Staphylococcus Aureus Products Subvert The Burkholderia

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

RESEARCH ARTICLE

Ji et al., Journal of Medical Microbiology 2019;68:1813–1822


DOI 10.1099/jmm.0.001100

Editor's Choice

Staphylococcus aureus products subvert the Burkholderia


cenocepacia-­induced inflammatory response in airway
epithelial cells
Yuan Ji, Albert Bolhuis and Malcolm L. Watson*

Abstract
Introduction. Chronic pulmonary infection is associated with colonization with multiple micro-­organisms but host–microbe and
microbe–microbe interactions are poorly understood.
Aim. This study aims to investigate the differences in host responses to mono- and co-­infection with S. aureus and B. cenoce-
pacia in human airway epithelial cells.
Methodology. We assessed the effect of co-­infection with B. cenocepacia and S. aureus on host signalling and inflammatory
responses in the human airway epithelial cell line 16HBE, using ELISA and western blot analysis.
Results. The results show that B. cenocepacia activates MAPK and NF-κB signalling pathways, subsequently eliciting robust
interleukin (IL)-8 production. However, when airway epithelial cells were co-­treated with live B. cenocepacia bacteria and S.
aureus supernatants (conditioned medium), the pro-­inflammatory response was attenuated. This anti-­inflammatory effect was
widely exhibited in the S. aureus isolates tested and was mediated via reduced MAPK and NF-κB signalling, but not via IL-1
receptor or tumour necrosis factor receptor modulation. The staphylococcal effectors were characterized as small, heat-­stable,
non-­proteinaceous and not cell wall-­related factors.
Conclusion. This study demonstrates for the first time the host response in a S. aureus/B. cenocepacia co-­infection model
and provides insight into a staphylococcal immune evasion mechanism, as well as a therapeutic intervention for excessive
inflammation.

Introduction respiratory infections in both hospital and community


Lower respiratory tract infections are a major cause of environments [3]. This is not only due to the wide range of
morbidity and mortality globally [1]. Individuals with an virulence factors deployed by S. aureus [4, 5], but also the
rapid emergence of multidrug-­resistant strains. S. aureus
immune deficiency, chronic obstructive pulmonary disease,
that gains resistance to β-lactam antibiotics as a conse-
or cystic fibrosis (CF) are particularly prone to such respira-
quence of adopting the staphylococcal cassette chromo-
tory infections [2].
somal mec element becomes methicillin-­resistant S. aureus
The opportunistic pathogen Staphylococcus aureus is one (MRSA). This emergence is posing serious challenges to
of the major threats that is commonly associated with antimicrobial therapy, as patients harbouring MRSA are

Received 03 July 2019; Accepted 05 October 2019; Published 01 November 2019


Author affiliations: 1Department of Pharmacy and Pharmacology, University of Bath, BA2 7AY, UK.
*Correspondence: Malcolm L. Watson, ​prsmlw@​bath.​ac.​uk
Keywords: Staphylococcus aureus; Burkholderia cepacia complex; host–pathogen interactions; immunomodulatory products.
Abbreviations: ANOVA, analysis of variance; ATCC, American Type Culture Collection; BCA, bicinchoninic acid assay; Bcc, Burkholderia cepacia
complex; CF, cystic fibrosis; DMEM, Dulbecco’s Modified Eagle Medium; DMSO, dimethyl sulfoxide; ELISA, enzyme-­linked immunosorbent assay;
IL, interleukin; JNK, c-­Jun N-­terminal kinases; LB, Luria-­Bertani Broth; LPS, lipopolysaccharide; MAPK, mitogen-­activated protein kinase;
MOI, multiplicity of infection; MRSA, methicillin-­resistant Staphylococcus aureus; MSSA, methicillin-­sensitive Staphylococcus aureus; MTT,
3-(4,5-­dimethylthiazol-2-­yl)-2,5-­diphenyltetrazolium bromide; MWCO, molecular weight cut-­off; NCTC, National Collection of Type Cultures; NF-κB,
nuclear factor κ-light-­chain-­enhancer of activated B cells; PI3K/Akt/mTOR, phosphoinositide 3-­kinases/ protein kinase B/mammalian target of
rapamycin; SDS-­PAGE, sodium dodecyl sulfate–polyacrylamide gel electrophoresis; SEM, standard error of the mean; SN, supernatant; TLR, toll-­like
receptor; TNF, tumour necrosis factor.

