Franc¸ois Vandenesch
2,3,4,5
,
Delphine Croisier-Bertin
1,6
, Thomas Henry
2,3,4,5
*
1Vivexia, Dijon, France, 2Universite
´de Lyon, Lyon, France, 3INSERM U1111, Lyon, France, 4CNRS, UMR 5308, Lyon, France, 5Ecole Normale Supe
´rieure, Lyon, France,
6Infectious Diseases Department, University Hospital, Dijon, France
Abstract
Objectives:
Community-acquired Staphylococcus aureus necrotizing pneumonia is a life-threatening disease. Panton
Valentine Leukocidin (PVL) has been associated with necrotizing pneumonia. PVL triggers inflammasome activation in
human macrophages leading to IL-1brelease. IL-1bactivates lung epithelial cells to release IL-8. This study aimed to assess
the relevance of this inflammatory cascade in vivo and to test the potential of an IL-1 receptor antagonist (IL-1Ra/Kineret) to
decrease inflammation-mediated lung injury.
Methods:
We used the sequential instillation of Heat-killed S. aureus and PVL or S. aureus infection to trigger necrotizing
pneumonia in rabbits. In these models, we investigated inflammation in the presence or absence of IL-1Ra/Kineret.
Results:
We demonstrated that the presence of PVL was associated with IL-1band IL-8 release in the lung. During PVL-
mediated sterile pneumonia, Kineret/IL-1Ra reduced IL-8 production indicating the relevance of the PVL/IL-1/IL-8 cascade in
vivo and the potential of Kineret/IL-1Ra to reduce lung inflammation. However, Kineret/IL-1Ra was ineffective in blocking IL-
8 production during infection with S. aureus. Furthermore, treatment with Kineret increased the bacterial burden in the
lung.
Conclusions:
Our data demonstrate PVL-dependent inflammasome activation during S.aureus pneumonia, indicate that IL-1
signaling controls bacterial burden in the lung and suggest that therapy aimed at targeting this pathway might be
deleterious during pneumonia.
Citation: Labrousse D, Perret M, Hayez D, Da Silva S, Badiou C, et al. (2014) Kineret
H
/IL-1ra Blocks the IL-1/IL-8 Inflammatory Cascade during Recombinant Panton
Valentine Leukocidin-Triggered Pneumonia but Not during S. aureus Infection. PLoS ONE 9(6): e97546. doi:10.1371/journal.pone.0097546
Editor: Suzan H. M. Rooijakkers, University Medical Center Utrecht, Netherlands
Received November 13, 2013; Accepted April 17, 2014; Published June 6, 2014
Copyright: ß2014 Labrousse et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: This work was supported by a FINOVI young investigator grant to TH. SOBI had no role in study design, data collection and analysis, decision to
publish, or preparation of the manuscript.
Competing Interests: Kineret H113/IL-1Ra employed in the study was provided by SOBI ß, SOBI had no role in study design, data collection and analysis,
decision to publish or preparation of the manuscript. Authors DL, DH, SDS and DCB are employees of Vivexia, a company specialized in the evaluation of the
efficacy of anti-infectious drug candidates. This does not alter the authors’ adherence to PLOS ONE policies on sharing data and materials.
* E-mail: Thomas.henry@inserm.fr
.These authors contributed equally to this work.
Introduction
The prevalence of community-acquired Staphylococcus aureus
pneumonia is low, but the disease can be very severe, with lethality
higher than 40% in children and young adults [1,2]. Due to the
spread of community-acquired methicillin-resistant S. aureus (CA-
MRSA) and the increased resistance of these strains to antibiotics,
it is crucial to understand the pathophysiological mechanisms at
play during severe CA-S. aureus pneumonia and to find novel
therapeutic options.
Panton Valentine Leukocidin (PVL) is a bi-component leuko-
toxin composed of LukS-PV and LukF-PV. PVL is very cytotoxic
to human neutrophils, monocytes and macrophages. Furthermore,
PVL triggers the production of IL-8 [3] by neutrophils and of IL-
1bby monocytes and macrophages [4,5]. We have recently shown
that IL-1breleased by rPVL-intoxicated macrophages activates
lung epithelial cells to release large amounts of IL-8. IL-1band IL-
8 are key cytokines to recruit neutrophils [6]. This inflammatory
cascade could thus contribute to acute lung inflammation observed
during infection. While inflammation is important to clear
bacteria, it can be detrimental to the host by triggering tissue
damage. Indeed, Diep et al. demonstrated that PVL was
associated with increased inflammation and neutrophil recruit-
ment, both of which trigger lung injury [7].
Kineret, also known as Anakinra, is a drug used to treat
rheumatoid arthritis and several inflammasome-related diseases
[8]. Kineret is a recombinant form of the naturally occurring IL-1
receptor antagonist (IL-1Ra). Kineret competes with the IL-1
PLOS ONE | www.plosone.org 1 June 2014 | Volume 9 | Issue 6 | e97546
receptor for the binding of IL-1aand IL-1b. The safety of Kineret
is well-characterized, thus allowing the drug to be used to treat
other diseases [9].
In this work, we first characterized IL-8 secretion by human
neutrophils, macrophages and lung epithelial cells in response to
PVL and toxin-containing bacterial supernatant in vitro. We then
performed an in vivo study with two specific aims: i) To assess
whether inflammasome activation and the rPVL/IL-1/IL-8
inflammatory cascade were relevant during pneumonia. ii) To
test whether Kineret/IL-1Ra could block this cascade and
alleviate lung inflammation and injury.
Our results confirmed that PVL is a virulence factor that
contributes to lung inflammation. Furthermore, we demonstrated
that the instillation of heat-killed S. aureus (HKS) and rPVL,
Kineret/IL-1ra reduced PVL-mediated IL-8 secretion, thus
indicating the functionality of the rPVL/IL-1/IL-8 cascade in
vivo. However, during infection with PVL
+
S. aureus, we found that
Kineret/IL-1ra had no effect on IL-8 levels, suggesting that other
inflammatory mechanisms were at play. Finally, treatment with
Kineret/IL-1ra increased bacterial replication in the lung,
indicating that the IL-1 inflammatory pathway contributed to
bacterial clearance. This latter result highlights the possible caveat
of targeting inflammatory pathways and indicates that if such a
therapeutic option were chosen, it could only be applied in the
presence of a potent adjunctive antibiotic therapy.
Materials and Methods
Ethics Statement
The experimental protocol was approved by the local ethics
committee for animal experimentation (Burgundy, Dijon, France-
protocol number CE 1311). Blood packs from healthy donors were
obtained anonymously through an agreement (InsermU1111/
11_02_2013) with the Etablissement Franc¸ais du Sang (EFS,
Lyon, France) in accordance with French laws #98-535 and 99-
1143 and with the principles of the Declaration of Helsinki. Each
donor provided written informed consent, which is available upon
request. The human cell lines (THP-1 and A549) used in this study
were previously described [4].
Bacterial strains, culture conditions and reagents
The MRSA Staphylococcus aureus USA 300 PVL
+
Los Angeles
Clone 0114 (hereafter termed USA300 LAC), SF8300 (BD425)
and their isogenic PVL
2
derivative strains (HT20060753 and
BD0452) kindly provided by Franck DeLeo [10] and Binh Diep
[7] were grown in CCY broth, which promotes the production of
leukocidins. The clinical MRSA strain ST20120376 (a clonal
complex CC121 PVL
+
strain) was isolated from a patient suffering
from necrotizing pneumonia. Supernatant was prepared from a
16 h culture and diluted 625 times before addition to cells. rPVL
was produced and purified as previously described [4].
Cells, culture conditions and in vitro tests
Human neutrophils were purified as previously described [11].
When applicable, Heat-killed S. aureus (HKS MOI 100:1) and
Kineret were added to the neutrophils 3 h before rPVL
intoxication.
THP-1 cells and A549 cells were cultured as previously
described [4]. For the mixed culture experiment, 10
5
A549 cells
were added to 10
3
PMA-differentiated THP-1 cells 24 h before
the addition of PVL. HKS was added for 16 h before cell
intoxication.
Production and treatment of necrotizing pneumonia in
rabbits.
Animals. Male New Zealand rabbits were bred and housed
at the Zootechnical Center (University of Burgundy, Dijon,
France) in accordance with current European Institute of Health
guidelines.
Inoculum. USA 300 LAC and its isogenic PVL mutant were
grown in CCY medium supplemented with pyruvic acid for
10 hours. The inoculum was adjusted to 3610
9
CFU/mL.
Rabbit Model of Necrotizing Pneumonia. The central
venous catheters were installed and the pneumonia model was
established as previously described [12,13]. Bacterial pneumonia
was induced by endobronchial challenge with 0.5 mL of saline
containing 3610
9
CFU/mL. rPVL-induced pneumonia was
achieved by endotracheal instillation of 12 mg each of LukS-PV
and LukF-PV 3 hours after the delivery of 0.5 mL of HKS.
