You are on page 1of 6

Dissemination of invasive Salmonella via

bacterial-induced extrusion of mucosal epithelia


Leigh A. Knodlera,1, Bruce A. Vallanceb, Jean Cellia, Seth Winfreea, Bryan Hansenc, Marinieve Monterob,
and Olivia Steele-Mortimera
a
Laboratory of Intracellular Parasites and cResearch Technologies Branch, Microscopy Unit, Rocky Mountain Laboratories, National Institute of Allergy and
Infectious Diseases, National Institutes of Health, Hamilton, MT 59840; and bDivision of Gastroenterology, Department of Paediatrics, University of British
Columbia, British Columbia Children’s Hospital, Vancouver, BC, Canada V6H 3V4

Edited by Pascale Cossart, Institut Pasteur, Paris, France, and approved August 24, 2010 (received for review May 5, 2010)

Salmonella enterica is an intracellular bacterial pathogen that resides grown on filters were used as a model polarized monolayer to ex-
and proliferates within a membrane-bound vacuole in epithelial cells amine the infectious cycle of Salmonella. Confocal microscopy
of the gut and gallbladder. Although essential to disease, how Sal- analysis of infected monolayers revealed two distinct populations of
monella escapes from its intracellular niche and spreads to secondary proliferating bacteria following the onset of replication ≥4 h post-
cells within the same host, or to a new host, is not known. Here, we infection (p.i.) (Fig. 1A and Fig. S1A). Compared with an average
demonstrate that a subpopulation of Salmonella hyperreplicating in doubling time of ≥95 min for the total population (Fig. S1A), some
the cytosol of epithelial cells serves as a reservoir for dissemination. bacteria were replicating at a much faster rate, with a doubling time
These bacteria are transcriptionally distinct from intravacuolar Sal- of ∼20 min (Movie S1). There was a temporal increase in the in-
monella. They are induced for the invasion-associated type III secre- cidence of these “hyperreplicating” Salmonella (defined as >50
tion system and possess flagella; hence, they are primed for invasion. bacteria per cell) (Fig. 1 A and B). By 10 h p.i., 11 ± 4.2% of infected
Epithelial cells laden with these cytosolic bacteria are extruded out of cells contained hyperreplicating bacteria (Fig. 1A). A similar phe-
the monolayer, releasing invasion-primed and -competent Salmo- notype was previously described for a Salmonella sifA mutant,
nella into the lumen. This extrusion mechanism is morphologically which hyperreplicates in the host cell cytosol because of a defect in
similar to the process of cell shedding required for turnover of the maintaining vacuolar integrity (7). We therefore assessed whether
intestinal epithelium. In contrast to the homeostatic mechanism, the hyperreplicating WT Salmonella we observed in polarized epi-
however, bacterial-induced extrusion is accompanied by an inflam- thelial cells are also free in the cytosol. Confocal microscopy in-
matory cell death characterized by caspase-1 activation and the apical dicated that many of these bacteria were not in a lysosome-
release of IL-18, an important cytokine regulator of gut inflammation. associated membrane protein 1 (LAMP1)-positive compartment
Although epithelial extrusion is obviously beneficial to Salmonella (Fig. 1B and Fig. S1B), and thus not in a mature SCV (8). Selective
for completion of its life cycle, it also provides a mechanistic explana- membrane permeabilization followed by immunostaining with
Downloaded from https://www.pnas.org by 103.252.33.114 on March 12, 2022 from IP address 103.252.33.114.

tion for the mucosal inflammation that is triggered during Salmonella polyclonal anti-Salmonella LPS antibody revealed that at least one-
infection of the gastrointestinal and biliary tracts. third of the hyperreplicating bacteria are cytosolic (Fig. 1C and Fig.
S2). These experiments demonstrate that WT Salmonella can rep-
caspase-1 | epithelial cells | flagella | IL-18 | type III secretion licate in a vacuole and the cytosol in epithelial cells, but they pro-
liferate more efficiently in the cytosolic environment (7, 9, 10).

I ntracellular pathogens reside either within a membrane-bound


vacuole or freely within the host cell cytosol. However, regardless
of their specific lifestyle, three distinct steps are common to their
Cytosolic Salmonella Are Invasion-Primed. We hypothesized that the
two distinct intracellular environments, intravacuolar and cyto-
infectious cycle: the ability to enter host cells, their establishment of solic, would differentially influence the expression of bacterial
an intracellular niche, and their escape from the host cell (1). Entry virulence genes. To assess this, we used a plasmid-derived tran-
and intracellular survival are critical virulence stages for these scriptional fusion assay based on destabilized GFP(LVA). Pro-
pathogens, but exit from the infected cell is essential for dissemi- moters were selected from well-characterized genes in each of the
nation and transmission to other hosts. Although considerable three type III secretion systems (T3SSs): PfliC-gfp[LVA] (flagel-
progress has been made in elucidating the first two facets of this lar T3SS), PprgH-gfp[LVA] (T3SS1), and PssaG-gfp[LVA]
cycle, the mechanism by which intracellular pathogens escape from (T3SS2) (11). The number of fluorescent bacteria was monitored
host cells has been comparably neglected. with time. Under the infection conditions used here, efficient
The Gram-negative bacterium Salmonella enterica causes a wide invasion requires both T3SS1 and flagellar-based motility (11–13)
range of food- and water-borne diseases ranging from self-limiting (Fig. S1A). This single-cell assay confirmed that both T3SS1
gastroenteritis to systemic typhoid fever in both humans and ani- (PprgH-gfp[LVA]) and flagella (PfliC-gfp[LVA]) were rapidly
mals. In enteric infections, Salmonella preferentially targets the down-regulated after bacterial internalization (11, 14, 15) (Fig.
single layer of polarized columnar epithelial cells lining the surface S1C). Surprisingly, these virulence factors were not completely
of the gastrointestinal tract (2–4), triggering an extensive in- inactivated (Fig. 2 and Fig. S1C). At 10 h p.i., ∼6% of infected
MICROBIOLOGY