001100 © 2019 The Authors


1813
Ji et al., Journal of Medical Microbiology 2019;68:1813–1822

associated with worse clinical prognosis compared to those of staphylococcal immunomodulatory factors and the nature
who never have MRSA [6, 7]. of immunomodulatory molecules released by S. aureus,
highlighting the anti-­inflammatory modulation of S. aureus
Burkholderia cepacia complex (Bcc) is a major threat for
immunocompromised individuals such as CF patients, as it during Bcc co-­infection and potential effects on the clinical
is transmissible between patients who can develop ‘cepacia outcome.
syndrome’, which is a fatal exacerbation of respiratory function
as a result of bacteraemia [8–11]. Together with Pseudomonas
aeruginosa and other species, Bcc are non-­fermenting Gram-­ Methods
negative bacilli that are not able to utilize glucose in the Bacterial strains and culture
absence of oxygen. Pathogens of these bacteria have become an The bacterial strains used in this study are listed in Table 1.
increasing challenge in hospital-­acquired infections due to the Bacteria were cultured in Luria-­Bertani (LB)broth (Thermo
high magnitude of intrinsic antibiotic resistance [12, 13]. While Fisher Scientific) at 37 °C with shaking. Bacterial superna-
the prevalence of S. aureus infection is about 60 % among CF tants were collected and filtered through a 0.2 µm membrane.
patients younger than 2 years and peaks between 11 to 17 years Where indicated, supernatants were heat-­treated by incuba-
old, most Bcc colonization is acquired after the age of 18 [14]. tion at 95 °C for 10 min. Heat-­killed S. aureus were prepared
Bcc bacteria express a wide range of virulence factors to facilitate
by resuspending the overnight culture to the desired multi-
the infection process, such as cable pili, flagella, several secre-
plicity of infection (m.o.i.) in Dulbecco's Modified Eagle
tion systems or homologous structures [15–17]. In addition,
Medium (DMEM) (Gibco, Thermo Fisher Scientific) and
the unusual modification of lipopolysaccharide (LPS) at least
heating at 100 °C for 1 h.
partially contributes to the virulence of Bcc. Acylation of lipid
A contributes to a much stronger pro-­inflammatory response Proteinase K immobilized on Eupergit C (Sigma-­Aldrich) was
induced by Bcc LPS even compared to LPS from P. aeruginosa used at a concentration of 100 µg ml−1 in Tris-­Cl (50 mM, pH
and Escherichia coli [18]. The resulting cytokine storm attracts 8)/CaCl2 (10 mM) at 37 °C for 1 h. At the end of digestion,
a large number of immune cells, such as neutrophils and mono- proteinase K was removed by centrifugation at 250 g for 10 min.
nuclear cells, to the lung. If the acute inflammatory response is Inactivated proteinase K was prepared by heating at 100 °C for
not resolved, it causes tissue injury, which leads to pulmonary 1 h.
fibrosis and possibly develops into a chronic inflammation [19].
Fractionated products of S. aureus were prepared by submit-
Chronic pulmonary infection is commonly associated with ting supernatants to 10 kDa and 3 kDa cutoff centrifugal
colonization with more than one micro-­organism, but little concentrators (Vivaspin 20, Sigma-­Aldrich) and centrifuging
is known regarding the complicated mechanisms of host– them at 5000 g for 17 min and 180 min, respectively, following
microbe and microbe–microbe interactions and their impact the manufacturer’s instructions. Supernatants were sepa-
on disease. The microbe–microbe interaction can be synergistic rated into three fractions: >10 000 molecular weight cut-­off
or antagonistic in nature and both forms of interaction can (MWCO), 3000–10000 MWCO and <3000 MWCO. Filtrates
lead to increased antibiotic resistance and virulence [20–22]. and concentrates were adjusted to the original volume with
Although many studies have reported worse clinical outcomes fresh LB, followed by filter sterilization.
resulting from microbe–microbe interactions [23–25], this is
not always the case. In contrast to the negative impact by Bcc
and P. aeruginosa, S. aureus co-­infection is positively associated Table 1. Bacterial strains
with better lung function, higher survival rate and delayed lung
disease progression [26–29]. An in vivo study that mimicked the Strain Characteristics Reference
sequential acquisition of early S. aureus and late P. aeruginosa in MRSA252 Hospital-­acquired MRSA, fatal post-­ [57]
the murine lung showed that preinfection with S. aureus signifi- op septicaemia
cantly reduced inflammation and the incidence of mortality
MSSA NCTC 6571 Methicillin-­sensitive S. aureus
caused by P. aeruginosa but enhanced P. aeruginosa persistence
[30]. We thus hypothesized that the protective effect of S. aureus MSSA Newman Isolated from a secondary [58]
results from dampening of the inflammatory response towards osteomyelitis infection of a TB patient
Bcc through interference with the host signalling pathways. MSSA209 [59]
Therefore, evaluating whether S. aureus colonization favours the
host and understanding the mechanism behind the unexpected MRSA41 [59]
protective effect of S. aureus may shed light on novel therapies MSSA SH1000 Laboratory strain, derivative of NCTC [60]
for persistent infections. 8325 by complementation of rsbU

In this study, in order to gain insights into the host response MSSA RN4220 Derivative of NCTC8325-4, with agrA [61]
to S. aureus/B. cenocepacia co-­colonization, we assessed the mutation
effect of B. cenocepacia on the host signalling cascade and MRSA USA400 Community-­associated S. aureus [62]
inflammatory responses in airway epithelial cells compared
B. cenocepacia J2315 Epidemic CF-­related ET12 lineage [63]
to that of co-­infection. Further, we identified the host target