Kineret/IL-1Ra was kindly provided EXW by SOBI
ß
and
administered at doses of 10 mg/kg (Fig. 1).
Evaluation of necrotizing pneumonia
Bacterial counts. The rabbits were euthanized 6 hours after
challenge (Fig. 1). Each pulmonary lobe was weighed and
homogenized in 5 mL of sterile saline for bacterial counts.
Bacterial concentrations in each lobe were determined after
adjusting for weight. The threshold value was 1 log
10
CFU/g.
Macroscopic scoring. Macroscopic pulmonary injury scores
were calculated according to a macroscopic scoring grid as
previously described [14].
Bronchoalveolar Lavage Fluid. At the time of euthanasia
(6 h after challenge), the lungs were excised and bronchoalveolar
lavage (BAL) was performed on both inferior lobes by the
instillation of 4 ml physiological saline. Total protein concentra-
tion in the BAL fluid (BALF) was determined by the Bradford
protein assay according to the manufacturer’s instructions (Pierce).
Measurements of IL-1band IL-8. Concentrations of rabbit
IL-1band IL-8 in the BALF and in the crude homogenate of each
pulmonary lobe were assessed by ELISA (Euromedex). The release
of human IL-1band IL-8 was quantified by ELISA using DuoSet
ELISA kits (R&D systems, Lille, France).
Statistical analysis. In vitro and in vivo data were analyzed
by an unpaired t-test and Mann-Whitney analysis, respectively,
using Prism software (GraphPad, San Diego, USA). For in vitro
data, means and standard deviations are shown. Two tailed p
values are shown: n.s.: not significant, * p,0.05; ** p,0.01; ***
p,0.0001.
Results
Kineret/IL-1Ra blocks the IL-1b/IL-8 inflammatory cascade
observed in a co-culture of human macrophages and
lung epithelial cells exposed to S. aureus supernatant,
but has no effect on PVL-mediated neutrophil response
Using a co-culture system of macrophages (1%) and lung
epithelial cells (99%), we previously showed that very low
concentrations of IL-1breleased by rPVL-intoxicated macrophag-
es triggered the release of large amounts of IL-8 by lung epithelial
cells [4]. In line with this result, the addition of Kineret/IL-1Ra to
a co-culture of macrophages (1%) and lung epithelial cells (99%)
exposed to supernatant from a clinical PVL
+
S. aureus strain
isolated from a patient suffering from necrotizing pneumonia
reduced IL-8 secretion in a dose-dependent manner (Fig. 2A).
Though Kineret/IL-1Ra also reduced IL-8 levels in the same
cellular model in response to supernatant from USA300 SF8300
Kineret H/IL-1Ra in CA-MRSA-Pneumonia
PLOS ONE | www.plosone.org 2 June 2014 | Volume 9 | Issue 6 | e97546
[7] or LAC strains, we found no difference in IL-8 levels between
WT USA300 strains and their isogenic DPVL mutants (Fig. 2B
and data not shown). This result indicates that other staphylococ-
cal secreted factors besides PVL can trigger the IL-1/IL-8 cascade.
Altogether, these results suggested that the majority of IL-8
produced early on during infection with S. aureus could be
secondary to IL-1 production triggered by the exposure of
macrophages to PVL or other staphylococcal secreted factors.
However, neutrophils are rapidly recruited to the lung and rapidly
out-compete alveolar macrophages. We thus decided to quantify
the production of IL-1band IL-8 by human neutrophils treated
with rPVL. In agreement with the low ability of human
neutrophils to produce IL-1b[15,16,17], IL-1bproduction by
rPVL-intoxicated primary human neutrophils was very low even
when they were primed with HKS (Figure 2C [4]). As previously
described [3,18], human neutrophils intoxicated with rPVL at
10 mg/ml produced high levels of IL-8 (Fig. 2D). In contrast, rPVL
at 100 mg/ml did not lead to consistent IL-8 production. This was
probably due to the rapid death of neutrophils at this concentra-
tion [7,11,19,20]. We then checked whether IL-8 production by
neutrophils or by lung epithelial cells exposed to PVL-intoxicated
neutrophils could be due to IL-1 signaling. Kineret/IL-1Ra had
no impact on IL-8 production by neutrophils (Fig. 2E) or by a co-
culture of neutrophils and lung epithelial cells (not shown), thus
indicating that IL-8 production in neutrophils is independent of
the IL-1/IL-8 cascade observed in macrophages ([4] and Fig. 2A).
Altogether, these results indicated that PVL triggers different
signaling pathways in human macrophages and neutrophils.
Furthermore, these in vitro results suggested that Kineret/IL-
1Ra could block IL-8 in vivo if the inflammatory response is driven
by macrophages, with no major contribution from neutrophils.
Infection with PVL
+
S. aureus triggers IL-1band IL-8
release in a rabbit model of necrotizing pneumonia
We and others have recently identified PVL as a major
inflammasome activator in human monocytes and macrophages
[4,5]. However, whether PVL activates the inflammasome
pathway in vivo remains to be determined. The rabbit model is
a well established model to study PVL
+
S. aureus diseases
[7,18,20,21,22] and to discriminate between active and inactive
anti-infective treatments [23][13]. We thus investigated the role of
PVL and its ability to activate the inflammasome pathway in a
rabbit model of pneumonia [7]. Rabbits were inoculated
intratracheally with 1.5610
9
CFU of the MRSA USA300 LAC
strain or its isogenic DPVL mutant strain (Fig. 1). This infectious
dose led to the rapid PVL-dependent death of infected rabbits
(Figure S1), with macroscopic lesions of the lung at 15 h PI
indicating necrosis (Figure S2). Necrotizing pneumonia is a
fulminant disease. We therefore focused on early events. Infected
animals were euthanized at 6 h PI. Bronchoalveolar lavage fluid
(BALF) and lung lysates were collected to quantify cytokines.
Infection with the PVL
+
strain was associated with greater IL-1b
release in the BALF than was the case with a DPVL mutant strain
(Fig. 3A). This cytokine is notoriously difficult to detect in
biological fluids [24] suggesting that the low levels of IL-1b
Figure 1. Study protocol.
doi:10.1371/journal.pone.0097546.g001
Kineret H/IL-1Ra in CA-MRSA-Pneumonia
PLOS ONE | www.plosone.org 3 June 2014 | Volume 9 | Issue 6 | e97546
detected in the BALF of PVL
+
S. aureus infected rabbits were
probably biologically relevant. As previously described [7], PVL
+
S. aureus-mediated pneumonia was associated with a greater
increase in IL-8 levels in the BALF (Fig. 3B) and the lung lysates
than that observed during infection with the DPVL strain (Fig. 3C).
This increase in inflammatory cytokines was associated with an
increase i) in the permeability of the alveolar-capillary barrier as
measured by the protein content of the BALF (Fig. 3D), ii) in the
overall lung histopathology as determined by macroscopic scoring
(Fig. 3E), iii) in the lung weight/body weight ratio (a measurement
of pulmonary edema-Fig. 3F) and iv) in the lung bacterial burden
(Fig. 3G).
Figure 2. IL-8 produced in response to
S. aureus
supernatant by a co-culture of macrophages and lung epithelial cells is dependent
on IL-1 signaling, while IL-8 produced by neutrophils is independent of IL-1. (A, B) THP-1 macrophages (10
3
cells), A549 lung epithelial cells
(10
5
) were cultured alone or mixed and treated as indicated with HKS, S. aureus CCY broth culture supernatant and Kineret/IL-1Ra. IL-8 was quantified
by ELISA at 6 h post-intoxication. (C–E) As indicated, primary human neutrophils were treated with HKS, Kineret/IL-1Ra (E) and rPVL. IL-1b(C) and IL-8
levels were quantified at 6 h PI (E) or at the indicated time post-intoxication (C, D). One experiment representative of three independent experiments
with three independent donors is shown.
doi:10.1371/journal.pone.0097546.g002
Kineret H/IL-1Ra in CA-MRSA-Pneumonia
PLOS ONE | www.plosone.org 4 June 2014 | Volume 9 | Issue 6 | e97546
Sequential instillation of Heat-killed S. aureus and rPVL
triggers pneumonia-like symptoms associated with IL-1b
and IL-8 production.
To ensure that the differences in IL-1band IL-8 production and
lung injury was due to the presence of PVL, we investigated
whether we could reproduce this inflammation using rPVL. In
vitro, inflammasome activation in response to pore-forming toxins
requires prestimulation with TLR agonists [25,26]. We thus
adapted the protocol established by Diep and collaborators [7]
and instilled HKS followed 3 h later by rPVL intratracheally.