flammatory response. After invading epithelial cells from the apical cells contained fluorescent PprgH-GFP[LVA] bacteria (3.8 ±
side, Salmonella resides and replicates within a membrane-bound 1.2% of the total bacterial population) (Fig. 2 and S1C). Strik-
vacuole, known as the Salmonella-containing vacuole (SCV).
Symptomatic and asymptomatic infections are characterized by
the fecal shedding of bacteria (5, 6), suggesting that Salmonella Author contributions: L.A.K., B.A.V., J.C., S.W., and O.S.-M. designed research; L.A.K., B.A.V.,
escapes from its intracellular niche back into the gut lumen as part J.C., S.W., B.H., and M.M. performed research; L.A.K., B.A.V., J.C., and S.W. analyzed data;
of its infectious cycle. Here, we report that Salmonella exits from and L.A.K. and O.S.-M. wrote the paper.

polarized epithelia by coopting a mechanism normally used by the The authors declare no conflict of interest.
host to remove senescent cells from the mucosal epithelium. This article is a PNAS Direct Submission.
Freely available online through the PNAS open access option.
Results and Discussion 1
To whom correspondence should be addressed. E-mail: lknodler@niaid.nih.gov.
WT Salmonella Hyperreplicates in the Cytosol of Epithelial Cells. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.
Human colonic epithelial cells (C2BBe1, a subclone of Caco-2) 1073/pnas.1006098107/-/DCSupplemental.

www.pnas.org/cgi/doi/10.1073/pnas.1006098107 PNAS | October 12, 2010 | vol. 107 | no. 41 | 17733–17738


Fig. 1. Infected epithelial cells contain two distinct populations of replicating Salmonella. (A) Polarized C2BBe1 monolayers were infected with WT Sal-
monella constitutively expressing mCherry. Monolayers were fixed at the indicated times and immunostained for the tight junction marker ZO-1 and the late
endosomal/lysosomal marker LAMP1. The percentage of infected cells containing >50 bacteria per cell was scored by fluorescence microscopy (mean ± SD, n ≥
3 independent experiments). (B) Confocal image showing the two different populations of replicating Salmonella at 8 h p.i. Many hyperreplicating bacteria
are not in a LAMP1-positive vacuole. The overlay shows mCherry Salmonella in green, LAMP1 in red, and ZO-1 in blue. (Scale bar, 10 μm; Inset scale bar, 2 μm.)
(C) Confocal image showing that some hyperreplicating bacteria are accessible to anti-LPS antibody delivered to the cytosol. At 8 h p.i., the plasma membrane
of infected monolayers was selectively permeabilized with digitonin. Cells were then incubated with anti-Salmonella LPS and anti-GM130 antibodies to detect
cytosolic bacteria and the cytosolic face of the Golgi, respectively. Nuclei were stained with Hoechst 33342. The overlay shows mCherry Salmonella in red,
cytosolic LPS in green, and nuclei in blue. GM130 is shown in the gray scale. Asterisks in the overlay indicate digitonin-permeabilized cells. (Scale bar, 10 μm;
GM130 scale bar, 10 μm; Inset scale bar, 2 μm.)

ingly, these T3SS1-induced bacteria were almost exclusively 8 h p.i., ∼60% of the T3SS1-induced bacteria were cytosolic and
found in cells containing hyperreplicating bacteria (Fig. 2A) and 23 ± 6% were in LAMP1-positive SCVs (Fig. 2 B, C, and E). By
associated with flagella (Fig. 2D). As expected, T3SS2 was in- contrast, the T3SS2-induced bacteria were intravacuolar (91 ±
duced intracellularly (16) (Fig. S1C); fluorescent PssaG-GFP 6% LAMP1-positive) and typically found in cells containing 5–20
[LVA] bacteria were not detected until >2 h p.i., and 32 ± 6.6% of bacteria (Fig. 2E and Fig. S3). Using live cell imaging, the motility
the bacteria were GFP-positive by 10 h p.i. (Figs. S1C and S3). At of intracellular Salmonella was assessed at 8 h p.i. (Fig. 2F).
Downloaded from https://www.pnas.org by 103.252.33.114 on March 12, 2022 from IP address 103.252.33.114.