1814
Ji et al., Journal of Medical Microbiology 2019;68:1813–1822

Human airway epithelial cell culture increase in IL-8 response in 16HBE cells (Fig. 1a). Although
Human bronchial epithelial 16HBE (obtained from Professor supernatants (in LB medium) of S. aureus MRSA252 or MSSA
Dieter C. Gruenert, University of California San Francisco) NCTC 6571 did not change IL-8 production compared to the
and human airway epithelial Calu-3 (ATCC) were maintained control, they significantly downregulated IL-8 induced by B.
in DMEM, supplemented with 10 % heat-­inactivated foetal cenocepacia (23 and 31% less than the B. cenocepacia-­treated
bovine serum, 100 units mL−1 penicillin and 100 µg ml−1 strep- group, respectively). In contrast, heat-­killed S. aureus did not
tomycin (all from Gibco, Thermo Fisher Scientific). Cells were have this suppressing effect (Fig. 1b).
incubated at 37 °C in a humidified atmosphere with 5 % CO2. The same experiments were performed with a collection of
IL-1β and tumour necrosis factor (TNF)-α (Peprotech) were S. aureus isolates (Fig. 1c). Supernatants from all the tested
used at the concentrations indicated. isolates downregulated the level of IL-8 production by
different amounts (5.4–38.0 %). Although some of the differ-
Cell viability assay ences were small, consistent changes were seen within each
The cell viability assay with the MTT [3-(4,5-­dimethylthiazol- experiment and these were statistically significant (P<0.05).
2-­yl)−2,5-­diphenyltetrazolium bromide; Sigma-­Aldrich] assay The ability of heat-­killed bacteria to inhibit B. cenocepacia-­
was performed as described previously [31]. Briefly, 16HBE cells elicited IL-8 production was also assessed (Fig. 1d). However,
were plated in 96-­well plates (5000 cells/well) and incubated for whole heat-­killed bacteria had no effect on B. cenocepacia-­
24 h. Next, the medium was replaced by 10 % S. aureus super- induced IL-8 production. These data indicate that anti-­
natants for 24 h. After incubating cells with 12 mM MTT for 2 inflammatory activity in these experiments is expressed in
h, formazan formed in viable cells was dissolved in DMSO for secreted products but not heat-­stable S. aureus cell surface
10 min before the optical density was read at 550 nm. structures. This is in contrast to the nasal isolate S. aureus
anti-­inflammatory activity reported by Peres et al. [34], which
Detection of cytokine production was exhibited by whole bacteria subjected to the same heat
treatment conditions.
At the end of stimulation, conditioned medium of 16HBE
cells was collected and the concentration of IL-8 was quan- To confirm that the suppressing effect is not due to cell damage,
tified using Maxisorp ELISA plates (Nunc, Thermo Fisher an MTT assay was used to assess cell viability (Fig. 1e). The
Scientific) and a commercially available enzyme-­ linked supernatants of the majority of S. aureus strains did not cause
immunosorbent assay (ELISA) kit (eBioscience, Thermo epithelial cell damage, indicating that there was no causality
Fisher Scientific) according to the manufacturer’s instructions. between IL-8 suppression and the induction of epithelial cell
damage. The exceptions were SH1000 and RN4220, which
Western blot analysis did reduce cell viability and, interestingly, these alone caused
Following stimulation, cell lysates were prepared and the concen- a doubling of IL-8 production (Fig. 1c).
tration of total protein was quantified using the Pierce BCA
Protein Assay kit (Thermo Fisher Scientific). Protein samples MSSA NCTC 6571 products block MAPK p38
were resolved on an SDS-­PAGE gel and immunoblotted on a and Erk, and NF-κB signalling activated by B.
nitrocellulose membrane (Bio-­Rad). The membrane was probed cenocepacia
with the antibodies anti-­phospho-­NF-κB-­p65, -phospho-­p38, The recognition of conserved microbial structures of patho-
-phospho-­Erk and -p38 (Cell Signaling Technology), followed gens by pattern recognition receptors leads to the activation
by incubation with secondary antibodies (Li-­Cor Biosciences). of the MAPK and NF-κB signalling pathways, subsequently
Quantitative analysis of the signal was performed using the initiating the expression of pro-­inflammatory genes to mount
Li-­Cor Odyssey Clx imaging system. an immune response [35]. The NF-κB and MAPK pathways
contribute to IL-8 expression at multiple levels [36]. Since B.
Statistical analysis cenocepacia induced strong IL-8 production, the effects of B.
Data were expressed as the mean±standard error of the mean cenocepacia on MAPK and NF-κB signalling in 16HBE cells
(sem) for n=3 independent biological repeats. The results were were assessed. Cells were challenged with B. cenocepacia at
compared using GraphPad Prism and one- or two-­way analysis an m.o.i. of 5 from 5 to 180 min (Fig. 2a–d). For the MAPK
of variance (ANOVA), followed by Tukey’s or Dunnett’s multiple family, phosphorylation of p38 was maximal at 120 min (200
comparisons tests, with P<0.05 considered significant. % more than time 0), while phosphorylation of Erk peaked at
5 min (156 % more than time 0), followed by a decrease and
then a secondary rise at 120 min (133 % more than time 0).
Results For NF-κB signalling, B. cenocepacia upregulated phospho-
B. cenocepacia-stimulated IL-8 response is rylation of p65, particularly after 120 min.
suppressed by S. aureus products In order to study the modulation of host signalling by this
IL-8 plays an important role at the early stage of infection by co-­stimulation model, 16HBE cells were treated with B.
chemoattracting granulocytes such as neutrophils [32]. Hence, cenocepacia (m.o.i. of 5) and 10 % S. aureus supernatant for
the level of IL-8 is a biomarker of pro-­inflammatory responses 2 h (Fig. 2e–h). B. cenocepacia led to the phosphorylation of
and inflammation [11, 33]. B. cenocepacia induced a robust p38, Erk and p65, whereas S. aureus supernatant alone did not

1815
Ji et al., Journal of Medical Microbiology 2019;68:1813–1822

Fig. 1. S. aureus supernatant inhibits the B. cenocepacia-­induced IL-8 response. IL-8 production in 16HBE cells treated with B. cenocepacia
(m.o.i. of 5) in the presence or absence of (a) 10 % S. aureus supernatants (SN) or (b) heat-­killed S. aureus (m.o.i. of 5) for 24 h. The
experiments were validated using (c) 10 % supernatants from S. aureus isolates or (d) heat-­killed S. aureus and B. cenocepacia (m.o.i.
of 5) co-­stimulation for 24 h. In (a) and (c), 10 % LB was used as a control. The data represent the mean±SEM of duplicate samples
from three identical experiments. Significance for comparison to B. cenocepacia-­treated cells was determined using two-­way ANOVA
followed by Dunnett’s multiple comparison test (*, P<0.05). (e) Cytotoxicity of S. aureus supernatants for 16HBE cells was assessed using
a MTT assay. The control was treated with 10 % LB. The results are presented as a percentage of the control, mean±sem from three
independent experiments. Significance for comparison to the control was determined using one-­way ANOVA followed by Dunnett’s
multiple comparison test.