Rabbits were euthanized 3 h post-rPVL instillation (Fig. 1). As
described above for infection, in this sterile model, the presence of
rPVL was associated with an increase in the levels of IL-1b
(Fig. 4A) and IL-8 (Fig. 4B) in the BALF, and IL-8 (Fig. 4C) in lung
lysates. Furthermore, the increase in inflammation mirrored an
increase in the lung histopathology as determined by macroscopic
scoring (Fig. 4D) and lung edema (Fig. 4E). Interestingly, at this
time point, HKS alone had no effect on the secretion of cytokines,
but worked in synergy with rPVL to induce IL-8 and pulmonary
pathological features.
Altogether, these two models of HKS-rPVL-mediated and
bacterial pneumonia indicated that the presence of PVL was
associated in vivo with inflammasome activation and confirmed
that PVL contributed to severe inflammation and lung injury in a
rabbit model of necrotizing pneumonia [7,18].
Kineret/IL-1Ra blocks the PVL/IL-1/IL-8 inflammatory
cascade observed in a HKS-rPVL-mediated pneumonia
model
IL-1band IL-8 are two key cytokines involved in the
recruitment of neutrophils [6]. Neutrophils contribute to the
pathology of necrotizing pneumonia [7]. We thus investigated
Figure 3. PVL is associated with an increase in IL-1band IL-8 release in a rabbit model of
S. aureus
-mediated pneumonia. (A–G)
Rabbits were inoculated intratracheally with 1.5610
9
cfu of USA300 LAC WT or DPVL strains and euthanized 6 h post-infection. IL-1b(A), IL-8 levels in
BALF (B) or in lung lysates (C) were quantified by ELISA. (D) Total protein levels in BALF were quantified by a Bradford assay. (E) Gross pathological
scoring was performed. (F) The ratio of lung weight to body weight is shown. (G) Bacterial burden as determined by CFU in each rabbit lung is shown.
(A–D) Each point represents the value obtained in the BALF from one lobe or (E–G) the value obtained for one rabbit. (A–G) The geometric mean is
shown. One experiment (n = 6) representative of two experiments (n = 12) is shown.
doi:10.1371/journal.pone.0097546.g003
Kineret H/IL-1Ra in CA-MRSA-Pneumonia
PLOS ONE | www.plosone.org 5 June 2014 | Volume 9 | Issue 6 | e97546
whether Kineret/IL-1Ra was effective in vivo to decrease the IL-
1/IL-8 inflammatory cascade and the inflammation-associated
lung damages. We first tested Kineret/IL-1Ra on pneumonia
triggered by sequential instillation of HKS and rPVL. To ensure
the highest potency of this drug, we administered Kineret IV 2 h
post-HKS treatment as well as in co-instillation with rPVL (Fig. 1).
Kineret was used at 10 mg/kg [27]. Interestingly and as observed
in vitro using human macrophages, treatment with Kineret
reduced IL-8 levels in both BALF (Fig. 5A) and lung lysates
(Fig. 5B). The reduction in IL-8 levels in lung lysate was much
greater than in BALF possibly due to the poor bioavailability of
IL-1Ra in the lung lumen. The inhibition of IL-1 signaling and IL-
8 production did not decrease the macroscopic pathological score
(Fig. 5C), edema formation (Fig. 5D) or the permeability of the
alveolar-capillary barrier (Fig. 5E). Overall, these results demon-
strated the presence of this PVL/IL-1/IL-8 cascade in vivo in the
lung and showed that Kineret/IL-1Ra targets this pathway but
has no detectable effect on lung pathophysiology.
Kineret/IL-1Ra does not block the PVL/IL-1/IL-8
inflammatory cascade observed during lung infection
with PVL
+
S. aureus
Since treatment with Kineret/IL-1Ra led to encouraging results
on the possibility to target the IL-1/IL-8 cascade, we decided to
investigate its potency during PVL
+
S. aureus-mediated pneumonia
(Fig. 1). In contrast to what we observed in the HKS-rPVL model,
treatment with Kineret/IL-1Ra did not reduce IL-8 levels in
either BALF (Fig. 6A) or lung lysates (Fig. 6B). As previously
described in the HKS-rPVL model, treatment with Kineret/IL-
1Ra was ineffective in reducing the pathological score (Fig. 6C),
lung edema (Fig. 6D) or the permeability of the alveolar-capillary
barrier (Fig. 6E). However, treatment with Kineret/IL-1Ra did
result in a significant increase in the bacterial burden per lung
(Fig. 6F) indicating that IL-1 signaling contributes to antibacterial
defenses during necrotizing pneumonia.
Figure 4. Sequential intratracheal instillation of HKS and rPVL triggers IL-1band IL-8 release and reproduces some key aspects of
S.
aureus
-mediated pneumonia. (A–E) Rabbits were inoculated intratracheally with HKS, 3 h later with PBS or with rPVL and euthanized 6 h post-HKS
instillation. IL-1b(A) and IL-8 levels in BALF (B) or in lung lysates (C) were quantified by ELISA. (D) Gross pathological scoring was performed. (E) The
ratio of lung weight on body weight is shown. (A–C) Each point represents the value obtained in the BALF from one lobe or (D, E) the value obtained
for one rabbit. (A–F) The geometric mean is shown. Results from three independent experiments (n = 12) are shown.
doi:10.1371/journal.pone.0097546.g004
Kineret H/IL-1Ra in CA-MRSA-Pneumonia
PLOS ONE | www.plosone.org 6 June 2014 | Volume 9 | Issue 6 | e97546
Discussion
CA-S. aureus necrotizing pneumonia is a severe disease with a
high percentage of fatal outcomes. The role of PVL in
experimental infections, in triggering specific human diseases or
in affecting disease outcome is still debated [28,29,30]. Here, using
infection or sequential instillations of HKS and rPVL, we
confirmed the role of PVL in triggering inflammation and lung
injury in a rabbit model of necrotizing pneumonia. Two recent
studies have described the ability of PVL to activate the
inflammasome in primary human monocytes and macrophages
[4,5]. We demonstrated in vivo that the presence of PVL was
associated with an increase in IL-1blevels in the BALF of infected
rabbits, thus highlighting the relevance of this pathway during
pneumonia. Furthermore, we observed that the blockage of IL-1
signaling using Kineret/IL-1Ra led to a ten-fold increase in the
bacterial burden in the lung of infected animals, indicating that IL-
1 is critical for the antibacterial activity during acute pneumonia.
The current guidelines for the treatment of necrotizing
pneumonia are the prompt and aggressive administration of
toxin-suppressing antibiotics such as linezolid [23,31,32,33].
Appropriate antibiotic therapy might not be sufficient in such
fulminant diseases in which inflammation is intense and at least
partly responsible for lung injury [7]. Indeed, numerous deaths
have been observed in patients treated in a timely manner with
effective antibiotic therapy [32,34]. Adjunctive anti-inflammatory
treatment is widely used in bacterial meningitis even though its
efficacy might be limited [35]. The use of non-steroidal
antiinflammatory drugs in CA pneumonia is associated with a
more complicated course [36] and the use of dexamethasone in
treating CA pneumonia is still debated [37,38]. Furthermore, CA-
pneumonia covers a wide range of diseases in terms of both the
causative pathogenic agent and severity, thus highlighting the need
to test adjunctive therapy in specific animal models of diseases.
Here, we used a rabbit model of pneumonia to test the therapeutic
potential of anti-inflammatory drugs. Due to the high sensitivity of
rabbit neutrophils to PVL [20] and as demonstrated by others
Figure 5. Kineret/IL-1Ra inhibits the IL-1/IL-8 cascade triggered by sequential intratracheal instillation of HKS and rPVL. (A–E) Rabbits
were inoculated intratracheally with HKS, 3 h later with rPVL and euthanized 6 h post-HKS instillation. When applicable, Kineret/IL-1Ra was injectedIV
at 2 h post-HKS and was co-instillated with rPVL. IL-8 levels in BALF (A) or in lung lysates (B) were quantified by ELISA. (C) Gross pathological scoring
was performed. (D) The ratio of lung weight to body weight is shown. (E) Total protein levels in BALF were quantified by Bradford assay. (A–B, E) Each
point represents the value obtained in the BALF from one lobe or (C, D) the value obtained for one rabbit. (A–E) The geometric mean is shown.
