Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

| |

Received: 2 July 2019    Revised: 13 August 2019    Accepted: 16 August 2019

DOI: 10.1111/omi.12268

ORIGINAL ARTICLE

Type IX secretion system is pivotal for expression of gingipain‐


associated virulence of Porphyromonas gingivalis

Malgorzata Benedyk1,2  | Agata Marczyk1 | Barbara Chruścicka2

1
Department of Microbiology, Faculty
of Biochemistry, Biophysics and Abstract
Biotechnology, Jagiellonian University, Porphyromonas gingivalis, a keystone pathogen in periodontitis, secretes an array
Krakow, Poland
2 of virulence factors including gingipains via the type IX secretion system (T9SS).
Malopolska Center of
Biotechnology, Jagiellonian University, Inactivation of any component of the T9SS leads to the accumulation of secreted pro‐
Krakow, Poland
teins in unprocessed and, in the case of progingipains, inactive forms in the periplasm.
Correspondence To cast light on the paradox that active gingipains are essential for P. gingivalis fitness
Malgorzata Benedyk, Department of
in vivo but a functional T9SS is not (Frontiers in Cellular and Infection Microbiology,
Microbiology, Faculty of Biochemistry,
Biophysics and Biotechnology, Jagiellonian 2017, 7:378), we have compared virulence of wild‐type P. gingivalis W83 and the
University, Krakow, Poland.
gingipain‐null strain with isogenic mutants deficient in individual T9SS components.
Email: malgorzata.benedyk@uj.edu.pl
Using an in vivo subcutaneous chamber mouse model of infection, gingipain‐null
Funding information
strain secretion mutants showed no virulence, but their pathogenic potential was
National Science Center Poland, Grant/
Award Number: 2017/01/X/NZ6/01953 reconstituted by coinfection with a low number of the parental strain. Apparently
The peer review history for this article is available the same mechanism compensated fitness of mutants lacking functional T9SS the
at https​://publo​ns.com/publo​n/10. 1111/
transposon library. In contrast to the parental strain, all mutants elicited significantly
omi.12268/​
lower but an effective inflammatory immune response, which cleared infection and
prevented systemic dissemination of P. gingivalis to organs. There were no significant
differences in immune responses to different secretion mutants, which were gener‐
ally more stimulatory than the gingipain‐null strain. Together, these results indicate
that functional T9SS is essential for P. gingivalis virulence apparently through delivery
of active gingipains to the bacterial surface. Therefore, T9SS is a legitimate target for
drug development to treat periodontitis.

KEYWORDS
fitness, gingipain, periodontitis, Porphyromonas gingivalis, protein secretion, T9SS, virulence

1 |  I NTRO D U C TI O N atherosclerotic cardiovascular disease (Tonetti & Van Dyke, 2013),
diabetes (Chapple, Genco, Worksh, & Working Group 2 of the Joint
Periodontitis is a chronic inflammatory disease of the tooth‐support‐ EFP/AAP Workshop, 2013), and rheumatoid arthritis (Potempa,
ing tissues. It is characterized by a progressive loss of attachment Mydel, & Koziel, 2017). The complex pathogenesis of periodontitis
caused by the destruction of the connective tissue and alveolar bone, implies interplay between the host response, cumulative effects
which can lead to tooth loss (Pihlstrom, Michalowicz, & Johnson, of various risk factors, and bacterial challenge posed by the dental
2005). Globally, mild to moderate periodontitis affects nearly plaque (Hajishengallis & Korostoff, 2017).
half of the human population, with severe periodontitis affecting Over the last two decades, extensive investigations of the oral
about 11% of individuals (Kassebaum et al., 2014). Moreover, peri‐ microbiome established that a wide variety of organisms are asso‐
odontitis is associated with various systemic conditions, including ciated with periodontal pathogenesis. In a synergistic manner they

Mol Oral Microbiol. 2019;34:237–244. wileyonlinelibrary.com/journal/omi   © 2019 John Wiley & Sons A/S. |  237
Published by John Wiley & Sons Ltd
|
238       BENEDYK et al.