1816
Ji et al., Journal of Medical Microbiology 2019;68:1813–1822

Fig. 2. Effect of S. aureus supernatants on by B. cenocepacia-­activated MAPK and NF-κB signalling. 16HBE cells were infected by B.
cenocepacia unwashed culture at an m.o.i. of 5 for the indicated time. TNFα (20 ng ml−1) for 5 min was used as the positive control. (a)
Western blot analysis for levels of phosphorylated p38, phosphorylated Erk, phosphorylated p65 and p38 in cell lysate followed by B.
cenocepacia stimulation. Phosphorylation of the p38 (b), ERK (c) and p65 (d) of cell lysates was measured by Western blot and normalized
against total p38. 16HBE cells were treated with 10% LB as the control, and B. cenocepacia (5 m.o.i.), with the presence or absence of
10% S. aureus supernatant, for 2 h. (e) Western blot analysis for levels of phosphorylated p38, phosphorylated Erk, phosphorylated p65
and p38 in cell lysate following S. aureus supernatant and B. cenocepacia co-­stimulation. Phosphorylation of the p38 (f), Erk (g) and p65
(h) of cell lysates was measured using Western blot and normalized against total p38. The protein bands are representative of three
independent experiments. The data are shown as the mean±sem. *, P<0.05, significant reduction compared to the B. cenocepacia-­treated
group using two-­way ANOVA followed by a one-­tailed Dunnett’s multiple comparisons test.

1817
Ji et al., Journal of Medical Microbiology 2019;68:1813–1822

Fig. 3. Characterization of anti-­inflammatory components in S. aureus supernatants. 16HBE cells were treated with B. cenocepacia (m.o.i.
of 5), in the presence or absence of 10% (a) heat-­treated supernatants (SN), (b) proteinase K (PK)-­treated S. aureus supernatants, or
(c) S. aureus supernatant fractions containing metabolites of >10 kDa, >3 kDa and <3 kDa for 24 h. IL-8 production was analysed using
ELISA. The data represent the mean±sem from three independent experiments performed in duplicate. *, P<0.05 significance compared
to B. cenocepacia-­treated group using two-­way ANOVA followed by Dunnett’s multiple comparisons test. (d) 16HBE cells were co-­treated
with IL-1 (1 ng ml−1)/TNFα (10 ng ml−1) and 10% S. aureus supernatants for 24 h. B. cenocepacia (m.o.i. of 5) was the positive control. The
data represent the mean±sem from three independent experiments performed in duplicate. *, P<0.05 compared to the B. cepacia-­treated
group using two-­way ANOVA followed by Tukey’s multiple comparisons test.

change any tested signalling events. Notably, MSSA NCTC These heat-­treated supernatants were still able to inhibit the
6571 but not MRSA252 supernatant reduced the phospho- B. cenocepacia-­stimulated IL-8 response (Fig. 3a), indicating
rylation of p38, Erk and p65 stimulated by B. cenocepacia. that the components are heat-­stable.
The reduction in p38 and p65 signalling was significant
Further, S. aureus supernatants were treated with solid-­phase
compared with that for B. cenocepacia alone (P<0.05). This
proteinase K for 1 h at 37 °C before removal to avoid any effect
strain selectivity is consistent with the differential inhibition
of the proteinase on released IL-8. Proteinase K-­digested
of the pro-­inflammatory response (Fig. 1a) by MSSA NTCT
supernatants did not restore IL-8 secretion caused by B.
6571 and MRSA252. Therefore, these findings suggest that
cenocepacia; rather, IL-8 secretion was reduced almost to the
MSSA NCTC 6571 supernatant may suppress the pro-­
baseline (Fig. 3b). Further, the 16HBE cultures incubated with
inflammatory effect of B. cenocepacia by inhibiting MAPK
proteinase-­treated supernatant alone did not inhibit basal
and NF-κB signalling.
IL-8 levels (Fig. 3b), indicating that no amount of protease K
was carried over into the production assay. Heat-­inactivated
Characterization of anti-inflammatory factors in S. proteinase K-­treated supernatants displayed a similar inhibi-
aureus supernatant tion level to the untreated supernatants (Fig. 1a), indicating
To investigate the nature of anti-­inflammatory components, that the components are either non-­ proteinaceous or
S. aureus supernatants were heated at 95 °C for 10 min. proteinase K-­resistant.