Results from two independent experiments (n = 12) are shown.
doi:10.1371/journal.pone.0097546.g005
Kineret H/IL-1Ra in CA-MRSA-Pneumonia
PLOS ONE | www.plosone.org 7 June 2014 | Volume 9 | Issue 6 | e97546
[7,18], the rabbit is a good animal model to study PVL
+
-S. aureus
pneumonia and to assess treatment efficacy. However, the
limitations of our model include the high inoculum required to
trigger robust pneumonia and the absence of an influenza-like
viral infection preceding S.aureus infection, which is observed in
most severe human clinical cases [1,2]. We found that dexameth-
asone had no effect on IL-8 or any of the other parameters
considered in our model of pneumonia (not shown). In contrast,
we demonstrated that Kineret/IL-1Ra was effective in inhibiting
the rPVL/IL-1/IL-8 cascade after sequential instillation of HKS
and rPVL. Surprisingly, this effect was not observed during
infection despite the detection of PVL-dependent IL-1band IL-8
release. Several hypotheses can explain this difference: i) while the
macrophage response may dominate in the sterile model (HKS+
rPVL), a neutrophil-based response may be predominant during
infection. Indeed, human neutrophils produced IL-8 in response to
rPVL independently of IL-1 signaling (Fig. 2E). Furthermore, IL-8
levels were strongly reduced in neutropenic rabbits [7,18]. ii) S.
aureus and leukocidins/PSMs-intoxicated neutrophils release nu-
merous proteases[39,40][41], which might degrade/inactivate
Kineret/IL-1Ra. iii) Endogenous IL-1Ra, which is up-regulated
during bacterial pneumonia [42], might mask the action of
exogenously added IL-1Ra/Kineret.
In our sterile pneumonia model, despite the efficient block of
IL-1 signaling and a reduction of 50% in IL-8 level, we found no
reduction in neutrophil infiltration into the lung (not shown). This
result suggests that neutrophil recruitment in the lung is based on
several redundant pathways [6] and would require the targeting of
numerous signaling pathways to be significantly reduced. Similar-
ly, the failure of Kineret/IL-1ra clinical trials in severe sepsis led to
the conclusion that strategies aimed at targeting a single
inflammatory mediator were unlikely to work in such complex
diseases. Importantly, such a lesson can be learned from S. aureus,
which has evolved a large number of virulence factors targeting
neutrophils [43,44].
Figure 6. Kineret/IL-1Ra does not reduce IL-8 levels during PVL
+
S. aureus
-mediated pneumonia but increases lung bacterial load.
(A–F) Rabbits were inoculated intratracheally with USA300 LAC strain and euthanized 6 h post-infection. When applicable, Kineret/IL-1Ra was injected
IV 1 h before and 3 h after infection and was co-instilled with S. aureus. IL-8 levels in BALF (A) or in lung lysates (B) were quantified by ELISA. (C) Gross
pathological scoring was performed. (D) The ratio of lung weight to body weight is shown. (E) Total protein levels in BALF were quantified by
Bradford assay. (F) Bacterial burden as determined by CFU assay is shown. (A, B, E) Each point represents the value obtained in the BALF from one
lobe or (C, D, F) the value obtained for one rabbit. (A–F) The geometric mean is shown. Results from two independent experiments (n = 12) are
shown.
doi:10.1371/journal.pone.0097546.g006
Kineret H/IL-1Ra in CA-MRSA-Pneumonia
PLOS ONE | www.plosone.org 8 June 2014 | Volume 9 | Issue 6 | e97546
Supporting Information
Figure S1 Survival curves of LAC PVL and LAC DPVL-
infected rabbits.
(PPTX)
Figure S2 Macroscopic Pulmonary Injury Scores of LAC
PVL and LAC A
¨PVL-infected rabbits.
(PPTX)
Acknowledgments
We thank Frank deLeo and Binh Diep for providing strains, Y. Jamilloux
and F. Ader for stimulating discussions. This work was performed within
the framework of the LABEX ECOFECT (ANR-11-LABX-0042) of
Universite´ de Lyon, within the program ‘‘Investissements d’Avenir’’ (ANR-
11-IDEX-0007) operated by the French National Research Agency (ANR).
We thank Philip Bastable for helpful comments on the manuscript.
Author Contributions
Conceived and designed the experiments: DL PC GL FV DCB TH.
Performed the experiments: DL MP DH SDS CB FC DCB. Analyzed the
data: DL MP CB DCB TH. Contributed reagents/materials/analysis tools:
MP MB CB DCB. Wrote the paper: TH DL DCB PC FV GL.
References
1. Li HT, Zhang TT, Huang J, Zhou YQ, Zhu JX, et al. (2011) Factors associated
with the outcome of life-threatening necrotizing pneumonia due to community-
acquired Staphylococcus aureus in adult and adolescent patients. Respiration;
international review of thoracic diseases 81: 448–460.
2. Gillet Y, Issartel B, Vanhems P, Fournet JC, Lina G, et al. (2002) Association
between Staphylococcus aureus strains carrying gene for Panton-Valentine
leukocidin and highly lethal necrotising pneumonia in young immunocompetent
patients. Lancet 359: 753–759.
3. Konig B, Koller M, Prevost G, Piemont Y, Alouf JE, et al. (1994) Activation of
human effector cells by different bacterial toxins (leukocidin, alveolysin, and
erythrogenic toxin A): generation of interleukin-8. Infection and immunity 62:
4831–4837.
4. Perret M, Badiou C, Lina G, Burbaud S, Benito Y, et al. (2012) Cross-talk
between Staphylococcus aureus leukocidins-intoxicated macrophages and lung
epithelial cells triggers chemokine secretion in an inflammasome-dependent
manner. Cellular microbiology 14: 1019–1036.
5. Holzinger D, Gieldon L, Mysore V, Nippe N, Taxman DJ, et al. (2012)
Staphylococcus aureus Panton-Valentine leukocidin induces an inflammatory
response in human phagocytes via the NLRP3 inflammasome. Journal of
leukocyte biology 92: 1069–1081.
6. Williams MR, Azcutia V, Newton G, Alcaide P, Luscinskas FW (2011)
Emerging mechanisms of neutrophil recruitment across endothelium. Trends in
immunology 32: 461–469.
7. Diep BA, Chan L, Tattevin P, Kajikawa O, Martin TR, et al. (2010)
Polymorphonuclear leukocytes mediate Staphylococcus aureus Panton-Valen-
tine leukocidin-induced lung inflammation and injury. Proceedings of the
National Academy of Sciences of the United States of America 107: 5587–5592.
8. Dinarello CA, Simon A, van der Meer JW (2012) Treating inflammation by
blocking interleukin-1 in a broad spectrum of diseases. Nature reviews Drug
discovery 11: 633–652.
9. Giampietro C, Ridene M, Lequerre T, Costedoat Chalumeau N, Amoura Z, et
al. (2013) Anakinra in adult-onset Still’s disease: long-term treatment in patients
resistant to conventional therapy. Arthritis care & research 65: 822–826.
10. Voyich JM, Otto M, Math ema B, Braughton KR, Whitney AR, et al. (2006) Is
Panton-Valentine leukocidin the major virulence determinant in community-
associated methicillin-resistant Staphylococcus aureus disease? The Journal of
infectious diseases 194: 1761–1770.
11. Spaan AN, Henry T, van Rooijen WJ, Perret M, Badiou C, et al. (2013) The
staphylococcal toxin panton-valentine leukocidin targets human c5a receptors.
Cell host & microbe 13: 584–594.
12. Croisier-Bertin D, Piroth L, Charles PE, Larribeau A, Biek D, et al. (2011)
Ceftaroline versus ceftriaxone in a highly penicillin-resistant pneumococcal
pneumonia rabbit model using simulated human dosing. Antimicrobial agents
and chemotherapy 55: 3557–3563.
13. Croisier-Bertin D, Hayez D, Da Silva S, Labrousse D, Biek D, et al. (2014) In
Vivo Efficacy of Ceftaroline Fosamil in a Methicillin-Resistant Pvl-Producing
Staphylococcus Aureus Rabbit Pneumonia Model. Antimicrobial agents and
chemotherapy.
14. Piroth L, Martin L, Coulon A, Lequeu C, Duong M, et al. (1999) Development
of a new experimental model of penicillin-resistant Streptococcus pneumoniae
pneumonia and amoxicillin treatment by reproducing human pharmacokinetics.
Antimicrobial agents and chemotherapy 43: 2484–2492.
15. Schroder AK, von der Ohe M, Kolling U, Altstaedt J, Uciechowski P, et al.
(2006) Polymorphonuclear leucocytes selectively produce anti-inflammatory
interleukin-1 receptor antagonist and chemokines, but fail to produce pro-
inflammatory mediators. Immunology 119: 317–327.
16. Malyak M, Smith MF Jr, Abel AA, Arend WP (1994) Peripheral blood
neutrophil production of interleukin-1 receptor antagonist and interleukin-1
beta. Journal of clinical immunology 14: 20–30.
17. Langereis JD, Oudijk EJ, Schweizer RC, Lammers JW, Koenderman L, et al.
(2011) Steroids induce a disequilibrium of secreted interleukin-1 receptor
antagonist and interleukin-1beta synthesis by human neutrophils. The European
respiratory journal : official journal of the European Society for Clinical
Respiratory Physiology 37: 406–415.