cause a shift in the microbial composition characterized by increased mouse model. To this end, we quantified numbers of bacteria in
levels of so‐called red‐complex bacteria, Porphyromonas gingivalis, chambers post‐inoculation, determined their ability to disseminate,
Treponema denticola, and Tannerella forsythia in the subgingival mi‐ and analyzed the local and systemic immune responses elicited by
croflora (Hajishengallis, Darveau, & Curtis, 2012; Hajishengallis & individual mutants.
Lamont, 2012; How, Song, & Chan, 2016). These anaerobic, Gram‐
negative microbes are uninvited and unwelcome guests in the very
2 | M ATE R I A L S A N D M E TH O DS
diverse community of microbial biofilm on the tooth surface and their
proliferation significantly changes the composition of the community
2.1 | Bacterial strains and cultures
(microbiota shift) (Hajishengallis, 2015) with all pathological conse‐
quences then following (Lamont & Hajishengallis, 2015). Among the The wild‐type strain of P. gingivalis (W83), its four T9SS mutants
red complex, P. gingivalis is the keystone pathogen with unsurpassed (∆Sov, ΔPorT, ΔPorZ, and ΔPorU; Table 1) and gingipains‐deletion
potential to initiate the transition of the commensal bacterial micro‐ null mutant (∆KRAB, Table 1) were grown under anaerobic condi‐
biota into a dysbiotic one that drives chronic inflammation in the tions (90% N2, 5% CO2, 5% H2) on blood (5% v/v sheep blood) agar
periodontium (Zenobia & Hajishengalliss, 2015). plates or in liquid tryptic soy broth (TSB; Sigma‐Aldrich) and yeast
Gram‐negative, anaerobic P. gingivalis bacterium expresses extract (5 mg/ml; BioShop) supplemented with hemin (5 μg/ml;
several well‐established virulence factors, including lipopolysac‐ Sigma‐Aldrich), l‐cysteine (250 μg/ml; Sigma‐Aldrich), and menadi‐
charide, phosphatase, fimbriae, hemagglutinins, and cysteine pro‐ one (0.5 μg/ml; Sigma‐Aldrich). The ∆Sov, ΔPorT, and ∆KRAB strains
teinases called gingipains (Bostanci & Belibasakis, 2012). The latter were cultured in medium supplemented with tetracycline (1 μg/ml;
are the most important P. gingivalis virulence factor with respect to Sigma‐Aldrich), and the ΔPorU and ΔPorZ strains were cultured in
pathogenicity and survival in vivo (Guo, Nguyen, & Potempa, 2010). medium supplemented with erythromycin (5 μg/ml; Sigma‐Aldrich).
Gingipains are encoded by three distinct genes: rgpA, rgpB, and kgp After overnight culture, bacteria were centrifuged (4,500 × g, 10 min)
(Potempa, Sroka, Imamura, & Travis, 2003). The Arg‐specific (RgpB to prepare inoculates. Subsequently, the bacterial pellet was washed
and RgpA) and Lys‐specific (Kgp) gingipains have been shown to three times with phosphate‐buffered saline (0.14 M NaCl, 0.0026 M
play a pivotal role in nutrient acquisition, colonization, penetration KCl, 0.01 M Na2HPO 4, 0.002 M KH2PO 4), pH 7.4 (PBS), and resus‐
into host tissue, deregulation of the immune response, and devel‐ pended in fresh PBS. Bacterial cell counts were standardized to an
opment of dysbiosis (Benedyk et al., 2016; Fleetwood et al., 2015; optical density of 1.0 at 600 nm (corresponding to 1 × 109 colony‐
Hajishengallis et al., 2012; Hajishengallis & Lamont, 2014). forming units [CFUs]/ml).
Gingipains are secreted to the bacterial cell surface and beyond
in a two‐step process (Fleetwood et al., 2015). First, preprogingipains
2.2 | Sov mutant construction
translocate the inner membrane using secretion pathways known as
the Sec system, and then progingipains across the outer membrane Using a similar strategy to that described previously to create the
using a protein secretion apparatus known as the type IX secretion ΔRgpA mutant (Nguyen et al., 2007), two 1.0 kb flanking regions,
system (T9SS) (Lasica, Ksiazek, Madej, & Potempa, 2017). The latter one 5′ and one 3′ to the sov gene (The Institute of Genomic Research
step is dependent on the presence of a conserved carboxy‐terminal [TIGR] accession numbers: PG0809 and PG0810), were amplified
domain, which tags them for secretion across the outer membrane by PCR using AccuPrime Pfx DNA Polymerase (Invitrogen Inc.)
by the T9SS (Nguyen, Travis, & Potempa, 2007). and inserted into the pUC19 plasmid (New England Biolabs Inc.) at
The T9SS is composed of several components localized in the
cytoplasmic membrane (PorL and PorM), in the periplasm (PorK and
PorN), in the outer membrane (β‐barrel proteins Sov, PorT, PorQ, TA B L E 1   Porphyromonas gingivalis strains used in this study
PorV, and PorP), and on the bacterial surface (PorU and PorZ) Strain Relevant genotype Source
(Lasica et al., 2017). Deletion of any component of that complex
P.g W83 WT Wild type Reference
system results in retention of the proteolytically inactive progin‐
strain
gipains in the mutant periplasm, together with 30 other proteins
P.g ∆PorU porU (NCBI: PG_RS00120; old Lasica et al.
transported by the T9SS (Nikolich, Shoemaker, & Salyers, 1992). locus PG0026) (Emr) (2016)
Interestingly, however, none of the T9SS components is essential P.g ∆PorT porT (NCBI: PG_RS03295; old Nguyen et al.
for the growth of P. gingivalis in epithelial cells and murine skin ab‐ locus PG0751) (Tcr) (2009)
scesses. This is a paradox considering that several proteins trans‐ P.g ∆PorZ porZ (NCBI: PGRS07070; old locus Lasica et al.
ported by this system, including gingipains, are absolutely essential PG1604 (Emr) (2016)
for P. gingivalis fitness in these models (Miller et al., 2017). Herein, P.g ∆Sov sov (NCBI: RC03550; old locus This study
we compared the propensity of P. gingivalis wild‐type (WT) strain PG0809/PG0810) (Tcr)
and individual secretory mutants (porU, ∆PorU; porZ, ∆PorZ; porT, P.g ∆KRAB kgpΔ598 rgpArgpBΔ410 Tcr Cmr Emr Rapala‐Kozik
∆PorT and sov, ∆Sov) to survive and proliferate in subcutaneous et al. (2011)