1818
Ji et al., Journal of Medical Microbiology 2019;68:1813–1822

To assess the approximate molecular weight of the secretory A whole-­genome study divided 88 S. aureus isolates into 25
S. aureus components causing a reduction in B. cenocepacia-­ clonal complex lineages and investigated 43 immune evasion
induced IL-8 production, S. aureus supernatants were genes [43]. The results revealed that although genes encoding
separated into different molecular cut-­off fractions using leucocidin are highly conserved, there are variants among
selective membranes (Fig. 3c). Components in the superna- other genes, indicating that each lineage applies different
tants with a molecular weight less than 3 kDa, but not above strategies and functions of immune evasion.
3 kDa, reduced IL-8 production back to the same level as
The MAPK family and NF-κB signalling pathways are known
the control, indicating that the anti-­inflammatory factors are
to play key roles in the host response to bacterial pathogens by
small molecules.
regulating the expression of cytokines and chemokines [44].
As staphylococcal virulence factors (such as SpA and However, pathogens have evolved to exploit these pathways.
Panton–Valentine leucocidin) target the IL-1 receptor (IL-­ For example, B. cenocepacia K56-2 can significantly upregu-
1R) and tumour necrosis factor receptor (TNFR), leading to late the phosphorylation of Akt, Erk and NF-κB in 16HBE
IL-8 expression [37, 38], in order to elucidate whether the cells within 30 min of stimulation [45]. In parallel, B. ceno-
S. aureus-­induced anti-­inflammatory response was selective cepacia BC7 activates the phosphorylation of p38 and JNK
for bacterial stimuli, 16HBE cells were treated with IL-1β in CF bronchial epithelial IB3 cells, whereas these pathways
and TNFα, resulting in the same amount of IL-8 as the B. remain unphosphorylated in contact with an environmental
cenocepacia-­treated group (Fig.  3d). However, in contrast B. cepacia strain [46]. As K56-2 and BC7 both belong to the
to the reduction seen with S. aureus supernatant and J2315 highly virulent ET-12 lineage, this indicates that hyperacti-
co-­stimulation, S. aureus supernatants did not suppress IL-1β/ vation of host signalling is related to the more severe lung
TNFα-induced IL-8 production, indicating that IL-­1R or inflammation caused by B. cenocepacia isolates.
TNFR signalling is not associated with this process and the
suppressive effect appears to be targeted toward pathways In our model, B. cenocepacia caused the phosphorylation of
involved in the B cenocepacia IL-8 response. p38, Erk and p65 at 120 min post-­infection. Of note, after a
transient increase at 5 min post-­infection, phosphorylation of
Erk peaked again at 120 min. The similar bi-­phasic activation
pattern of Erk and p38 was observed in the response to B.
Discussion pseudomallei both in vitro and in vivo [47]. Thirty minutes
This study is the first to demonstrate the host response caused post-­infection, murine alveolar macrophages exhibited
by S. aureus/B. cenocepacia co-­infection in airway epithelial transient phosphorylation of p38 and Erk1, and at 2 h post-­
cells. We demonstrate that S. aureus-­secreted products, but infection exhibited sustained activation. As Erk regulates
not cell wall-­related components, are able to suppress the apoptosis in response to oxidative stress [48], D’Elia et al.
pro-­inflammatory response induced by B. cenocepacia. The [47] proposed that the transient activation at the early stage
active compound(s) are primarily characterized as small, is associated with host defence, while during the later stage
heat-­stable, non-­proteinaceous, and targeting MAPK and of infection host cells might initiate apoptosis, as indicated
NF-κB signalling. by continuous Erk activation, and by means of this B. pseu-
Previous studies have concentrated on the interactions domallei gains time for intracellular proliferation by delaying
between S. aureus and P. aeruginosa, as they are two preva- apoptosis through modulating the dynamics of Erk activation.
lent pathogens involved in CF lung infections [20, 24, 39–42]. Upon co-­stimulation with B. cenocepacia live bacteria and
However, the impact of S. aureus/Bcc interactions on host S. aureus supernatant, activation of MAPK and NF-κB was
responses has never been assessed. Herein, we demonstrate assessed where MSSA NCTC 6571 supernatant abolished
that in the context of S. aureus/B. cenocepacia co-­infection, phosphorylation of p38, Erk and p65 caused by B. cenoce-
supernatants from S. aureus isolates subverted B. cenocepacia pacia. MRSA252 supernatant did not affect either baseline
live bacteria-­induced IL-8 production in airway epithelial or B. cenocepacia-­induced phosphorylation, which correlates
cells, suggesting that S. aureus releases anti-­inflammatory with its lower inhibitory ability with respect to IL-8 produc-
compounds into the extracellular milieu. This effect of S. tion compared to MSSA NCTC 6571. Our results are in
aureus provides a possible explanation of the protective effect partial agreement with the results displayed by Chekabab et
of S. aureus colonization in terms of lung function and patient al. [41] as they showed that S. aureus supernatants inhibited
survival [27, 28]. P. aeruginosa supernatant-­induced inflammatory response
Interestingly, all of the supernatants from S. aureus isolates through blocking NF-κB activation but not p38. However,
tested were able to subvert B. cenocepacia-­induced IL-8 Chekabab et al. [41] stimulated airway epithelial Beas-­2B cells
production (P<0.05). Even hyperinflammatory isolates such as with synthetic Toll-­like receptor (TLR) 2/1 agonist Pam3CSK4
SH1000 and RN4220, where the supernatants induced strong but not P. aeruginosa, but bacteria themselves contain far
IL-8 production (Fig. 1c), were able to reduce the inflamma- more complicated ligands that could potentially activate
tory response, indicating that the components may be widely the MAPK family. Additionally, the downstream signalling
expressed in S. aureus. The secretome of S. aureus includes a cascades engaged in the anti-­inflammatory response may be
large arsenal of immune evasion factors that inhibit comple- cell type-­specific. For example, in human peripheral blood
ment, chemotaxis, phagocytosis, and induce neutrophil lysis. mononuclear cells, among a group of nasal S. aureus isolates,