Fig. 2. Flagella and the invasion-associated T3SS1 are expressed by cytosolic bacteria late during infection. (A–D) Polarized C2BBe1 monolayers were infected
with WT Salmonella carrying a plasmid that expresses destabilized GFP [GFP(LVA)] under the control of the T3SS1-associated prgH promoter. At 8 h p.i.,
monolayers were fixed and immunostained for confocal microscopy. (A) T3SS1 is induced late during infection in “hyperreplicating” bacteria. PprgH-induced
bacteria (green in overlay), Salmonella LPS (red), and the tight junction marker ZO-1 (blue) are shown. (Scale bars, 10 μm.) (B) T3SS1-induced bacteria are not in
a mature LAMP1-positive SCV. PprgH-induced bacteria (green in overlay), LAMP1 (red), and ZO-1 (blue) are shown. (Scale bar, 10 μm; Inset scale bar, 2 μm.) (C)
T3SS1-induced bacteria are cytosolic. The plasma membrane of polarized cells was selectively permeabilized with digitonin before the cytosolic delivery of anti-
LPS and anti-GM130 (permeabilization control) antibodies. PprgH-induced bacteria (green in overlay), cytosolic bacteria (LPS, red), and Hoechst 33342 (blue) are
shown. Asterisks indicate cells permeabilized by digitonin. GM130 is shown in the gray scale. (Scale bar, 10 μm; GM130 scale bar, 10 μm; Inset scale bar, 2 μm.) (D)
T3SS1-induced bacteria are flagellated. PprgH-induced bacteria (green in overlay), FliC (red), and ZO-1 (blue) are shown. (Scale bar, 10 μm; Inset scale bar, 2 μm.)
(E) T3SS1-induced bacteria are cytosolic, whereas T3SS2-induced bacteria are vacuolar. Infected cells were fixed at 8 h p.i. and immunostained for LAMP1 or
selectively permeabilized with digitonin and incubated with anti-LPS antibodies to detect cytosolic bacteria. The number of T3SS1-induced (PprgH-GFP[LVA])
and T3SS2-induced (PssaG-GFP[LVA]) bacteria positive for LAMP1 or cytosolic LPS staining was scored by fluorescence microscopy (mean ± SD, n ≥ 3 independent
experiments.) (F) Some T3SS1-induced bacteria are motile. The motility of T3SS1-induced (PprgH-GFP[LVA]) and T3SS2-induced (PssaG-GFP[LVA]) bacteria was
assessed at 8–9 h p.i. by live cell imaging. (Inset) Average instantaneous velocities (μm/s) for motile PprgH-induced bacteria (n = 3 independent experiments).

17734 | www.pnas.org/cgi/doi/10.1073/pnas.1006098107 Knodler et al.


Consistent with flagellin (FliC) expression (Fig. 2D), a subset of infected epithelial cells occurs in vivo, we used a mouse model in
T3SS1-induced bacteria was motile (Fig. 2F and Movie S2). This which Salmonella rapidly breaches the intestinal barrier and
population moved at speeds consistent with flagellar-based mo- spreads systemically to various tissues, including the mucosal
tility (4–15 μm/s) (17), whereas T3SS2-induced bacteria were epithelium lining the gallbladder. In this murine infection model,
immobile (Fig. 2F and Movie S2). Hence, there are at least two Salmonella causes inflammation similar to cholecystitis seen in
transcriptionally distinct intracellular populations of replicating humans during acute typhoid fever (18). We observed both free
bacteria in epithelial cells: T3SS2-induced intravacuolar bacteria bacteria and bacteria-laden epithelial cells in the gallbladder lu-
and T3SS1-induced flagellated bacteria that are cytosolic. men (Fig. 3 D and E). In agreement with our observations,
sloughing of enterocytes laden with Salmonella from villus tips has
Invasion-Primed Salmonella Are Released into the Lumen by also been reported in rabbit ileal loop studies (2). Therefore,
Extruding Cells. Coincident with the onset and kinetics of hyper- extrusion of Salmonella-infected epithelial cells is evident in both
enteric and systemic infections.
replication, bacteria-laden cells extruding toward the apical side
Salmonella-associated extrusion resembles cell extrusion in-
were observed by EM (Fig. 3 A and B). Bacteria in these extruded
volved in the rapid turnover of polarized epithelial cells in the gut.
cells were not surrounded by a vacuolar membrane [Fig. 3B (ii)], This process occurs when neighboring cells contract to push
in agreement with the confocal microscopy data for hyper- a dying cell out of the monolayer and is characterized by re-
replicating bacteria (Figs. 1C and 2E). By contrast, SCV mem- organization of adherens and tight junctions to maintain the in-
branes were readily apparent around replicating bacteria in cells tegrity of the epithelial monolayer (19–22). Here, we observed
within the monolayer (Fig. 3C). To examine whether extrusion of similar actin contractile rings and tight junction “rosettes” at the
base of extruding Salmonella-infected cells (Fig. S4 and Movie
S3), suggesting that the Salmonella-associated and homeostatic
extrusion events occur via similar cell biological processes.
The incidence of extrusion was significantly increased on in-
fection: 10 ± 2.7% of cells containing Salmonella showed evidence
of extrusion at 10 h p.i. compared with only 0.85 ± 0.89% of un-
infected cells (Fig. 4A). Furthermore, cells containing T3SS1-
induced Salmonella were more likely to be extruded than those
containing T3SS2-induced bacteria (20 ± 4.7% vs. 2.4 ± 0.9%;
Fig. 4 A and B). In infected gallbladders, T3SS1-induced bacteria
were found within epithelial cells lining the gallbladder at 4 d p.i.
and did not immunostain for FliC (Fig. S5). By 5 d p.i., flagellated
T3SS1-induced bacteria were predominantly found within cells
that had been sloughed into the lumen or were free in the lumen
Downloaded from https://www.pnas.org by 103.252.33.114 on March 12, 2022 from IP address 103.252.33.114.