chambers or be eliminated by the immune system in an in vivo Abbreviations: P.g, Porphyromonas gingivalis; WT, wild‐type.
BENEDYK et al. |
      239

the SacI/SmaI and XbaI/SalI sites, respectively. All primers used in


2.5 | CFU assay
this study are listed in Table 2. An intervening tetracycline resist‐
ance cassette (tetQ) from the pNFD13‐2 plasmid (Saiki & Konishi, After 24, 48, and 72  hr, 100  µl of the fluids were taken from the
2007) was amplified and inserted between the flanking regions in chamber and then 10‐fold serially diluted chamber fluids were plated
the modified pUC19 at the SmaI/XbaI site to create the final plas‐ on blood agar plates and cultured anaerobically for 14 days at 37°C.
mid construct, pSovAtB‐C. Correct placement and orientation of the Thereafter, the number of visible colonies was counted and the total
DNA segments were confirmed by sequencing. Purified plasmids number of CFUs was calculated by multiplying this number by the
were electroporated into electro‐competent P. gingivalis W83 cells dilution factor.
as described previously (Nguyen et al., 2007) and integration of the
plasmid construct into the P. gingivalis genome by a double crossover
2.6 | Myeloperoxidase assay
recombination event was selected for using 1 µg/ml tetracycline in
TSB agar. Deletion of the sov gene in the resulting mutants was con‐ Polymorphonuclear neutrophil (PMN) influx into the subcutane‐
firmed by PCR and DNA sequencing. ous chambers was analyzed by measuring the enzymatic activity of
myeloperoxidase (MPO; as a marker of PMN accumulation). Briefly,
24 hr after bacterial injection into the chamber, 50 µl of 25‐fold di‐
2.3 | Experimental animals and ethical approval
luted chamber fluid was mixed with 100 µl o‐phenylenediamine dihy‐
Specific pathogen‐free (SPF) female BALB/c mice, 8–10  weeks of drochloride (OPD) reagent (5 mg OPD, 5 µl 30% hydrogen peroxide,
age, were purchased from Jackson Laboratory. Mice were housed in 5 ml buffer containing 0.05 M citric acid, and 0.05 M sodium phos‐
individually ventilated cages and fed a standard laboratory diet and phate, pH 5) and incubated for 10 min at room temperature in the
water ad libitum in SPF conditions within the animal care facility at the dark. The reaction was terminated by addition of 50 µl 2.5 M H2SO4.
Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian Then, absorbance was measured at 490 nm using a microplate reader
University, Krakow, Poland. Mice were maintained under a 60 ± 5% (SpectraMax Gemini; Molecular Devices) as direct accumulation
relative humidity 12 hr‐light/dark cycle and at 22 ± 2°C. Control and of MPO, which has been expressed as a percentage of the control
bacterially infected mice were housed in separate cages. All animal groups (chamber fluid from animals inoculated with PBS only).
experiments were reviewed and approved by the I Regional Ethics
Committee on Animal Experimentation, Krakow, Poland (approval
2.7 | Gingipain activity
no: 131/2018).
On day 1 (24 hr after bacterial injection into the chamber), the pro‐
teolytic activity of P. gingivalis gingipains was measured. Briefly,
2.4 | Subcutaneous chamber model procedure
5 µl chamber fluid was added to 95 µl TGC buffer (100 mM Tris
To enable the best operative outcome, all mice used had a minimum pH 7.6, 200  mM Gly‐Gly, 5  mM CaCl2, 100 µg/ml PEFABLOCK,
weight of 20 g. All mice were anesthetized by intraperitoneal in‐ and 20 µg/ml aprotinin) supplemented with 10 mM l‐cysteine in
jection of ketamine (22 mg/kg; VetaKetam, VetAgro) and xylazine 96‐well microtitre plates. After 5  min preincubation at 37°C, a
(2 mg/kg; Sedasin, Biowet), and the eyes were lubricated with oint‐ substrate of gingipain R (benzoyl‐arginine‐p‐nitroanilide [BAPNA];
ment (Puralube Vet; Pharmaderm). Following general anesthesia, Sigma‐Aldrich) was added to give a working concentration of 1 mM.
using sterile instruments, the skin was incised on the right side of The protease activity of gingipain R was represented by the release
the back and a sterile titanium‐coil chamber was implanted subcu‐ of p‐nitroaniline from the substrate by determination of absorb‐
taneously. After 10 days, the mice were injected into the lumen of ance at 405 nm using a microplate reader (SpectraMax Gemini;
the subcutaneous chamber with 1 × 109 CFUs of WT P. gingivalis Molecular Devices).
W83, ∆Sov, ΔPorT, ΔPorZ, ΔPorU, or ∆KRAB resuspended in a total
volume of 80 µl sterile PBS. Control mice were injected with 80 µl
2.8 | Cytokine and C‐reactive protein measurement
sterile PBS into subcutaneous chambers (sham‐infection).
The level of IL‐6, TNFα, IL‐10, MCP‐1 in serum samples was deter‐
mined using a CBA Mouse Inflammatory Cytokine Kit (BD Bioscience)
according to the manufacturer's instructions and results were ana‐
TA B L E 2   Primers used for SOV mutant construction
lyzed using an FCAP Array v2.0.2 (BD Bioscience). Briefly, 24 hr after
SovFrASacIF ctGAGCTCGCCTACAATCTCAAGCGTATGC bacterial injection into the chamber, mice were sacrificed by cervi‐
cal dislocation, and whole blood was immediately collected and al‐
SovFrASmaIR ttaaCCCGGGCGTGATCCTTCTTTGTTTGAGA
lowed to clot at room temperature (60 min), and then centrifuged at
SovFrBXbaIF acTCTAGATAGCCTAACGGCATTACCCAC
2,000 × g for 10 min to separate the serum. C‐reactive protein (CRP)
SovFrBSalIR attaGTCGACCTTGTATGCAGGAAGCAGGATAG
levels in the serum were measured using a commercially available
tetQSmaIF tataCCCGGGACAACGAATTATCTCCTTAACGT
ELISA (Mouse CRP ELISA kit; Innovative Research), according to the
tetQXbaIR cgcTCTAGATTTTATTGCCAAGTTCTAATGCT
manufacturer's recommendations.
|
240       BENEDYK et al.