18. Ma X, Chang W, Zhang C, Zhou X, Yu F (2012) Staphy lococcal Panton-
Valentine leukocidin induces pro-inflammatory cytokine production and nuclear
factor-kappa B activation in neutrophils. PLoS One 7: e34970.
19. Niemann S, Ehrhardt C, Medina E, Warnking K, Tuchscherr L, et al. (2012)
Combined action of influenza virus and Staphylococcus aureus panton-valentine
leukocidin provokes severe lung epithelium damage. The Journal of infectious
diseases 206: 1138–1148.
20. Loffler B, Hussain M, Grundmeier M, Bruck M, Holzinger D, et al. (2010)
Staphylococcus aureus panton-valentine leukocidin is a very potent cytotoxic
factor for human neutrophils. PLoS Pathog 6: e1000715.
21. Prevost G, Cribier B, Couppie P, Petiau P, Supersac G, et al. (1995) Panton-
Valentine leucocidin and gamma-hemolysin from Staphylococcus aureus ATCC
49775 are encoded by distinct genetic loci and have different biolog ical activities.
Infection and immunity 63: 4121–4129.
22. Cremieux AC, Dumitrescu O, Lina G, Vallee C, Cote JF, et al. (2009) Panton-
valentine leukocidin enhances the severity of community-associated methicillin-
resistant Staphylococcus aureus rabbit osteomyelitis. PLoS One 4: e7204.
23. Diep BA, Afasizheva A, Le HN, Kajikawa O, Matute-Bello G, et al. (2013)
Effects of Linezolid on Suppressing In Vivo Production of Staphylococcal Toxins
and Improving Survival Outcomes in a Rabbit Model of Methicillin-Resist ant
Staphylococcus aureus Necrotizing Pneumonia. The Journal of infectious
diseases.
24. Hoffman HM, Rosengren S, Boyle DL, Cho JY, Nayar J, et al. (2004)
Prevention of cold-associated acute inflammation in familial cold autoinflam-
matory syndrome by interleukin-1 receptor antagonist. Lancet 364: 1779–1785.
25. Juliana C, Fernandes-Alnemri T, Kang S, Farias A, Qin F, et al. (2012) Non-
transcriptional Priming and Deubiquitination Regulate NLRP3 Inflammasome
Activation. The Journal of biological chemistry 287: 36617–36622.
26. Bauernfeind FG, Horvath G, Stutz A, Alnemri ES, MacDonald K, et al. (2009)
Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors
license NLRP3 inflammasome activation by regulating NLRP3 expression.
Journal of immunology 183: 787–791.
27. McDuffie RS, Jr., Davies JK, Leslie KK, Lee S, Sherman MP, et al. (2001) A
randomized controlled trial of interleukin-1 receptor antagonist in a rabbit
model of ascending infection in pregnancy. Infectious diseases in obstetrics and
gynecology 9: 233–237.
28. Shallcross LJ, Fragaszy E, Johnson AM, Hayward AC (2012) The role of the
Panton-Valentine leucocidin toxin in staphylococcal disease: a systematic review
and meta-analysis. The Lancet infectious diseases.
29. Day NP (2012) Panton-Valentine leucocidin and staphylococcal disease. The
Lancet infectious diseases.
30. Diep BA, Gillet Y, Etienne J, Lina G, Vandenesch F (2013) Response to
Shallcross et al. : Panton-Valentine Leukocidin and pneumonia: does infrequent
means not severe? Lancet Infect Dis In Press.
31. Rouzic N, Janvier F, Libert N, Javouhey E, Lina G, et al. (2010) Prompt and
successful toxin-targeting treatment of three patients with necrotizing pneumo-
nia due to Staphylococcus a ureus strains carrying the Panton-Valentine
leukocidin genes. Journal of clinical microbiology 48: 1952–1955.
32. Morgan MS (2007) Diagnosis and treatment of Panton-Valentine leukocidin
(PVL)-associated staphylococcal pneumonia. International journal of antimicro-
bial agents 30: 289–296.
33. Gillet Y, Dumitrescu O, Tristan A, Dauwalder O, Javouhey E, et al. (2011)
Pragmatic management of Panton-Valentine leukocidin-associated staphylococ-
cal diseases. International journal of antimicrobial agents 38: 457–464.
34. Wargo KA, Eiland EH 3rd (2005) Appropriate antimicrobial therapy for
community-acquired methicillin-resistant Staphylococcus aureus carrying the
Panton-Valentine leukocidin genes. Clin Infect Dis 40: 1376–1378; author reply
1378–1379.
35. van de Beek D, Brouwer MC, Thwaites GE, Tunkel AR (2012) Advances in
treatment of bacterial meningitis. Lancet 380: 1693–1702.
36. Voiriot G, Dury S, Parrot A, Mayaud C, Fartoukh M (2011) Nonsteroidal
antiinflammatory drugs may affect the presentation and course of community-
acquired pneumonia. Chest 139: 387–394.
Kineret H/IL-1Ra in CA-MRSA-Pneumonia
PLOS ONE | www.plosone.org 9 June 2014 | Volume 9 | Issue 6 | e97546
37. Joseph L, Goldberg S, Picard E (2011) Dexa methasone in community-acquired
pneumonia. Lancet 378: 980; author reply 981.
38. Meijvis SC, Hardeman H, Remmelts HH, Heijligenberg R, Rijkers GT, et al.
(2011) Dexamethasone and length of hospital stay in patients with community-
acquired pneumonia: a randomised, double-blind, placebo-controlled trial.
Lancet 377: 2023–2030.
39. Surewaard BG, de Haas CJ, Vervoort F, Rigby KM, DeLeo FR, et al. (2013)
Staphylococcal alpha-phenol soluble modulins contribute to neutrophil lysis after
phagocytosis. Cellular microbiology 15: 1427–1437.
40. Malachowa N, Kobayashi SD, Freedman B, Dorward DW, DeLeo FR (2013)
Staphylococcus aureus leukotoxin GH promotes formation of neutrophil
extracellular traps. Journal of immunology 191: 6022–6029.
41. Dubin G (2002) Extracellular proteases of Staphylococcus spp. Biological
chemistry 383: 1075–1086.
42. Herold S, Tabar TS, Janssen H, Hoegner K, Cabanski M, et al. (2011) Exudate
macrophages attenuate lung injury by the release of IL-1 receptor antagonist in
gram-negative pneumonia. American journal of respiratory and critical care
medicine 183: 1380–1390.
43. Vandenesch F, Lina G, Henry T (2012) Staphylococcus aureus Hemolysins, bi-
component Leukocidins, and Cytolytic Peptides: A Redundant Arsenal of
Membrane-Damaging Virulence Factors? Frontiers in cellular and infection
microbiology 2: 12.
44. van Wamel WJ, Rooijakkers SH, Ruyken M, van Kessel KP, van Strijp JA
(2006) The innate immune modulators staphylococcal complement inhibitor and
chemotaxis inhibitory protein of Staphylococcus aureus are located on beta-
hemolysin-converting bacteriophages. Journal of bacteriology 188: 1310–1315.
Kineret H/IL-1Ra in CA-MRSA-Pneumonia
PLOS ONE | www.plosone.org 10 June 2014 | Volume 9 | Issue 6 | e97546
... 16 In addition, recombinant PVL triggers the secretion of the proinflammatory IL-1 via the NOD-like receptor protein 3 (NLRP3) inflammasome in macrophages, which contributes to lung tissue damage in rabbits, to a similar extent as PVLexpressing S. aureus. 10,14,17,18 Dampening inflammation may thus be beneficial, particularly as IL-1 administration increases and inhibition of NLRP3 decreases S. aureus lung burdens in mice. [19][20][21] PVL, however, does not affect lung IL-1 levels in humanized mice after S. aureus infection, 16 and preventing IL-1 signaling can lead to increased S. aureus lung burdens in rabbits. ...
... [19][20][21] PVL, however, does not affect lung IL-1 levels in humanized mice after S. aureus infection, 16 and preventing IL-1 signaling can lead to increased S. aureus lung burdens in rabbits. 18 The conflicting outcomes in S. aureus infection studies are related to the discovery that leukocidins such as PVL and LukAB recognize human but not mouse cell surface receptors. 22,23 The PVL subunits S and F interact with complement C5a receptor 1 (C5aR1) 23,24 and protein tyrosine phosphatase receptor type C (also known as CD45), 25 respectively, whereas LukAB binds CD11b. ...