(Fig. 4C and Fig. S5). Altogether, these data imply a strong cor-
relation between T3SS1 induction and Salmonella-induced ex-
trusion in vitro and in vivo. To address the invasion competence
of these bacteria, we developed a secondary infection assay
(Fig. S6A). Consistent with the onset of cell extrusion from the
polarized monolayer, significant numbers of bacteria could be
recovered from a naive population of epithelial cells from 6 h p.i.
and increasing thereafter. This demonstrates that Salmonella re-
leased from extruded cells are invasion-primed and -competent.

Extruding Cells Undergo Inflammatory Cell Death. During gut ho-


meostasis, epithelial cells are shed into the lumen as a result of
anoikis, a form of apoptosis characterized by activated caspase-2,
-3, and -9 but not caspase-1 or -8 (23–26). Because the Salmonella-
laden extruding cells also showed structural features typical of cell
death both in vivo and in vitro (Fig. 3 B, D, and E), we next assayed
Fig. 3. Apical extrusion of infected cells from mucosal epithelium in vitro and for caspase activity. Epithelial monolayers were incubated with
in vivo. (A) SEM of the apical surface of an infected monolayer. Polarized cell-permeable fluorescent probes that bind irreversibly to acti-
C2BBe1 cells were infected with WT Salmonella, and at 10 h p.i., samples were vated caspases. Uninfected extruding cells were positive for active
fixed and processed for SEM. (i) Extruding cell is bacteria-laden. Microvilli on caspase-3/-7 (74 ± 8.1%) but not for active caspase-1 (21 ± 9.4%),
the apical surface of the underlying monolayer are evident. (Scale bar, 2 μm.)
as previously described (27) (Fig. 5B). By contrast, the majority of
(ii) Inset from i. (Scale bar, 0.5 μm.) (B) Transmission EM (TEM) of an extruded
infected extruding cells were positive for both active caspase-1
MICROBIOLOGY

cell. Cells were infected as for A, and samples were fixed and processed for
TEM. (i) Extruded cell is filled with bacteria and is adjacent to the apical surface (83 ± 4.5%) and caspase-3/-7 (85 ± 7.3%) (Fig. 5 A and B). Ho-
of the monolayer, distinguished by microvilli (MV). (Scale bar, 2 μm.) (ii) Inset meostatic extrusion of epithelial cells is not prevented by general
from i showing the absence of a vacuolar membrane around Salmonella in caspase inhibition (21). Addition of a caspase-1 inhibitor did not
extruded cells. N, epithelial cell nucleus. (Scale bar, 0.5 μm.) (C) TEM of an block, but significantly decreased, extrusion of infected cells
infected C2BBe1 cell that was not extruding. Cells were infected as for A, and (Fig. 4A), suggesting that the signal for homeostatic and bacterial-
samples were fixed and processed for TEM. Salmonella are clearly surrounded induced extrusion precedes caspase activation.
by a vacuolar membrane (arrowheads). (Scale bar, 0.5 μm.) (D and E) EM of Caspase-1–dependent programmed cell death, also known as
infected gallbladders. Gallbladders from C57BL/6 mice were collected 6 d p.i.
pyroptosis, is characterized by pore formation in the plasma
and processed for TEM. Apical extrusion of Salmonella-infected epithelial cells
into the lumen (L) is evident. Some bacteria are also free in the lumen. (Scale
membrane, followed by cell swelling and lysis, and the proteolytic
bars, 2 μm.) (E) Infected cell is undergoing extrusion into the luminal space (L), processing of proinflammatory cytokines, leading to the secretion
with an obvious constriction site formed by the junctional complexes of of mature active IL-1β and IL-18 (28). To monitor plasma
neighboring cells (arrowheads). Microvilli (MV) on the apical surface of the membrane integrity, infected cells were incubated with SYTOX
mucosal epithelium are indicated. Orange nucleic acid dye, which can only enter cells with a com-

Knodler et al. PNAS | October 12, 2010 | vol. 107 | no. 41 | 17735
Fig. 4. Extruding epithelial cells contain invasion-primed Salmonella. (A) Quantification of extrusion. Polarized C2BBe1 monolayers were infected with
Salmonella constitutively expressing mCherry or carrying destabilized GFP reporters for T3SS1 (PprgH-GFP[LVA]) or T3SS2 (PssaG-GFP[LVA]). At 10 h p.i.,
monolayers were fixed and immunostained for ZO-1. DNA was stained with Hoechst 33342. Where indicated (+YVAD), 100 μM Ac-YVAD-CMK was added
before, and maintained throughout, the infection. Infected (mCherry bacteria) cells, or cells containing PprgH-positive or PssaG-positive bacteria, showing
signs of extrusion were scored by fluorescence microscopy (mean ± SD, n ≥ 4 independent experiments). Asterisks indicate significantly different from
infected cells (P < 0.05, ANOVA with Dunnett’s post hoc test). (B) Confocal image showing an extruding cell containing T3SS1-induced bacteria. Polarized
C2BBe1 monolayers infected with WT Salmonella carrying PprgH-GFP[LVA] were fixed and immunostained at 10 h p.i. DNA was stained with Hoechst 33342.
PprgH-induced bacteria (green in overlay), apical plasma membrane marker, villin (red), DNA (cyan), and tight junction marker ZO-3 (gray) are shown. The x–z
section is indicated by a dashed line. (Scale bar, 10 μm.) (C) Fluorescence microscopy image showing that epithelial cells containing T3SS1-induced, flagellated
bacteria are extruded into the gallbladder lumen (L). C57BL/6 mice were infected with Salmonella carrying a plasmid that expresses destabilized GFP under
the control of the T3SS1-associated invF promoter (PinvF-GFP[LVA]). Mice were killed 5–6 d p.i., and gallbladders processed for immunostaining. DNA was
stained with DAPI. PinvF-induced bacteria (green in overlay), FliC (red), and DNA (cyan) are shown. The arrowhead indicates an extruded cell containing
numerous T3SS1-induced, flagellated bacteria. (Scale bar, 10 μm; Inset scale bar, 2 μm.)