while subcutaneous chamber inocula of WT W83 at 107 CFUs and


2.9 | Statistical analysis
the ∆Sov mutant at 109 CFUs alone each had no effect, when ad‐
All the data were reported as the mean  ±  standard deviation (SD). ministered together they caused sickness and eventually killed the
Significant differences between groups were examined using mice (Figure 1b). This result suggests that the WT strain restored the
Student's t test or two‐way repeated measures ANOVA (multi‐pa‐ virulence of the secretory mutant.
rameter data) and one‐way ANOVA with the Bonferroni correction Levels of gingipains in the chamber fluid were evaluated at the
or Dunnett's post‐test (multiple comparisons). Kaplan–Meier graphs end of the experiment and were found at very low (Rgp) or unde‐
were analyzed using the log‐rank test with the Bonferroni correction tectable (Kgp) levels (Figure 2). This confirms that the lack of any
(post‐test) to correct for multiple comparisons. All statistical analyses component of the T9SS impairs the secretion of gingipains (Figure 2)
were performed using GraphPad Prism software (v5.0d GraphPad). and probably that of all other cargo proteins in vivo.
Values of p ≤ .05 were considered to be statistically significant.

3.2 | Functional T9SS is necessary for P. gingivalis


3 |   R E S U LT S survival in subcutaneous chambers
To establish the importance of T9SS components on P. gingivalis
3.1 | Inhibition of gingipain secretion prevents
survival/proliferation in vivo, we determined the number of bacteria
fatality in the subcutaneous chamber model
(CFUs) in the chamber fluid 24 hr after inoculation. We found that
A subcutaneous chamber infection mouse model was used to com‐ while the number of CFUs of WT P. gingivalis W83 increased by 30‐
pare the virulence of the WT P. gingivalis W83 and the gingipain‐null fold, CFUs of all strains deficient in the functional T9SS significantly
mutant ∆KRAB with secretion‐mutant strains ∆PorU, ∆PorZ, ∆PorT, decreased in numbers (Figure 3). Although there were variations
or ∆Sov. When 1 × 109 CFUs of bacteria were inoculated into the in CFUs numbers among secretion mutants, with a tendency for
chamber, only WT P. gingivalis W83 bacteria were lethal, with all ∆PorU > ∆PorZ > ∆PorT > ∆Sov, the differences between individual
mice dying by 48  hr. By contrast, animals infected with the same strains were not significant. On the other hand, the gingipain‐null
dose of the gingipain‐null mutant ∆KRAB strain or secretion mu‐ mutant strain ∆KRAB was cleared more efficiently than secretion
tants (∆PorU, ∆PorZ, ∆PorT, and ∆Sov) showed no signs of disease
throughout the course of the experiment (Figure 1a). Furthermore,

F I G U R E 2   Gingipain activity is at a very low or undetectable


F I G U R E 1   Functional T9SS is necessary to maintain level in chamber fluid after infection with secretion mutants.
Porphyromonas gingivalis virulence. BALB/c mice were challenged Arginine‐specific (a) and lysine‐specific (b) gingipain activity
with wild‐type (WT) P. gingivalis W83, four different P. gingivalis was determined using l‐BAPNA and N‐(p‐Tosyl)‐Gly‐Pro‐Lys‐4‐
T9SS‐secretion mutants (ΔPorU, ΔPorT, ΔSov, or ΔPorZ), and nitroanilide acetate salt as a substrate, respectively. Data are
gingipain‐null mutant (ΔKRAB), each at 1 × 109 CFUs (a) or with presented as means ± SD of assays performed in triplicate and
P. gingivalis W83 (1 × 107 CFUs) alone or with ΔSov (1 × 109 CFUs) were analyzed by one‐way ANOVA with the Bonferroni post‐test
(b). Mouse survival was monitored throughout the entire procedure correction (***p < .0001)
BENEDYK et al. |
      241