Article
Staphylococcus aureus causes necrotizing pneumonia by secreting toxins such as leukocidins that target front‐line immune cells. The mechanism by which leukocidins kill innate immune cells and trigger inflammation during S. aureus lung infection, however, remains unresolved. Here, we explored human‐induced pluripotent stem cell‐derived macrophages (hiPSC‐dMs) to study the interaction of the leukocidins Panton–Valentine leukocidin (PVL) and LukAB with lung macrophages, which are the initial leukocidin targets during S. aureus lung invasion. hiPSC‐dMs were susceptible to the leukocidins PVL and LukAB and both leukocidins triggered NLPR3 inflammasome activation resulting in IL‐1β secretion. hiPSC‐dM cell death after LukAB exposure, however, was only temporarily dependent of NLRP3, although NLRP3 triggered marked cell death after PVL treatment. CRISPR/Cas9‐mediated deletion of the PVL receptor, C5aR1, protected hiPSC‐dMs from PVL cytotoxicity, despite the expression of other leukocidin receptors, such as CD45. PVL‐deficient S. aureus had reduced ability to induce lung IL‐1β levels in human C5aR1 knock‐in mice. Unexpectedly, inhibiting NLRP3 activity resulted in increased wild‐type S. aureus lung burdens. Our findings suggest that NLRP3 induces macrophage death and IL‐1β secretion after PVL exposure and controls S. aureus lung burdens. S. aureus leukocidin PVL triggers NLRP3 mediated cell death in human stem‐cell derived macrophages and humanized C5aR1 macrophages, leading to increased bacterial lung burdens.
... FBXO11 2/2 macrophages show dysregulated IL-1β expression PVL not only causes macrophage cell death but also activates the NLRP3 inflammasome to induce IL-1β secretion (Holzinger et al, 2012;Labrousse et al, 2014;Chow et al, 2020). To address whether FBXO11 regulates inflammation after exposure to bacterial toxins, we next determined IL-1β secretion as an indicator of NLRP3 activity. ...
Article
Full-text available
Staphylococcus aureus causes severe infections such as pneumonia and sepsis depending on the pore-forming toxin Panton–Valentine leukocidin (PVL). PVL kills and induces inflammation in macrophages and other myeloid cells by interacting with the human cell surface receptor, complement 5a receptor 1 (C5aR1). C5aR1 expression is tighly regulated and may thus modulate PVL activity, although the mechanisms involved remain incompletely understood. Here, we used a genome-wide CRISPR/Cas9 screen and identified F-box protein 11 (FBXO11), an E3 ubiquitin ligase complex member, to promote PVL toxicity. Genetic deletion of FBXO11 reduced the expression of C5aR1 at the mRNA level, whereas ectopic expression of C5aR1 in FBXO11 −/− macrophages, or priming with LPS, restored C5aR1 expression and thereby PVL toxicity. In addition to promoting PVL-mediated killing, FBXO11 dampens secretion of IL-1β after NLRP3 activation in response to bacterial toxins by reducing mRNA levels in a BCL-6–dependent and BCL-6–independent manner. Overall, these findings highlight that FBXO11 regulates C5aR1 and IL-1β expression and controls macrophage cell death and inflammation following PVL exposure.
... Similarly, mice treated with Anakinra, an IL-1 receptor antagonist (IL-1RA), exhibited more severe septic arthritis with an increased mortality [121]. In a murine model of Staphylococcus aureus pneumonia, Anakinra injection increased the bacterial burden in the lung [122]. In fact, IL-1β mediated neutrophil recruitment to inflamed tissues and activation of immune cells including dendritic cells and monocytes, as proved by Guo et al. using a cecal ligation and puncture (CLP) model in mice treated with IL-1β [123][124][125]. ...
Article
Sepsis is defined as a life-threatening organ dysfunction induced by a dysregulated host immune response to infection. Immune response induced by sepsis is complex and dynamic. It is schematically described as an early dysregulated systemic inflammatory response leading to organ failures and early deaths, followed by the development of persistent immune alterations affecting both the innate and adaptive immune responses associated with increased risk of secondary infections, viral reactivations, and late mortality. In this review, we will focus on the role of NACHT, leucin-rich repeat and pyrin-containing protein 3 (NLRP3) inflammasome in the pathophysiology of sepsis. NLRP3 inflammasome is a multiproteic intracellular complex activated by infectious pathogens through a two-step process resulting in the release of the pro-inflammatory cytokines IL-1β and IL-18 and the formation of membrane pores by gasdermin D, inducing a pro-inflammatory form of cell death called pyroptosis. The role of NLRP3 inflammasome in the pathophysiology of sepsis can be ambivalent. Indeed, although it might protect against sepsis when moderately activated after initial infection, excessive NLRP3 inflammasome activation can induce dysregulated inflammation leading to multiple organ failure and death during the acute phase of the disease. Moreover, this activation might become exhausted and contribute to post-septic immunosuppression, driving impaired functions of innate and adaptive immune cells. Targeting the NLRP3 inflammasome could thus be an attractive option in sepsis either through IL-1β and IL-18 antagonists or through inhibition of NLRP3 inflammasome pathway downstream components. Available treatments and results of first clinical trials will be discussed.
... Here, the treatment with CXCL9(74-103) did not reduce macrophage accumulation in BALF, but significantly decreased the number of neutrophils. At this point, some studies demonstrated that neutrophils are necessary for IL-1β production by macrophages [55,56], which may explain the lower detection of this cytokine in CXCL9(74-103)-treated mice. In addition, neutrophils can also be a source of IL-1β during acute inflammation [57]. ...
Article
Full-text available
Klebsiella pneumoniae is an important pathogen associated with hospital-acquired pneumonia (HAP). Bacterial pneumonia is characterized by a harmful inflammatory response with a massive influx of neutrophils, production of cytokines and chemokines, and consequent tissue damage and dysfunction. Targeted therapies to block neutrophil migration to avoid tissue damage while keeping the antimicrobial properties of tissue remains a challenge in the field. Here we tested the effect of the anti-inflammatory properties of the chemokine fragment CXCL9(74-103) in pneumonia induced by Klebsiella pneumoniae in mice. Mice were infected by intratracheal injection of Klebsiella pneumoniae and 6 h after infection were treated systemically with CXCL9(74-103). The recruitment of leukocytes, levels of cytokines and chemokines, colony-forming units (CFU), and lung function were evaluated. The treatment with CXCL9(74-103) decreased neutrophil migration to the airways and the production of the cytokine interleukin-1β (IL-1β) without affecting bacterial control. In addition, the therapeutic treatment improved lung function in infected mice. Our results indicated that the treatment with CXCL9(74-103) reduced inflammation and improved lung function in Klebsiella pneumoniae-induced pneumonia.
... The reduction of this inflammation is one way to prevent lung tissue damage. This has been demonstrated in pneumonia mouse models, in which cytokine production was hindered by the inhibition of NF-κB signaling (181), NLRP3 inflammasome inhibition (169), and IL-1R signaling (182). Another way is to inhibit S. aureus toxins, notably by using passive immunotherapy approaches, such as neutralization with antibodies targeting Hla, PVL, HlgACB, and LukDE. ...
Article
Full-text available
Staphylococcus aureus is both a commensal and a pathogenic bacterium for humans. Its ability to induce severe infections is based on a wide range of virulence factors. S. aureus community-acquired pneumonia (SA-CAP) is rare and severe, and the contribution of certain virulence factors in this disease has been recognized over the past 2 decades. First, the factors involved in metabolism adaptation are crucial for S. aureus survival in the lower respiratory tract, and toxins and enzymes are required for it to cross the pulmonary epithelial barrier. S. aureus subsequently faces host defense mechanisms, including the epithelial barrier, but most importantly the immune system. Here, again, S. aureus uses myriad virulence factors to successfully escape from the host’s defenses and takes advantage of them. The impact of S. aureus virulence, combined with the collateral damage caused by an overwhelming immune response, leads to severe tissue damage and adverse clinical outcomes. In this review, we summarize step by step all of the S. aureus factors implicated in CAP and described to date, and we provide an outlook for future research.
... Treatment with Anakinra (Swedish Orphan Biovitrum AB, Stockholm, Sweden), human IL-1 receptor antagonist protein used in rheumatoid arthritis [172], resulted in decreased cytokine production in a PVL-induced NP rabbit model. Unfortunately, Anakinra therapy did not show a comparable effect when probed in SAP [173]. ...
Article
Full-text available
Pneumonia is an acute pulmonary infection associated with high mortality and an immense financial burden on healthcare systems. Staphylococcus aureus is an opportunistic pathogen capable of inducing S. aureus pneumonia (SAP), with some lineages also showing multidrug resistance. Given the high level of antibiotic resistance, much research has been focused on targeting S. aureus virulence factors, including toxins and biofilm-associated proteins, in an attempt to develop effective SAP therapeutics. Despite several promising leads, many hurdles still remain for S. aureus vaccine research. Here, we review the state-of-the-art SAP therapeutics, highlight their pitfalls, and discuss alternative approaches of potential significance and future perspectives. Key Contribution: S. aureus pneumonia is associated with high fatality and a large array of virulence factors. This review offers an in-depth analysis of the development of non-antibiotic alternatives against S. aureus pneumonia.