1819
Ji et al., Journal of Medical Microbiology 2019;68:1813–1822

some isolates induce anti-­inflammatory IL-10 production staphylococcal anti-­inflammatory factors that suppress B.
through PI3K/Akt/mTOR and Erk signalling whereas some cenocepacia-­ induced IL-8 production through blocking
induce pro-­inflammatory TNFα through a p38-­dependent MAPK and NF-κB signalling. To conclude, we reveal immune
pathway [34]. This suggests that S. aureus manipulates evasion factors secreted by S. aureus that are small, heat-­stable
different signalling cascades to initiate immune tolerance and non-­proteinaceous, and reduce the pro-­inflammatory
through the heterogenic expression of ligands in the process response induced by CF-­isolated B. cenocepacia in 16HBE
of infection. cells. These results suggest a potential treatment for patients
with chronic pulmonary infections, whereby drugs targeting
In further experiments, treatments with heat, proteinase K and
MAPK and NF-κB pathways could be applied to reduce
different molecular weight cut-­off fractions were examined
in order to determine the nature of the anti-­inflammatory inflammation without facilitating infections.
compounds in S. aureus supernatants. Consistent with
previous studies, the compounds were characterized as small Funding information
(<3 kDa) and heat-­resistant [41, 49]. As secondary or tertiary Y. J. was funded by a University of Bath scholarship.
structures of proteins are usually disrupted during boiling, the
Conflicts of interest
staphylococcal factors are unlikely to be proteins. Similarly, The authors declare that there are no conflicts of interest.
when co-­stimulated with B. cenocepacia, IL-8 production elic-
ited by proteinase K-­treated S. aureus supernatants showed a References
50 % decrease compared to deactivated proteinase K-­treated 1. Troeger C, Forouzanfar M, Rao PC, Khalil I, Brown A et  al. Esti-
mates of the global, regional, and national morbidity, mortality, and
S. aureus supernatants, indicating that anti-­inflammatory aetiologies of lower respiratory tract infections in 195 countries: a
components are less likely to be peptides due to the nonspe- systematic analysis for the global burden of disease study 2015.
cific protease activity of proteinase K. A possible explana- Lancet Infect Dis 2017;17:1133–1161.
tion is that proteinaceous pro-­inflammatory components in 2. Pragman AA, Berger JP, Williams BJ. Understanding persis-
S. aureus supernatants were degraded by proteinase K and tent bacterial lung infections: clinical implications informed
by the biology of the microbiota and biofilms. Clin Pulm Med
thus the anti-­inflammatory effectors overpowered the pro-­ 2016;23:57–66.
inflammatory counterparts. For example, the major virulence 3. Tong SYC, Davis JS, Eichenberger E, Holland TL, Fowler VG.
factor SpA not only contributes to immune evasion by binding Staphylococcus aureus infections: epidemiology, pathophysiology,
to immunoglobulin, but also elicits a strong pro-­inflammatory clinical manifestations, and management. Clin Microbiol Rev
response through interacting with tumour necrosis factor 2015;28:603–661.
receptor 1 and epidermal growth factor receptor [37, 50, 51]. 4. Haaber J, Penadés JR, Ingmer H. Transfer of antibiotic resistance
in Staphylococcus aureus. Trends Microbiol 2017;25:893–905.
To date, only one research group has reported small heat-
5. Thammavongsa V, Kim HK, Missiakas D, Schneewind O. Staphylo-
and protease-­resistant molecules derived from MRSA- and coccal manipulation of host immune responses. Nat Rev Microbiol
S. epidermidis-­conditioned medium that induce nitric oxide 2015;13:529–543.
in human upper airway epithelial cells [52, 53]. Although 6. Dasenbrook EC, Checkley W, Merlo CA, Konstan MW, Lechtzin N
these studies did not assess the production of cytokines and et  al. Association between respiratory tract methicillin-­resistant
chemokines in response to the factors, the result strongly Staphylococcus aureus and survival in cystic fibrosis. JAMA
2010;303:2386–2392.
indicates the existence of small heat- and protease-­resistant
molecules that are secreted by S. aureus. 7. Moran GJ, Krishnadasan A, Gorwitz RJ, Fosheim GE,
Albrecht V et al. Prevalence of methicillin-­resistant Staphylococcus
We also investigated whether S. aureus factors interact with aureus as an etiology of community-­ acquired pneumonia. Clin
Infect Dis 2012;54:1126–1133.
other receptors such as IL-­1R and TNFR that promote the
8. Hauser N, Orsini J. Cepacia syndrome in a non-­cystic fibrosis
inflammatory response. The results showed that the inhibi- patient. Case Rep Infect Dis 2015;2015:1–4.
tory effect of S. aureus-­secreted products was B. cenocepacia
9. Kenna DTD, Lilley D, Coward A, Martin K, Perry C et  al. Preva-
stimuli-­specific, as S. aureus supernatants had no effect on lence of Burkholderia species, including members of Burkholderia
IL-1β/TNFα-induced IL-8 production. This is not surprising, cepacia complex, among UK cystic and non-­cystic fibrosis patients.
as S. aureus can colonize human airway asymptomatically, J Med Microbiol 2017;66:490–501.
and therefore is able to trigger a low level of inflammatory 10. LiPuma JJ, Mortensen JE, Dasen SE, Edlind TD, Schidlow DV et al.
response and even suppress it in order to induce immune Ribotype analysis of Pseudomonas cepacia from cystic fibrosis
treatment centers. J Pediatr 1988;113:859–862.
tolerance. Known staphylococcal factors that antagonize
11. Palfreyman RW, Watson ML, Eden C, Smith AW. Induction of
the vital microbial sensor TLRs include staphylococcal biologically active interleukin-8 from lung epithelial cells by
superantigen-­like protein 3 and TIR domain protein targeting Burkholderia (Pseudomonas) cepacia products. Infect Immun
TLR2 [54, 55], and phenol-­soluble modulin α1–3 targeting 1997;65:617–622.
TLR4 [56]. However, little is known about whether the 12. Oliveira MEF, Araújo DG, Oliveira SR. Resistance of non-­fermenting
immunosuppression of S. aureus is beneficial to the host. To gram-­negative bacilli isolated from blood cultures from an emer-
gency Hospital. J Bras Patol Med Lab 2017;53:87–91.
further explore our findings, the impact of S. aureus on B.
13. Shommu NS, Vogel HJ, Storey DG. Potential of metabolomics to
cenocepacia infection needs to be investigated using an in reveal Burkholderia cepacia complex pathogenesis and antibiotic
vivo respiratory infection model. Our findings illustrate for resistance. Front Microbiol 2015;6:668.
the first time the host response to S. aureus and B. cenoce- 14. Cystic Fibrosis Foundation. Cystic Fibrosis Foundation Patient
pacia co-­infection in the airway epithelial cells, and reveal Registry 2016 Annual Data Report. Maryland: Bethesda; 2017.