the ∆KRAB group and around sevenfold higher than in the groups
challenged with T9SS knockouts (∆PorU, ∆PorZ, ∆PorT, or ∆Sov;
Figure 4a). In contrast to the number of PMNs infiltrating chambers
being similar for all secretion mutants, the level of lymphocytes and
monocytes in the chamber fluid showed the following trends: ∆PorZ > 
∆PorT > ∆Sov > ∆PorU > ∆KRAB and ∆PorU > ∆PorT > ∆PorZ > ∆Sov 
> ∆KRAB, respectively (Figure 4b,c). The trend observed for monocyte
levels reflects the number of surviving bacteria in the chamber, which
was highest in ∆PorU and the lowest in ∆KRAB (Figure 3).
In addition to cytological evaluation, MPO activity was mea‐
sured in the chamber fluid to estimate PMN infiltration and ac‐
tivation. In line with cytological findings, the MPO activity was
F I G U R E 3   Functional T9SS is essential for Porphyromonas
gingivalis survival and proliferation in chambers. Mice were
challenged with 1 × 109 CFUs wild‐type (WT) P. gingivalis W83,
T9SS‐secretion mutants (ΔPorU, ΔPorT, ΔSov, or ΔPorZ) and
gingipain‐null strain ΔKRAB. Survival of bacteria in the chamber
was assessed by counting CFUs on agar plates inoculated with
chamber fluid at 24 hr post‐infection. Data are presented as
means ± SD and were analyzed by one‐way ANOVA (***p < .0001)

mutants, suggesting that gingipains are of pivotal importance for


P. gingivalis survival under attack by the immune system. In keeping
with WT P. gingivalis W83 having the exclusive ability to evade the
host immune system and proliferate inside chambers, only this strain
escaped from the chamber and systematically disseminated to vari‐
ous organs, including the lungs, liver, and kidneys as shown by plating
homogenates from these tissues prepared at 24 and 48 hr post‐in‐
oculation (Table 3). This confirms the importance of the functional
T9SS for the virulence of P. gingivalis in vivo and reaffirms the validity
of the T9SS as a drug target for the treatment of periodontitis.

3.3 | Functional T9SS is pivotal for robust host


inflammatory response
Cytological analysis of the chamber fluid showed the presence of
PMNs, lymphocytes, and monocytes regardless of the inoculated
strain. However, the number of each inflammatory cell type in the
chamber fluid determined 24 hr post‐challenge was far higher in ani‐
mals infected with WT P. gingivalis W83 than with any other strain.
The number of PMNs was around eightfold higher as compared with

TA B L E 3   Presence of bacteria in organs at 24 hr post‐infection

  Lungs Liver Kidney Spleen

P.g W83 WT + + + + F I G U R E 4   Operational T9SS is necessary for the recruitment


of inflammatory cells to the site of inflammation. Numbers of
P.g ∆PorU − − − −
polymorphonuclear neutrophils (PMNs) (a), lymphocytes (b), and
P.g ∆PorT − − − − monocytes (c) in chamber fluid after challenge (24 hr) with wild‐
P.g ∆PorZ − − − − type (WT) Porphyromonas gingivalis W83, T9SS‐secretion mutants
P.g ∆Sov − − − − (∆PorU, ∆PorT, ∆PorZ, or ∆Sov), and gingipain‐null mutant (ΔKRAB)
were counted in smears stained using Wright's stain. Cells were
P.g ∆KRAB − − − −
identified by morphology. Data are presented as a means ± SD
Abbreviations: P.g, Porphyromonas gingivalis; WT, wild‐type. (***p < .0001)
|
242       BENEDYK et al.

in the level of PMN activation was observed among the secretion


mutants.

3.4 | Functional T9SS is crucial for the


development of systemic inflammatory responses
during subcutaneous chamber infection with
P. gingivalis
Cytokine and chemokine production are an important part of the
host's defense system and play a critical role in regulation of the
inflammatory response against bacterial infections. To assess the
F I G U R E 5   Functional T9SS is important for polymorphonuclear
neutrophil activation in Porphyromonas gingivalis‐infected role of the T9SS in P. gingivalis‐stimulated systemic inflammation,
chambers. Subcutaneous chambers in BALB/c mice were inoculated the concentration of TNFα, MCP‐1, IL‐6, and IL‐10 in the peripheral
with 1 × 109 CFUs wild‐type (WT) P. gingivalis W83, T9SS‐secretion blood was assessed 24 hr post‐inoculation. As expected, in the blood
mutants (∆PorU, ∆PorT, ∆PorZ, or ∆Sov), and the gingipain‐null of animals infected with WT P. gingivalis W83, a strong, statistically
strain (∆KRAB). Then, 24 hr post‐infection myeloperoxidase (MPO)
significant increase of all cytokines tested was observed. Infection
activity in chamber fluid was determined. The MPO activity was
with ∆PorU, ∆PorZ, ∆PorT, ∆Sov, or ∆KRAB resulted in only a small
expressed as a % of control activity (control mice challenged with
PBS) increase in cytokine levels (Figure 6a–d). Similarly, CRP, another im‐
portant marker of inflammation, showed the highest increase in the
around fourfold higher in the chamber fluid from P. gingivalis W83 blood of animals challenged with WT P. gingivalis W83, while chal‐
infected animals compared with ∆PorU‐, ∆PorZ‐, ∆PorT‐, ∆Sov‐, lenge with secretion mutants (∆PorU, ∆PorZ, ∆PorT, and ∆Sov) and
or ∆KRAB‐infected animals (Figure 5). In this case, no clear trend the gingipain‐null mutant ∆KRAB caused a moderate increase during