promised plasma membrane, and Hoechst 33342, a cell-perme- extrusion, releasing the invasion-primed Salmonella into the lu-
able nuclear stain. Salmonella-infected extruding cells were pos- men of the gastrointestinal and biliary tracts. Escape into the lu-
itive for both dyes, indicating plasma membrane rupture, whereas men allows Salmonella to infect secondary cells rapidly, and may
neighboring cells within the monolayer stained only with Hoechst also contribute to host-to-host transmission. Thus, by subverting
a host-dependent cell turnover event, Salmonella completes its
Downloaded from https://www.pnas.org by 103.252.33.114 on March 12, 2022 from IP address 103.252.33.114.

33342 (Fig. 5C). We next quantified the apical and basolateral


release of cytokines from infected C2BBe1 cells. Although IL-1β infectious cycle (Fig. S6B). Given the prevalence of mucosal-
release was below the limits of detection (0.8 pg/mL), we observed dwelling pathogens, other pathogens may also use this host cell
a steady temporal increase in the release of IL-18, which was re- process as an exit strategy.
stricted to the apical side of monolayers (Fig. 5D) and dependent
on caspase-1 and -3 (Fig. 5E). Collectively, these data highlight Materials and Methods
a clear difference in the activated cell death program between Bacterial Strains and Plasmids. WT S. enterica serovar Typhimurium
Salmonella-induced and homeostatic extrusion. (S. Typhimurium) SL1344 (37) and ΔSPI2::kan (38), ΔSPI1::kan (15), and flgB::
We have demonstrated that extruding Salmonella-infected cells Tn10 mutants (11) have been described previously. For constitutive expression of
GFP or mCherry, WT S. Typhimurium was electroporated with pFPV25.1 (39) or
undergo inflammatory cell death. A complex but still largely un-
pFPV-mCherry (40), respectively. S. Typhimurium carrying the destabilized GFP
explained pathological feature of salmonelloses is an overwhelming (GFP[LVA]) (41) reporter plasmids pMPMA3ΔPlac-PprgH-gfp[LVA], pMPMA3Δ-
inflammatory response. We propose that bacterial-induced ex- Plac-PinvF-gfp[LVA], pMPMA3ΔPlac-PfliC-gfp[LVA], or pMPMA3ΔPlac-PssaG-
trusion provides one mechanistic explanation for the pathogenesis gfp[LVA] (11) were used to analyze intracellular virulence gene expression.
of mucosal inflammation during Salmonella infections of the in-
testine and gallbladder. In support of our data, activation of IL-18 in Infection of Cultured Epithelial Cells with Salmonella. Cell lines were obtained
porcine intestinal mucosa has been reported for S. enterica serovar from the American Type Culture Collection and used at a passage number ≤13
Choleraesuis infections (29). Interestingly, in inflammatory bowel for all experiments. C2BBe1 human colorectal adenocarcinoma cells (CRL-
diseases, chronic inflammation is also associated with elevated 2012), a clone of Caco-2, were maintained in DMEM (Mediatech) containing
10 μg/mL human transferrin (Sigma) and 10% (vol/vol) heat-inactivated FCS
IL-18 levels (30–32). Therefore, caspase-1–dependent IL-18 pro-
(Gibco). HeLa human cervical adenocarcinoma cells were maintained in
duction by intestinal epithelial cells might prove to be a mediator
Eagle’s modified Eagle medium (Mediatech) containing 10% (vol/vol) heat-
of mucosal inflammation associated with both autoimmune dis- inactivated FCS. Polarized monolayers were established by seeding 2 × 105
orders (33, 34) and bacterial infections. C2BBe1 cells in basal seeding medium containing MITO+ serum extender
Conceptually, an increased turnover of mucosal epithelium (Becton Dickinson) on BIOCOAT fibrillar collagen 24-well inserts with a 1-μm
provides the host with an ideal defense mechanism against in- membrane pore size (Becton Dickinson). After 24 h, the seeding medium was
fection. Indeed, it has been implicated as a protective mechanism replaced with enterocyte differentiation medium containing MITO+ serum
in the gut against both bacteria and parasites (35, 36). However, we extender. Cells were incubated in differentiation medium for a total of 3 d,
believe that Salmonella takes advantage of this process as a unique replaced with fresh medium each day, until the transepithelial electrical re-
means of bacterial egress. Critical to this is the finding that sistance was ≥250 Ω.cm2, as measured using a Millicell Electrical Resistance
System (Millipore). The medium was changed to DMEM containing 10% (vol/
a population of Salmonella is cytosolic and expresses the virulence
vol) heat-inactivated FCS (growth medium, GM) before infection.
genes required for invasion. We hypothesize that a vacuole mat-
Preparation of invasive Salmonella and infection of HeLa cells at a multi-
uration defect leads to the cytosolic release of a small, but signif- plicity of infection (MOI) of ∼50 were as previously described (42). Polarized
icant, fraction of bacteria. The nutrient-rich cytosol supports monolayers were infected apically at an MOI of ∼50–100 for 10 min and then
a high bacterial replication rate and reprograms virulence gene washed three times apically and twice basolaterally in HBSS (Mediatech). Cells
expression toward invasion. The cytosolic load of bacteria is were incubated in antibiotic-free GM until 30 min p.i. Thereafter, GM con-
sensed by the host cell, leading to inflammatory cell death and taining 50 μg/mL gentamicin (Sigma) was added for 1 h to kill any remaining