... P. aeruginosa infection destroys lung architecture and function due to inflammatory-and neutrophil-mediated degradation of mucin layers and structural proteins of the pulmonary connective tissue [1,2]. Cytokines such as IL-1b [3,4] and IL-8 [5], the latter itself regulated by IL-1b [6], initiate and maintain this neutrophil-dependant inflammatory cycle in contrast to their normally host-protective roles [7][8][9]. Anti-inflammatory agents can mitigate tissue destruction to preserve pulmonary function during P. aeruginosa pneumonia [10] and CF [11,12]. ...
Article
Full-text available
Background: Pulmonary damage by Pseudomonas aeruginosa during cystic fibrosis lung infection and ventilator-associated pneumonia is mediated both by pathogen virulence factors and host inflammation. Impaired immune function due to tissue damage and inflammation, coupled with pathogen multidrug resistance, complicates the management of these deep-seated infections. Pathological inflammation during infection is driven by interleukin-1β (IL-1β), but the molecular processes involved are not fully understood. Methods: We examined IL-1β activation in a pulmonary model infection of Pseudomonas aeruginosa and in vitro using genetics, specific inhibitors, recombinant proteins, and targeted reporters of protease activity and IL-1β bioactivity. Findings: Caspase-family inflammasome proteases canonically regulate maturation of this proinflammatory cytokine, but we report that plasticity in IL-1β proteolytic activation allows for its direct maturation by the pseudomonal protease LasB. LasB promotes IL-1β activation, neutrophilic inflammation, and destruction of lung architecture characteristic of severe P. aeruginosa pulmonary infection. Interpretation: Preservation of lung function and effective immune clearance may be enhanced by selectively controlling inflammation. Discovery of this IL-1β regulatory mechanism provides a distinct target for anti-inflammatory therapeutics, such as matrix metalloprotease inhibitors that inhibit LasB and limit inflammation and pathology during P. aeruginosa pulmonary infections. Funding: Full details are provided in the Acknowledgements section.
... IL-1Ra competitively inhibits IL-1b binding to IL-1R, and it exerts an inhibitory effect on the IL-1bmediated inflammatory response. [21,22] As a pro-inflammatory factor, IL-12 mainly has 2 subunits, p40 and p35. These 2 subunits are connected to each other through a disulfide bond, to form p70, which has relevance to immune regulation and inflammation. ...
Article
Full-text available
Background: Our study aimed to screen and explore the expression of inflammatory factors in keloid patients and to investigate how hyperbaric oxygen (HBO) therapy affects the expression levels of interleukin-12p40 (IL-12p40), macrophage inflammatory protein-1β (MIP-1β), platelet-derived growth factor-BB (PDGF-BB), and interleukin-1 receptor antagonist (IL-1Ra). Objective: 30 patients were randomly selected and divided into the following 3 groups: keloid samples from keloid patients treated with HBO therapy (A), keloid samples from keloid patients treated without HBO therapy (B), and normal control skin samples derived from individuals who had no clear scarring (C). Each group included 10 samples. Methods: Inflammatory factors in the keloid tissues were measured with the MILLIPLEX multiplexed Luminex system. Hematoxylin and eosin staining, immunohistochemical staining, and Western blotting were used to observe the morphological differences in different tissues and the expression levels. Results: The expression levels of inflammatory mediators, including IL-12p40, MIP-1β, PDGF-BB, and IL-1Ra, in keloid tissues were significantly different from those in samples of normal skin. Hematoxylin and eosin staining showed significantly greater inflammatory infiltration in keloid tissue. Significantly different expression levels were observed in group A, B, and C. Conclusion: Significantly altered levels of inflammatory factors in the samples from keloid patients were observed, suggesting that formation of a keloid is potentially related to inflammatory responses. HBO therapy could significantly affect the expression levels of IL-12p40, MIP-1β, PDGF-BB, and IL-1Ra, indicating that the effects of HBO therapy are associated with the attenuation of inflammatory responses.
... Furthermore, studies also have indicated the involvement of AKT/NF-κB pathway in LPS-induced inflammatory responses in murine macrophages (15,25). In addition, the activation of NF-κB is a prerequisite for production of various inflammatory cytokines, including TNF-α, IL-1 and IL-6 (26), generating a much more severe inflammatory response in stimulated macrophages (27). Expression levels of proinflammatory cytokines including IL-6, TNF-α and IL-1β are correlated with pneumonia (28), which are also reported to be induced by LPS in the in vitro pneumonia model in A549 cells or monocytes cells (29,30). ...
Preprint
Full-text available
Pneumonia accounts for approximately 15% mortalities in adolescents worldwide. MicroRNAs (miRNAs) regulate numerous diseases including pneumonia. miRNA and mRNA expression levels were detected by real time polymerase chain reaction (RT-qPCR). Protein expression levels were determined by enzyme-linked immunosorbent assay (ELISA) and western blot. The interaction between phosphatase and tensin homolog on chromosome ten (PTEN) and miR-103a-3p was explored by dual luciferase reporter assay. Cell viability and cell apoptosis were detected by cell Counting Kit-8 (CCK-8) and flow cytometry. Herein, we discovered that PTEN was decreased and miR-103a-3p was overexpressed in Ana-1 cells of in vitro pneumonia model. miR-103a-3p downregulated the expression levels of PTEN. AntagomiR-103a-3p reversed the increased cell apoptosis and decreased cell viability and inflammatory cytokine expression levels (tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and IL-6) induced by LPS in Ana-1 cells by PTEN. AntagomiR-103a-3p inhibited the activation of PTEN/PI3K/AKT/NF-κB signaling pathway induced by LPS in Ana-1 cells. Taken together, our findings exhibited that miR-103a-3p attenuated LPS induced pneumonia by blocking the activation of PTEN/PI3K/AKT/NF-κB signaling pathway and the following cell apoptosis as well as release of proinflammatory cytokines, suggesting that miR-103a-3p might serve as a novel therapeutic target for the treatment of pneumonia.
Chapter
The NLRP3 inflammasome senses the activity of pore-forming toxins secreted by Staphylococcus aureus. The bacterial toxins compromise plasma membrane integrity which activates the NLRP3 inflammasome to induce host pore-forming proteins and cellular suicide, termed pyroptosis. Host cell death rates are routinely determined at pre-defined time points and on whole cell populations. To capture the dynamic interactions between bacterial pore-forming toxins and host cell death factors, we have applied live-cell imaging techniques capable of analyzing single cell events in real time. Here, we describe methods using live-cell imaging to determine the host responses, such as plasma membrane integrity, mitochondrial health, and apoptotic caspases, towards pore-forming toxins.
Article
Full-text available
Ceftaroline, the active metabolite of the prodrug ceftaroline fosamil, is a cephalosporin with broad-spectrum in vitro activity against Gram-positive organisms, including methicillin-resistant Staphylococcus aureus (MRSA) and multidrug-resistant Streptococcus pneumoniae (MDRSP), and common Gram-negative pathogens. This study investigated the in vivo activity of ceftaroline fosamil compared with clindamycin, linezolid, and vancomycin in a severe pneumonia model due to MRSA-producing Panton-Valentine leukocidin (PVL). A USA300 PVL-positive clone was used to induce pneumonia in rabbits. Infected rabbits were randomly assigned to no treatment or simulated human-equivalent dosing with ceftaroline fosamil, clindamycin, linezolid, or vancomycin. Residual bacterial concentrations in the lungs and spleen were assessed after 48 h of treatment. PVL expression was measured using a specific enzyme-linked immunosorbent assay (ELISA). Ceftaroline, clindamycin, and linezolid considerably reduced mortality rates compared with the control, whereas vancomycin did not. Pulmonary and splenic bacterial titers and PVL concentrations were greatly reduced by ceftaroline, clindamycin, and linezolid. Ceftaroline, clindamycin, and linezolid were associated with reduced pulmonary tissue damage based on significantly lower macroscopic scores. Ceftaroline fosamil, clindamycin, and, to a lesser extent, linezolid were efficient in reducing bacterial titers in both the lungs and spleen and decreasing macroscopic scores and PVL production compared with the control.