F I G U R E 6   Inactivation of T9SS
suppresses the immune response to
Porphyromonas gingivalis. Subcutaneous
chambers in BALB/c mice were
challenged with 1 × 109 CFUs wild‐type
(WT) P. gingivalis W83, T9SS‐secretion
mutants (∆PorU, ∆PorT, ∆PorZ, or ∆Sov)
or gingipain‐null strain (∆KRAB). Then,
24 hr post‐infection level of IL‐6 (a), TNFα
(b), MCP‐1 (c), IL‐10 (d), and CRP (e) was
determined in blood serum. Significance
of observed differences between
P. gingivalis W83 WT and P. gingivalis
∆PorU‐, ∆PorT‐, ∆PorZ‐, ∆Sov‐, and
∆KRAB‐challenged animals was verified
with Student's t test, *p < .001, **p <
.0013, ***p < .0001 (a–d) or with one‐way
ANOVA test, ***p < .0001 (e)
BENEDYK et al. |
      243

the infection (Figure 6e). Interestingly, in the case of IL‐6 and TNFα, a involved in cleavage of the CTD and attachment of T9SS cargo pro‐
trend similar to that observed for CFUs, lymphocyte, and monocyte teins, it is interesting to note a clear trend for the higher CFU and
numbers in the chamber fluid was noticed (Figure 6a,b). monocyte accumulation in chambers inoculated with delta‐PorU
than delta‐Sov. This may be related to the “partial secretion” pheno‐
type of the former mutant in which significant amount of T9SS cargo
4 | D I S CU S S I O N proteins were found on the cell surface (Glew et al., 2012).
Previous studies using the subcutaneous chamber model
The protein secretion system known as the T9SS is present in many have shown that challenge with bacteria possessing a properly
species of the Bacteroidetes phylum. In P. gingivalis, the keystone per‐ functioning T9SS, and therefore capable of secretion of pro‐
iodontal pathogen T9SS is responsible for the secretion of about 30 teins including gingipains, leads to exaggerated inflammation
different proteins, including pivotal virulence factors: the gingipains and immune activation, and an increase in bacterial tissue bur‐
(Abby et al., 2016; Nguyen et al., 2009). It is surprising that to date no den with resultant bacteria dissemination, chamber exfoliation,
virulence analysis of T9SS‐secretion mutants has been carried out tissue injury, and ultimately death. The microbicidal mechanisms
using an in vivo model of infection necessary to confirm the impor‐ of phagocytes with production of TNF and other cytokines seem
tance of T9SS for the pathogenicity of P. gingivalis. to be primarily responsible for the necrotic lesions of infection.
In this study, we used the well‐established subcutaneous cham‐ None of the above effects were observed after chamber inoc‐
ber model that allows monitoring of bacterial growth or eradication ulation with secretion mutants (∆PorT, ∆PorU, ∆PorZ, or ∆Sov)
of infection in vivo. This provides a reliable way to sample and eval‐ or the gingipain‐null mutant ∆KRAB, confirming the pivotal role
uate inflammatory exudates to study the host local and systemic of gingipains as the primary virulence factor responsible for an
immune response. Results of our in vivo experiments conclusively immediate and uncontrolled inflammatory response, bacteria dis‐
demonstrated that a functional T9SS is pivotal for P. gingivalis growth, semination, and subsequently death of the animals (Guo et al.,
dissemination, and pathogenic effects in vivo, including induction of 2010; Kadowaki et al., 2007; Lin, Huang, Lai, Huang, & Hu, 2005).
a strong inflammatory reaction. Disabling the T9SS in P. gingivalis These results are in line with our previous studies using an animal
(as in the ∆PorU, ∆PorZ, ∆PorT, and ∆Sov mutants) led to a dras‐ model of aspiration pneumonia, where infection with P. gingivalis
tic decrease in CFUs in the chamber, which is in stark contrast to W83 induced fatal pneumonia, while the gingipain‐null mutant
WT P. gingivalis, which proliferated. Despite this finding, none of the ∆KRAB induced only a transient and non‐threatening infection
components of T9SS disabled by gene deletion were found to con‐ (Benedyk et al., 2016).
tribute to P. gingivalis fitness in three different models, in contrast In summary, we conclude that the complex T9SS and all of its
to several cargo proteins (Miller et al., 2017). Apparently, inoculating identified components play an important role at the site of infec‐
with the transposon library of mutants, the lack of functional T9SS tion as the source of gingipain, the most important virulence fac‐
was compensated by the mutants with a functional secretion system. tor of P. gingivalis. More importantly, our data clearly confirm that
This contention was confirmed in the present study with the find‐ all components of T9SS are a viable target for the development
ing that a relatively small contingent of WT P. gingivalis restored the of novel treatment strategies for periodontitis as T9SS inhibition
bacterium virulence in vivo. It can be speculated that T9SS is leaky leads to utter loss of bacterial virulence and quick clearance from
and transported proteins essential for fitness are released into the the tissue.
outside environment where they are processed by extracellular com‐
ponents of T9SS from secretion‐competent mutants in the library.
AC K N OW L E D G E M E N T S
Such a mechanism would be especially important in processing
and activating gingipains. Only infection with WT P. gingivalis W83 This work was funded by the National Science Center, Poland
was able to trigger a local and systemic immune response as mani‐ (2017/01/X/NZ6/01953).
fested by the mass influx of inflammatory cells into the chamber and
the increase in proinflammatory cytokine production. Furthermore,
C O N FL I C T O F I N T E R E S T
only in response to infection with WT P. gingivalis W83 we did ob‐
serve a rapid recruitment of a large number of lymphocytes, PMNs, The authors have no conflicts of interest to declare.
and monocytes to the site of infection, leading to increased produc‐
tion of IL‐6, TNFα, MCP‐1, and IL‐10 cytokines as well as an increase
ORCID
in CRP. Apparently, the lack of gingipain activity in T9SS mutants
most significantly contributed to abolishment of P. gingivalis viru‐ Malgorzata Benedyk  https://orcid.org/0000-0001-5432-8739
lence, which corroborates the finding that inhibition of gingipains
completely eliminates P. gingivalis virulence in vivo (Bostanci &
Belibasakis, 2012; Guo et al., 2010; Kadowaki et al., 2007). Finally, REFERENCES