17736 | www.pnas.org/cgi/doi/10.1073/pnas.1006098107 Knodler et al.


two transwells per well). Coincubation of C2BBe1 and HeLa cells continued
in GM containing 10 μg/mL gentamicin. Polarized monolayers were solubi-
lized in TX-100/SDS as described above. For quantification of the secondary
infection, the HeLa cells were washed extensively in HBSS, solubilized in 1 mL
of TX-100/SDS, and plated on LB agar plates.

Immunofluorescence Staining. Infected monolayers were washed twice, api-


cally and basolaterally, with PBS and then fixed apically and basolaterally with
3.5% (wt/vol) paraformaldehyde (PFA) for 20 min at room temperature (RT).
Monolayers were then washed apically and basolaterally with PBS, followed
by incubation in 75 mM ammonium chloride/20 mM glycine in PBS for 10 min
at RT to quench free aldehyde groups. After washing in PBS, monolayers
were permeabilized in 10% (vol/vol) normal goat serum/0.1% (wt/vol) sa-
ponin in PBS (SS-PBS) for 20 min at RT. Primary and secondary antibodies
were diluted in SS-PBS and applied apically and basolaterally in a humid box
for 1 h at RT. Filters were excised from transwell supports and placed cell side
up on a drop of Prolong Gold antifade reagent (Invitrogen) on a glass slide.
Another drop of mounting media was applied to the filter, and a coverslip
was placed directly on top. Samples were cured overnight at RT.
Live cell staining was carried out by incubation for 10 min at 37 °C with 2 μg/
mL Hoechst 33342 (Invitrogen) to stain nucleic acids or 0.5 μM SYTOX Orange
(Invitrogen) to stain nucleic acids in cells with a compromised plasma mem-
brane. For determination of activated caspase-1 and caspase-3/-7, live cells
were incubated with FAM-YVAD-FMK or FAM-DEVD-FMK, respectively, in
GM for 1 h according to the manufacturer’s instruction (Immunochemistry
Technologies) before fixation.
To determine whether intracellular bacteria were vacuolar or cytosolic,
polarized C2BBe1 cells were infected as described, and at 8 h p.i., the apical
plasma membrane was selectively permeabilized using digitonin to allow
access of antibody to the cytosol. Briefly, transwells were washed three times
in KHM buffer [110 mM potassium acetate, 20 mM Hepes, 2 mM MgCl2 (pH
Fig. 5. Salmonella-infected extruding epithelial cells undergo inflammatory 7.3)] and incubated apically with 150 μg/mL digitonin (Sigma) in KHM buffer
cell death. (A) Confocal image of an infected extruding cell positive for active
for 90 s at RT. Transwells were then immediately washed with KHM buffer.
caspase-1. Polarized C2BBe1 monolayers were infected with WT Salmonella
Rabbit polyclonal anti-Salmonella LPS antibody and mouse anti-human
constitutively expressing mCherry (red in overlay). At 9 h p.i., live cells were GM130 monoclonal antibody were added for 15 min at 37 °C to label cy-
incubated with the active caspase-1 probe FAM-YVAD-FMK (green). Nuclei tosolic bacteria and the cytosolic face of the Golgi, respectively. Monolayers
Downloaded from https://www.pnas.org by 103.252.33.114 on March 12, 2022 from IP address 103.252.33.114.