1820
Ji et al., Journal of Medical Microbiology 2019;68:1813–1822

15. Leitão JH, Sousa SA, Ferreira AS, Ramos CG, Silva IN et al. Patho- 34. Peres AG, Stegen C, Li J, Xu AQ, Levast B et al. Uncoupling of pro-
genicity, virulence factors, and strategies to fight against Burk- and anti-­inflammatory properties of Staphylococcus aureus. Infect
holderia cepacia complex pathogens and related species. Appl Immun 2015;83:1587–1597.
Microbiol Biotechnol 2010;87:31–40. 35. Pandey S, Kawai T, Akira S. Microbial sensing by Toll-­like receptors
16. Urban TA, Goldberg JB, Forstner JF, Sajjan US. Cable pili and the and intracellular nucleic acid sensors. Cold Spring Harb Perspect
22-­kilodalton adhesin are required for Burkholderia cenocepacia Biol 2014;7:a016246.
binding to and transmigration across the squamous epithelium. 36. Hoffmann E, Dittrich-­Breiholz O, Holtmann H, Kracht M.
Infect Immun 2005;73:5426–5437. Multiple control of interleukin-8 gene expression. J Leukoc Biol
17. Urban TA, Griffith A, Torok AM, Smolkin ME, Burns JL et al. Contri- 2002;72:847–855.
bution of Burkholderia cenocepacia flagella to infectivity and 37. Gómez MI, Lee A, Reddy B, Muir A, Soong G et al. Staphylococcus
inflammation. Infect Immun 2004;72:5126–5134. aureus protein A induces airway epithelial inflammatory responses
18. De Soyza A, Silipo A, Lanzetta R, Govan JR, Molinaro A. Chemical by activating TNFR1. Nat Med 2004;10:842–848.
and biological features of Burkholderia cepacia complex lipopoly- 38. Labrousse D, Perret M, Hayez D, Da Silva S, Badiou C et  al.
saccharides. Innate Immun 2008;14:127–144. Kineret®/IL-­1ra blocks the IL-1/IL-8 inflammatory cascade during
19. Tisoncik JR, Korth MJ, Simmons CP, Farrar J, Martin TR et al. Into recombinant Panton Valentine Leukocidin-­ triggered pneumonia
the eye of the cytokine storm. Microbiol Mol Biol Rev 2012;76:16–32. but not during S. aureus infection. PLoS One 2014;9:e97546.
20. Orazi G, O’Toole GA. Pseudomonas aeruginosa Alters Staphy- 39. Ahlgren HG, Benedetti A, Landry JS, Bernier J, Matouk E et  al.
lococcus aureus Sensitivity to Vancomycin in a Biofilm Model of Clinical outcomes associated with Staphylococcus aureus and
Cystic Fibrosis Infection. MBio 2017;8:e00873-17:4. Pseudomonas aeruginosa airway infections in adult cystic fibrosis
patients. BMC Pulm Med 2015;15:918.
21. Perez AC, Pang B, King LB, Tan L, Murrah KA et al. Residence of
Streptococcus pneumoniae and Moraxella catarrhalis within polymi- 40. Baldan R, Cigana C, Testa F, Bianconi I, De Simone M et al. Adapta-
crobial biofilm promotes antibiotic resistance and bacterial persis- tion of Pseudomonas aeruginosa in Cystic Fibrosis airways influ-
tence in vivo. Pathog Dis 2014;70:280–288. ences virulence of Staphylococcus aureus in vitro and murine
models of co-­infection. PLoS One 2014;9:e89614.
22. Ramsey MM, Whiteley M. Polymicrobial interactions stimulate
resistance to host innate immunity through metabolite perception. 41. Chekabab SM, Silverman RJ, Lafayette SL, Luo Y, Rous-
Proc Natl Acad Sci USA 2009;106:1578–1583. seau S et al. Staphylococcus aureus inhibits IL-8 responses induced
by Pseudomonas aeruginosa in airway epithelial cells. PLoS One
23. Bragonzi A, Farulla I, Paroni M, Twomey KB, Pirone L et al. Model- 2015;10:e0137753–19.
ling co-­infection of the cystic fibrosis lung by Pseudomonas aerugi-
nosa and Burkholderia cenocepacia reveals influences on biofilm 42. DeLeon S, Clinton A, Fowler H, Everett J, Horswill AR et  al.
formation and host response. PLoS One 2012;7:e52330. Synergistic interactions of Pseudomonas aeruginosa and Staph-
ylococcus aureus in an in vitro wound model. Infect Immun
24. Fugère A, Lalonde Séguin D, Mitchell G, Déziel E, Dekimpe V et al. 2014;82:4718–4728.
Interspecific small molecule interactions between clinical isolates
43. McCarthy AJ, Lindsay JA. Staphylococcus aureus innate immune
of Pseudomonas aeruginosa and Staphylococcus aureus from adult
evasion is lineage-­ specific: a bioinfomatics study. Infection,
cystic fibrosis patients. PLoS One 2014;9:e86705.
Genetics and Evolution 2013;19:7–14.
25. Korgaonkar A, Trivedi U, Rumbaugh KP, Whiteley M. Commu-
44. Krachler AM, Woolery AR, Orth K. Manipulation of kinase signaling
nity surveillance enhances Pseudomonas aeruginosa viru-
by bacterial pathogens. J Cell Biol 2011;195:1083–1092.
lence during polymicrobial infection. Proc Natl Acad Sci USA
2013;110:1059–1064. 45. Gillette DD, Shah PA, Cremer T, Gavrilin MA, Besecker BY et  al.
Analysis of human bronchial epithelial cell proinflammatory
26. Emerson J, Rosenfeld M, McNamara S, Ramsey B, Gibson RL.
response to Burkholderia cenocepacia infection: inability to secrete
Pseudomonas aeruginosa and other predictors of mortality and
IL-1β. J Biol Chem 2013;288:3691–3695.
morbidity in young children with cystic fibrosis. Pediatr Pulmonol
2002;34:91–100. 46. Sajjan US, Hershenson MB, Forstner JF, LiPuma JJ. Burkholderia
cenocepacia ET12 strain activates TNFR1 signalling in cystic
27. Liou TG, Adler FR, Fitzsimmons SC, Cahill BC, Hibbs JR et  al. fibrosis airway epithelial cells. Cell Microbiol 2008;10:188–201.
Predictive 5-­year survivorship model of cystic fibrosis. Am J Epide-
miol 2001;153:345–352. 47. D'Elia RV, Saint RJ, Newstead SL, Clark GC, Atkins HS. Mitogen-­
activated protein kinases (MAPKs) are modulated during in vitro
28. Mayer-­Hamblett N, Aitken ML, Accurso FJ, Kronmal RA, and in vivo infection with the intracellular bacterium Burkholderia
Konstan MW et  al. Association between pulmonary function and pseudomallei. Eur J Clin Microbiol Infect Dis 2017;36:2147–2154.
sputum biomarkers in cystic fibrosis. Am J Respir Crit Care Med
2007;175:822–828. 48. Li Y, Jiang Z, Xue D, Deng G, Li M et  al. Mycoplasma ovipneumo-
niae induces sheep airway epithelial cell apoptosis through an
29. Nixon GM, Armstrong DS, Carzino R, Carlin JB, Olinsky A et  al. ERK signalling-­ mediated mitochondria pathway. BMC Microbiol
Clinical outcome after early Pseudomonas aeruginosa infection in 2016;16:222.
cystic fibrosis. J Pediatr 2001;138:699–704.
49. Tajima A, Seki K, Shinji H, Masuda S. Inhibition of interleukin-8
30. Cigana C, Bianconi I, Baldan R, De Simone M, Riva C et al. Staphy- production in human endothelial cells by Staphylococcus aureus
lococcus aureus impacts Pseudomonas aeruginosa chronic respira- supernatant. Clin Exp Immunol 2006;147:148–154.
tory disease in murine models. J Infect Dis 2018;217:933–942.
50. Graille M, Stura EA, Corper AL, Sutton BJ, Taussig MJ et al. Crystal
31. Wachsmann P, Lamprecht A. Polymeric nanoparticles for the structure of a Staphylococcus aureus protein A domain complexed
selective therapy of inflammatory bowel disease. Methods Enzymol with the Fab fragment of a human IgM antibody: structural basis
2012;508:377–397. for recognition of B-­cell receptors and superantigen activity. Proc
32. Perret M, Badiou C, Lina G, Burbaud S, Benito Y et  al. Cross-­ Natl Acad Sci USA 2000;97:5399–5404.
talk between Staphylococcus aureus leukocidins-­intoxicated 51. Soong G, Martin FJ, Chun J, Cohen TS, Ahn DS et  al. Staphylo-
macrophages and lung epithelial cells triggers chemokine secre- coccus aureus protein A mediates invasion across airway epithelial
tion in an inflammasome-­ dependent manner. Cell Microbiol cells through activation of RhoA GTPase signaling and proteolytic
2012;14:1019–1036. activity. J Biol Chem 2011;286:35891–35898.
33. Dennehy R, Romano M, Ruggiero A, Mohamed YF, Dignam SL 52. Carey RM, Chen B, Adappa ND, Palmer JN, Kennedy DW et  al.
et al. The Burkholderia cenocepacia peptidoglycan-­associated lipo- Human upper airway epithelium produces nitric oxide in
protein is involved in epithelial cell attachment and elicitation of response to Staphylococcus epidermidis. Int Forum Allergy Rhinol
inflammation. Cell Microbiol 2017;19:e12691. 2016;6:1238–1244.