taking into account that Sov is the OM beta‐barrel secretion pore Abby, S. S., Cury, J., Guglielmini, J., Néron, B., Touchon, M., & Rocha, E.
essential for OM translocation while PorU is the surface sortase P. C. (2016). Identification of protein secretion systems in bacterial
|
244       BENEDYK et al.

genomes. Scientific Reports, 6, 23080. https​://doi.org/10.1038/srep2​ of PorZ, an essential bacterial surface component of the type-IX
3080 secretion system of human oral-microbiomic Porphyromonas gingiva‐
Benedyk, M., Mydel, P., Delaleu, N., Płaza, K., Gawron, K., Milewska, A., lis. Scientific Reports, 6, 37708. https​://doi.org/10.1038/srep3​7708
… Potempa, J. (2016). Gingipains: Critical factors in the development Lasica, A. M., Ksiazek, M., Madej, M., & Potempa, J. (2017). The Type
of aspiration pneumonia caused by P. gingivalis. Journal of Innate IX Secretion System (T9SS): Highlights and recent insights into its
Immunity, 8(2), 185–198. https​://doi.org/10.1159/00044​1724 structure and function. Frontiers in Cellular and Infection Microbiology,
Bostanci, N., & Belibasakis, G. N. (2012). Porphyromonas gingivalis: An in‐ 7, 215. https​://doi.org/10.3389/fcimb.2017.00215​
vasive and evasive opportunistic oral pathogen. FEMS Microbiology Lin, Y. Y., Huang, J. H., Lai, Y. Y., Huang, H. C., & Hu, S. W. (2005). Tissue destruc‐
Letters, 333, 1–9. https​://doi.org/10.1111/j.1574-6968.2012.02579.x tion induced by P. gingivalis infection in a mouse chamber model is asso‐
Chapple, I. L. C., Genco, R., Worksh, W. G., & Working Group 2 of the ciated with host tumor necrosis factor generation. Infection and Immunity,
Joint EFP/AAP Workshop. (2013). Diabetes and periodontal diseases: 73(12), 7946–7952. https​://doi.org/10.1128/IAI.73.12.7946-7952.2005
Consensus report of the Joint EFP/AAP Workshop on Periodontitis and Miller, D. P., Hutcherson, J. A., Wang, Y., Nowakowska, Z. M., Potempa,
Systemic Diseases (Reprinted from Journal of Clinical Periodontology, J., Yoder‐Himes, D. R., … Lamont, R. J. (2017). Genes contributing to
vol 40, pg S106–S112, 2013). Journal of Periodontology, 84, S106–S112. P. gingivalis fitness in abscess and epithelial cell colonization environ‐
https​://doi.org/10.1902/jop.2013.1340011 ments. Frontiers in Cellular and Infection Microbiology, 7, 378. https​://
Fleetwood, A. J., O'Brien‐Simpson, N. M., Veith, P. D., Lam, R. S., doi.org/10.3389/fcimb.2017.00378​
Achuthan, A., Cook, A. D., … Hamilton, J. A. (2015). Porphyromonas Nguyen, K. A., Travis, J., & Potempa, J. (2007). Does the importance of the
gingivalis‐derived RgpA‐Kgp complex activates the macrophage uro‐ C‐terminal residues in the maturation of RgpB from Porphyromonas
kinase plasminogen activator system: Implications for Periodontitis. gingivalis reveal a novel mechanism for protein export in a subgroup
Journal of Biological Chemistry, 290, 16031–16042. https​://doi. of Gram‐negative bacteria? Journal of Bacteriology, 189, 833–843.
org/10.1074/jbc.M115.645572 https​://doi.org/10.1128/JB.01530-06
Glew, M. D., Veith, P. D., Peng, B., Chen, Y. Y., Gorasia, D. G., Yang, Q., Nguyen, K. A., Zylicz, J., Szczesny, P., Sroka, A., Hunter, N., & Potempa, J.
… Reynolds, E. C. (2012) PG0026 is the C‐terminal signal peptidase (2009). Verification of a topology model of PorT as an integral outer
of a novel secretion system of Porphyromonas gingivalis. Journal of membrane protein in Porphyromonas gingivalis. Microbiology, 155,
Biological Chemistry, 287(29), 24605‐24617. https​://doi.org/10.1074/ 328–337. https​://doi.org/10.1099/mic.0.024323-0
jbc.M112.369223 Nikolich, M. P., Shoemaker, N. B., & Salyers, A. A. (1992). A Bacteroides tet‐
Guo, Y., Nguyen, K.‐A., & Potempa, J. (2010). Dichotomy of gingipains racycline resistance gene represents a new class of ribosome protec‐
action as virulence factors: From cleaving substrates with the pre‐ tion tertracycline resistance. Antimicrobial Agents and Chemotherapy,
cision of a surgeon's knife to a meat chopper‐like brutal degra‐ 36(5), 1005–1012. https​://doi.org/10.1128/aac.36.5.1005