were stained with Hoechst 33342 (cyan). (Upper) 3D-rendered view in the x–z
were then fixed and quenched as described above, followed by nonselective
plane. (Scale bar, 10 μm.) (B) C2BBe1 cells were infected as in A and incubated
permeabilization in 0.1% (wt/vol) saponin and 10% (vol/vol) horse serum in
with FAM-YVAD-FMK (active caspase-1) or FAM-DEVD-FMK (active caspase-3
PBS for 15 min at RT. Anti-Salmonella LPS antibody (1:200; Difco) and anti-
and -7) at 9 h p.i. Uninfected or infected extruding cells positive for active cas-
GM130 monoclonal antibody (1:50; BD Transduction Laboratories) were
pases were scored by fluorescence microscopy (mean ± SD, n ≥ 3 independent
detected using Alexa Fluor- and Cy5-conjugated secondary antibodies as
experiments). (C) Confocal image of an extruding cell labeled by a membrane
described above. Nucleic acids were subsequently stained with Hoechst
impermeant nucleic acid dye. Polarized monolayers were infected with WT
33342 (2 μg/mL) for 10 min at RT. Intracellular bacteria were then scored for
Salmonella constitutively expressing GFP (green in overlay). At 10 h p.i., live cells
LPS staining. Extruding cells were excluded from analysis because they have
were incubated with two nucleic acid dyes, the membrane impermeant SYTOX
a compromised plasma membrane and delivery of antibodies could occur in
Orange (red in overlay) and the membrane permeant Hoechst 33342 (cyan). The
a digitonin-independent manner. For each experiment, one transwell was
extruding cell contains numerous Salmonella and is positive for both nucleic acid
used as a permeabilization control to ensure that the apical plasma mem-
stains, indicating a compromised plasma membrane. The x–z section is indicated
brane (but not the endomembranes) was permeabilized. For this, the digi-
by a dashed line. (Scale bar, 10 μm.) (D) Apical release of IL-18 from infected
tonin-treated cells were incubated with two LAMP1 antibodies: rabbit
epithelial cells. Polarized monolayers were mock-infected (○) or infected with
polyclonal antibody directed against the cytoplasmic tail of LAMP1 (1:250;
Salmonella (●). Apical supernatants were collected and assayed for IL-18 con-
Novus Biologicals) and a mouse monoclonal antibody directed against the
centration by sandwich ELISA (mean ± SD, n ≥ 3 independent experiments). (E)
luminal portion of LAMP1 (1:1,000; clone H4A3, Developmental Studies
Caspase-1 and caspase-3 activation are required for IL-18 release. Polarized
Hybridoma Bank) (Fig. S2). We excluded any experiments in which cells
monolayers were mock-infected or infected with WT Salmonella. Where in-
stained with both anti-LAMP1 antibodies.
dicated, monolayers were pretreated and incubated for the entire infection
Immunostaining of mouse gallbladder tissues was performed using pre-
with the general caspase inhibitor Z-VAD-FMK (50 μM), the caspase inhibitor
viously described procedures (44). Gallbladders were fixed in 4% (wt/vol) PFA
negative control Z-FA-FMK (50 μM), the caspase-1 inhibitor Ac-YVAD-CMK (100
for 1 h at RT, washed in PBS, embedded in optimal cutting template com-
μM), or the caspase-3 inhibitor, Ac-DEVD-CMK (50 μM). At 10 h p.i., apical
pound (Sakura Finetek), and then frozen with isopentane and liquid N2 and
supernatants were collected and IL-18 was assayed by ELISA (mean ± SD, n ≥ 3
stored at −70 °C. Serial sections were cut at a thickness of 4 μm for immu-
independent experiments). Asterisks indicate data significantly different from
nohistochemical staining with Alexa Fluor 488-conjugated rabbit anti-GFP
WT Salmonella infection (P < 0.05, ANOVA with Dunnett’s post hoc test).
MICROBIOLOGY

antibody (1:500; Invitrogen) and mouse monoclonal antibody anti-FliC


(1:100; BioLegend). Nucleic acids were stained with DAPI.
extracellular bacteria, followed by GM containing 10 μg/mL gentamicin for the
remainder of the experiment to restrict the extracellular growth of bacteria. Animal Infections. WT S. Typhimurium (Fig. 3 D and E) or S. Typhimurium
carrying pMPMA3ΔPlac-PinvF-gfp[LVA] (Fig. 4C) were grown overnight at
Enumeration of Intracellular and Extracellular Bacteria. For quantification of 37 °C with shaking in LB or LB containing 50 μg/mL carbenicillin, respectively.
viable intracellular bacteria, polarized monolayers were washed apically and Cultures were diluted in PBS to ∼5–8 × 103 cfu/mL. Female C57BL/6 mice
basolaterally three times with PBS and then lysed in 1 mL of 1% (vol/vol) Triton (Charles River) were infected with 100 μL of the diluted culture by tail vein
X-100/0.1% (wt/vol) SDS (TX-100/SDS). Serial dilutions were plated on LB agar injection. Additionally, for infections with plasmid-bearing Salmonella, mice
plates. Bacterial doubling time was calculated as described previously (43). were treated with carbenicillin (100 mg/kg) by i.p. injection daily throughout
To measure the invasion competence of apically released bacteria, we the infection to maintain the plasmid. The protocols used were in direct
developed a secondary infection protocol. Polarized C2BBe1 monolayers accordance with guidelines drafted by the University of British Columbia’s
were infected and treated with 50 μg/mL gentamicin to kill extracellular Animal Care Committee and the Canadian Council on the Use of Laboratory
bacteria as described above. At 1.5 h p.i., transwells were inverted onto Animals. Gallbladders were collected 4–6 d p.i. and processed for EM or
a monolayer of HeLa cells seeded in six-well plates (2 × 105 cells per well and immunohistochemistry as described above.

Knodler et al. PNAS | October 12, 2010 | vol. 107 | no. 41 | 17737
Information on reagents, quantification of cytokine release, fluorescence oratory for discussion and critique of this manuscript. This research was sup-
microscopy, determination of bacterial velocities, and EM is provided in SI ported by the Intramural Research Program of the National Institute of
Allergy and Infectious Diseases, National Institutes of Health (O.S.-M. and
Materials and Methods.
J.C.) and by grants from the Canadian Institutes of Health Research and the
Crohn’s and Colitis Foundation (to B.A.V.). M.M. was supported by the Cana-
ACKNOWLEDGMENTS. We thank Caixia Ma and Tina Huang for their expert dian Institute of Gastroenterology/Crohn’s and Colitis Foundation of Canada/
technical assistance; Anita Mora for graphics assistance; the Genomics Core Canadian Institutes of Health Research Fellowship. B.A.V. is the Canada Re-
Facility at Rocky Mountain Laboratories for DNA sequence analysis; and Rey search Chair in Pediatric Gastroenterology and the Children with Intestinal
Carabeo, Ed Miao, Staffan Svärd, and members of the Steele–Mortimer lab- and Liver Disorders (CHILD) Foundation Research Scholar.