Article
Full-text available
To determine whether treatment with interleukin-1 receptor antagonist (IL1-ra) would affect amniotic fluid concentrations of tumor necrosis factor alpha (TNF-alpha) and prostaglandins or clinical or microbiological outcomes in a model of ascending bacterial infection in pregnancy. Timed pregnant New Zealand white rabbits at 70% of gestation underwent endoscopic inoculation of the cervices with 10(6) - 10(7) cfu Escherichia coli. Animals were randomly assigned in a blinded manner to a 5-h intravenous infusion of human IL1-ra (10 mg/kg) or placebo beginning 1-2 h after inoculation. Blood was drawn from the does for assay of serum IL1-ra concentration before inoculation, at mid-infusion, after the infusion ended and at necropsy. At necropsy, temperature and cultures were taken, and aspirated amniotic fluid was pooled for assays of TNF-aalpha, prostaglandin E2 (PGE2) and ILI-ra. Serum IL1-ra concentrations rose to a mean of 2 microg/ml at mid-infusion and fell markedly after the infusion to concentrations barely detectable at necropsy. Between the two groups, there were no significant differences in the rates of fever or positive cultures or in amniotic fluid concentrations of PGE2 or TNF-alpha. One unique finding was the demonstration that administration of human IL1-ra to the does resulted in measurable concentrations of human IL1-ra in the amniotic fluid. Treatment with an intravenous infusion of human IL1-ra after cervical inoculation with E. coli did not affect clinical or microbiological outcomes or amniotic fluid concentrations of TNF-alpha or PGE2. This experiment providesthefirstdemonstration of passage of human IL1-ra from the maternal bloodstream to the amniotic fluid.
Article
Full-text available
Background: Linezolid is recommended for treatment of pneumonia and other invasive infections caused by methicillin-resistant Staphylococcus aureus (MRSA). The premise underlying this recommendation is that linezolid inhibits in vivo production of potent staphylococcal exotoxins, including Panton-Valentine leukocidin (PVL) and α-hemolysin (Hla), although supporting evidence is lacking. Methods: A rabbit model of necrotizing pneumonia using MRSA clone USA300 was used to compare therapeutic effects of linezolid (50 mg/kg 3 times/day) and vancomycin (30 mg/kg 2 times/day) administered 1.5, 4, and 9 hours after infection on host survival outcomes and in vivo bacterial toxin production. Results: Mortality rates were 100% for untreated rabbits and 83%-100% for vancomycin-treated rabbits. In contrast, mortality rates were 25%, 50%, and 100% for rabbits treated with linezolid 1.5, 4, and 9 hours after infection, respectively. Compared with untreated and vancomycin-treated rabbits, improved survival of rabbits treated 1.5 hours after infection with linezolid was associated with a significant decrease in bacterial counts, suppressed bacterial production of PVL and Hla, and reduced production of the neutrophil-chemoattractant interleukin 8 in the lungs. Conclusions: Across the study interval, only early treatment with linezolid resulted in significant suppression of exotoxin synthesis and improved survival outcomes in a rabbit model of MRSA necrotizing pneumonia.
Article
Full-text available
Staphylococcus aureus community-acquired (CA) MRSA strains are highly virulent and can cause infections in otherwise healthy individuals. The most important mechanism of the host for clearing S. aureus is phagocytosis by neutrophils and subsequent killing of the pathogen. Especially CA-MRSA strains are very efficient in circumventing this neutrophil killing. Interestingly, only a relative small number of virulence factors have been associated with CA-MRSA, one of which are the phenol soluble modulins (PSMs). We have recently shown that the PSMs are functionally inhibited by serum lipoproteins, indicating that PSMs may exert their cytolytic function primarily in the intracellular environment. To further investigate the intracellular role of the PSMs we measured the effect of the α-type and β-type PSMs on neutrophil killing after phagocytosis. Using fluorescently labeled S. aureus, we measured bacterial survival after phagocytosis in a plate reader, which was employed next to flow cytometry and time-lapse microscopy. Phagocytosis of the CA-MRSA strain MW2 by human neutrophils resulted in rapid host cell death. Using mutant strains of MW2, we demonstrated that in the presence of serum, the intracellular expression of only the psmα operon is both necessary and sufficient for both increased neutrophil cell death and increased survival of S. aureus. Our results identify PSMα peptides as prominent contributors to killing of neutrophils after phagocytosis, a finding with major implications for our understanding of S. aureus pathogenesis and strategies for S. aureus vaccine development.
Article
Full-text available
Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) is epidemic in the United States, even rivaling HIV/AIDS in its public health impact. The pandemic clone USA300, like other CA-MRSA strains, expresses Panton-Valentine leukocidin (PVL), a pore-forming toxin that targets polymorphonuclear leukocytes (PMNs). PVL is thought to play a key role in the pathogenesis of necrotizing pneumonia, but data from rodent infection models are inconclusive. Rodent PMNs are less susceptible than human PMNs to PVL-induced cytolysis, whereas rabbit PMNs, like those of humans, are highly susceptible to PVL-induced cytolysis. This difference in target cell susceptibility could affect results of experimental models. Therefore, we developed a rabbit model of necrotizing pneumonia to compare the virulence of a USA300 wild-type strain with that of isogenic PVL-deletion mutant and -complemented strains. PVL enhanced the capacity of USA300 to cause severe lung necrosis, pulmonary edema, alveolar hemorrhage, hemoptysis, and death, hallmark clinical features of fatal human necrotizing pneumonia. Purified PVL instilled directly into the lung caused lung inflammation and injury by recruiting and lysing PMNs, which damage the lung by releasing cytotoxic granule contents. These findings provide insights into the mechanism of PVL-induced lung injury and inflammation and demonstrate the utility of the rabbit for studying PVL-mediated pathogenesis.
Article
Background. Linezolid is recommended for treatment of pneumonia and other invasive infections caused by methicillin-resistant Staphylococcus aureus (MRSA). The premise underlying this recommendation is that linezolid inhibits in vivo production of potent staphylococcal exotoxins, including Panton-Valentine leukocidin (PVL) and α-hemolysin (Hla), although supporting evidence is lacking. Methods. A rabbit model of necrotizing pneumonia using MRSA clone USA300 was used to compare therapeutic effects of linezolid (50 mg/kg 3 times/day) and vancomycin (30 mg/kg 2 times/day) administered 1.5, 4, and 9 hours after infection on host survival outcomes and in vivo bacterial toxin production. Results. Mortality rates were 100% for untreated rabbits and 83%—100% for vancomycin-treated rabbits. In contrast, mortality rates were 25%, 50%, and 100% for rabbits treated with linezolid 1.5, 4, and 9 hours after infection, respectively. Compared with untreated and vancomycin-treated rabbits, improved survival of rabbits treated 1.5 hours after infection with linezolid was associated with a significant decrease in bacterial counts, suppressed bacterial production of PVL and Hla, and reduced production of the neutrophil-chemoattractant interleukin 8 in the lungs. Conclusions. Across the study interval, only early treatment with linezolid resulted in significant suppression of exotoxin synthesis and improved survival outcomes in a rabbit model of MRSA necrotizing pneumonia.
Article
Staphylococcus aureus secretes numerous virulence factors that facilitate evasion of the host immune system. Among these molecules are pore-forming cytolytic toxins, including Panton-Valentine leukocidin (PVL), leukotoxin GH (LukGH; also known as LukAB), leukotoxin DE, and γ-hemolysin. PVL and LukGH have potent cytolytic activity in vitro, and both toxins are proinflammatory in vivo. Although progress has been made toward elucidating the role of these toxins in S. aureus virulence, our understanding of the mechanisms that underlie the proinflammatory capacity of these toxins, as well as the associated host response toward them, is incomplete. To address this deficiency in knowledge, we assessed the ability of LukGH to prime human PMNs for enhanced bactericidal activity and further investigated the impact of the toxin on neutrophil function. We found that, unlike PVL, LukGH did not prime human neutrophils for increased production of reactive oxygen species nor did it enhance binding and/or uptake of S. aureus. Unexpectedly, LukGH promoted the release of neutrophil extracellular traps (NETs), which, in turn, ensnared but did not kill S. aureus. Furthermore, we found that electropermeabilization of human neutrophils, used as a separate means to create pores in the neutrophil plasma membrane, similarly induced formation of NETs, a finding consistent with the notion that NETs can form during nonspecific cytolysis. We propose that the ability of LukGH to promote formation of NETs contributes to the inflammatory response and host defense against S. aureus infection.
Article
Panton-Valentine Leukocidin (PVL) is a staphylococcal bicomponent pore-forming toxin linked to severe invasive infections. Target-cell and species specificity of PVL are poorly understood, and the mechanism of action of this toxin in Staphylococcus aureus virulence is controversial. Here, we identify the human complement receptors C5aR and C5L2 as host targets of PVL, mediating both toxin binding and cytotoxicity. Expression and interspecies variations of the C5aR determine cell and species specificity of PVL. The C5aR binding PVL component, LukS-PV, is a potent inhibitor of C5a-induced immune cell activation. These findings provide insight into leukocidin function and staphylococcal virulence and offer directions for future investigations into individual susceptibility to severe staphylococcal disease.