1821
Ji et al., Journal of Medical Microbiology 2019;68:1813–1822

53. Carey RM, Workman AD, Chen B, Adappa ND, Palmer JN et  al. analysis of staphylococcal genomes: polymorphism and evolution
Staphylococcus aureus triggers nitric oxide production in human of two major pathogenicity islands. J Bacteriol 2008;190:300–310.
upper airway epithelium. Int Forum Allergy Rhinol 2015;5:808–813. 59. Collins J, Rudkin J, Recker M, Pozzi C, O'Gara JP et  al. Offset-
54. Askarian F, van Sorge NM, Sangvik M, Beasley FC, ting virulence and antibiotic resistance costs by MRSA. Isme J
Henriksen JR et  al. A Staphylococcus aureus TIR domain protein 2010;4:577–584.
virulence factor blocks TLR2-­mediated NF-κB signaling. J Innate 60. O'Neill AJ. Staphylococcus aureus SH1000 and 8325-4: compara-
Immun 2014;6:485–498. tive genome sequences of key laboratory strains in staphylococcal
55. Koymans KJ, Goldmann O, Karlsson CAQ, Sital W, Thänert R et al. research. Lett Appl Microbiol 2010;51:358–361.
The TLR2 antagonist staphylococcal superantigen-­like protein 3 61. Nair D, Memmi G, Hernandez D, Bard J, Beaume M et al. Whole-­
acts as a virulence factor to promote bacterial pathogenicity in genome sequencing of Staphylococcus aureus strain RN4220, a
vivo. J Innate Immun 2017;9:561–573. key laboratory strain used in virulence research, identifies muta-
56. Chu M, Zhou M, Jiang C, Chen X, Guo L et al. Staphylococcus aureus tions that affect not only virulence factors but also the fitness of
phenol-­soluble modulins α1–α3 act as novel Toll-­like receptor the strain. J Bacteriol 2011;193:2332–2335.
(TLR) 4 antagonists to inhibit HMGB1/TLR4/NF-κB signaling 62. Voyich JM, Otto M, Mathema B, Braughton KR, Whitney AR et al.
pathway. Front Immunol 2018;9:596–13. Is Panton-­Valentine leukocidin the major virulence determinant
57. Holden MTG, Feil EJ, Lindsay JA, Peacock SJ, Day NPJ et  al. in community-­associated methicillin-­resistant Staphylococcus
Complete genomes of two clinical Staphylococcus aureus strains: aureus disease? J Infect Dis 2006;194:1761–1770.
evidence for the rapid evolution of virulence and drug resistance. 63. Holden MTG, Seth-­Smith HMB, Crossman LC, Sebaihia M,
Proc Natl Acad Sci USA 2004;101:9786–9791. Bentley SD et al. The genome of Burkholderia cenocepacia J2315,
58. Baba T, Bae T, Schneewind O, Takeuchi F, Hiramatsu K. Genome an epidemic pathogen of cystic fibrosis patients. J Bacteriol
sequence of Staphylococcus aureus strain Newman and comparative 2009;191:261–277.

Five reasons to publish your next article with a Microbiology Society journal
1.  The Microbiology Society is a not-for-profit organization.
2.  We offer fast and rigorous peer review – average time to first decision is 4–6 weeks.
3.  Our journals have a global readership with subscriptions held in research institutions around
the world.
4.  80% of our authors rate our submission process as ‘excellent’ or ‘very good’.
5.  Your article will be published on an interactive journal platform with advanced metrics.

Find out more and submit your article at microbiologyresearch.org.

1822

You might also like