dation of proteins. Periodontology 2000, 54, 15–44. https​ ://doi. Pihlstrom, B. L., Michalowicz, B. S., & Johnson, N. W. (2005). Periodontal
org/10.1111/j.1600-0757.2010.00377.x diseases. Lancet, 366, 1809–1820. https​://doi.org/10.1016/
Hajishengallis, G. (2015). Periodontitis: From microbial immune subver‐ S0140-6736(05)67728-8
sion to systemic inflammation. Nature Reviews Immunology, 15, 30– Potempa, J., Mydel, P., & Koziel, J. (2017). The case for periodontitis in the
44. https​://doi.org/10.1038/nri3785 pathogenesis of rheumatoid arthritis. Nature Reviews Rheumatology,
Hajishengallis, G., Darveau, R. P., & Curtis, M. A. (2012). The keystone‐ 13, 606–620. https​://doi.org/10.1038/nrrhe​um.2017.132
pathogen hypothesis. Nature Reviews Microbiology, 10, 717–725. Potempa, J., Sroka, A., Imamura, T., & Travis, J. (2003). Gingipains, the
https​://doi.org/10.1038/nrmic​ro2873 major cysteine proteinases and virulence factors of Porphyromonas
Hajishengallis, G., & Korostoff, J. M. (2017). Revisiting the Page & gingivalis: Structure, function and assembly of multidomain protein
Schroeder model: The good, the bad and the unknowns in the peri‐ complexes. Current Protein and Peptide Science, 4, 397–407.
odontal host response 40 years later. Periodontology 2000, 75, 116– Rapala‐Kozik, M., Bras, G., Karkowska‐Kuleta, J., Sroka, A., Herwald, H.,
151. https​://doi.org/10.1111/prd.12181​ Nguyen, K. A., … Kozik, A. (2011). Adsorption of components of the
Hajishengallis, G., & Lamont, R. J. (2012). Beyond the red com‐ plasma kinin‐forming system on the surface of Porphyromonas gin‐
plex and into more complexity: The polymicrobial synergy givalis involves gingipains as the major docking platforms. Infection
and dysbiosis (PSD) model of periodontal disease etiol‐ and Immunity, 79(2), 797–805. https​://doi.org/10.1128/IAI.00966-10
ogy. Molecular Oral Microbiology, 27(6), 409–419. https​://doi. Saiki, K., & Konishi, K. (2007). Identification of a Porphyromonas gin‐
org/10.1111/j.2041-1014.2012.00663.x givalis novel protein sov required for the secretion of gingipains.
Hajishengallis, G., & Lamont, R. J. (2014). Breaking bad: Manipulation of Microbiology and Immunology, 51(5), 483–491.
the host response by Porphyromonas gingivalis. European Journal of Tonetti, M. S., & Van Dyke, T. E. (2013). Periodontitis and atherosclerotic car‐
Immunology, 44, 328–338. https​://doi.org/10.1002/eji.20134​4202 diovascular disease: Consensus report of the Joint EFP/AAPWorkshop
How, K. Y., Song, K. P., & Chan, K. G. (2016) Porphyromonas gingivalis: An on Periodontitis and Systemic Diseases. Journal of Periodontology, 84,
overview of periodontopathic pathogen below the gum line. Frontiers S24–S29. https​://doi.org/10.1902/jop.2013.1340019
in Microbiology, 7, 53. https​://doi.org/10.3389/fmicb.2016.00053​ Zenobia, C., & Hajishengalliss, G. (2015). Porphyromonas gingivalis vir‐
Kadowaki, T., Takii, R., Yamatake, K., Kawakubo, T., Tsukuba, T., & ulence factors involved in subversion of leukocytes and microbial
Yamamoto, K. (2007). A role for gingipains in cellular responses and dysbiosis. Virulence, 6, 236–243. https​://doi.org/10.1080/21505​
bacterial survival in Porphyromonas gingivalis‐infected cells. Frontiers 594.2014.999567
in Bioscience, 12, 4800–4809.
Kassebaum, N. J., Bernabe, E., Dahiya, M., Bhandari, B., Murray, C. J., &
Mercenes, W. (2014). Global burden of severe periodontitis in 1990–
How to cite this article: Benedyk M, Marczyk A, Chruścicka B.
2010: A systematic review and meta‐regression. Journal of Dental
Type IX secretion system is pivotal for expression of gingipain‐
Research, 93, 1045–1053. https​://doi.org/10.1177/00220​34514​552491
Lamont, R. J., & Hajishengallis, G. (2015). Polymicrobial synergy and associated virulence of Porphyromonas gingivalis. Mol Oral
dysbiosis in inflammatory disease. Trends in Molecular Medicine, 21, Microbiol. 2019;34:237–244. https​://doi.org/10.1111/
172–183. https​://doi.org/10.1016/j.molmed.2014.11.004 omi.12268​
Lasica, A. M., Goulas, T., Mizgalska, D., Zhou, X., De Diego, I., Ksiazek,
M., … Gomis-Rüth, F. X. (2016). Structural and functional probing

You might also like