1. Hybiske K, Stephens RS (2008) Exit strategies of intracellular pathogens. Nat Rev 22. Baron DA, Miller DH (1990) Extrusion of colonic epithelial cells in vitro. J Electron
Microbiol 6:99–110. Microsc Tech 16:15–24.
2. Wallis TS, et al. (1986) The nature and role of mucosal damage in relation to Salmonella 23. Papaconstantinou HT, et al. (2000) Prevention of mucosal atrophy: Role of glutamine
typhimurium-induced fluid secretion in the rabbit ileum. J Med Microbiol 22:39–49. and caspases in apoptosis in intestinal epithelial cells. J Gastrointest Surg 4:416–423.
3. Kent TH, Formal SB, Labrec EH (1966) Salmonella gastroenteritis in rhesus monkeys. 24. Grossmann J, et al. (2002) Induction of apoptosis before shedding of human intestinal
Arch Pathol 82:272–279. epithelial cells. Am J Gastroenterol 97:1421–1428.
4. Santos RL, et al. (2001) Animal models of Salmonella infections: Enteritis versus 25. Fouquet S, et al. (2004) Early loss of E-cadherin from cell-cell contacts is involved in the
typhoid fever. Microbes Infect 3:1335–1344. onset of Anoikis in enterocytes. J Biol Chem 279:43061–43069.
5. Vogelsang TM, Bøe J (1948) Temporary and chronic carriers of Salmonella typhi and 26. Bullen TF, et al. (2006) Characterization of epithelial cell shedding from human small
Salmonella paratyphi B. J Hyg (Lond) 46:252–261. intestine. Lab Invest 86:1052–1063.
6. Buchwald DS, Blaser MJ (1984) A review of human salmonellosis: II. Duration of 27. Slattum G, McGee KM, Rosenblatt J (2009) P115 RhoGEF and microtubules decide the
excretion following infection with nontyphi Salmonella. Rev Infect Dis 6:345–356.
direction apoptotic cells extrude from an epithelium. J Cell Biol 186:693–702.
7. Beuzón CR, Salcedo SP, Holden DW (2002) Growth and killing of a Salmonella enterica
28. Bergsbaken T, Fink SL, Cookson BT (2009) Pyroptosis: Host cell death and
serovar Typhimurium sifA mutant strain in the cytosol of different host cell lines.
inflammation. Nat Rev Microbiol 7:99–109.
Microbiology 148:2705–2715.
29. Foss DL, Zilliox MJ, Murtaugh MP (2001) Bacterially induced activation of interleukin-
8. Drecktrah D, Knodler LA, Howe D, Steele-Mortimer O (2007) Salmonella trafficking is
18 in porcine intestinal mucosa. Vet Immunol Immunopathol 78:263–277.
defined by continuous dynamic interactions with the endolysosomal system. Traffic 8:
30. Monteleone G, et al. (1999) Bioactive IL-18 expression is up-regulated in Crohn’s
212–225.
disease. J Immunol 163:143–147.
9. Beuzón CR, et al. (2000) Salmonella maintains the integrity of its intracellular vacuole
31. Salvati VM, et al. (2002) Interleukin 18 and associated markers of T helper cell type 1
through the action of SifA. EMBO J 19:3235–3249.
10. Paz I, et al. (2010) Galectin-3, a marker for vacuole lysis by invasive pathogens. Cell activity in coeliac disease. Gut 50:186–190.
Microbiol 12:530–544. 32. Wiercinska-Drapalo A, Flisiak R, Jaroszewicz J, Prokopowicz D (2005) Plasma
11. Ibarra JA, et al. (2010) Induction of Salmonella pathogenicity island 1 under different interleukin-18 reflects severity of ulcerative colitis. World J Gastroenterol 11:605–608.
growth conditions can affect Salmonella-host cell interactions in vitro. Microbiology 33. Dupaul-Chicoine J, et al. (2010) Control of intestinal homeostasis, colitis, and colitis-
156:1120–1133. associated colorectal cancer by the inflammatory caspases. Immunity 32:367–378.
12. Galán JE, Ginocchio C, Costeas P (1992) Molecular and functional characterization of 34. Zaki MH, et al. (2010) The NLRP3 inflammasome protects against loss of epithelial
the Salmonella invasion gene invA: Homology of InvA to members of a new protein integrity and mortality during experimental colitis. Immunity 32:379–391.
family. J Bacteriol 174:4338–4349. 35. Cliffe LJ, et al. (2005) Accelerated intestinal epithelial cell turnover: A new mechanism
13. Van Asten FJ, Hendriks HG, Koninkx JF, Van der Zeijst BA, Gaastra W (2000) of parasite expulsion. Science 308:1463–1465.
Inactivation of the flagellin gene of Salmonella enterica serotype enteritidis strongly 36. Sherman MP, Petrak K (2005) Lactoferrin-enhanced anoikis: A defense against
Downloaded from https://www.pnas.org by 103.252.33.114 on March 12, 2022 from IP address 103.252.33.114.

reduces invasion into differentiated Caco-2 cells. FEMS Microbiol Lett 185:175–179. neonatal necrotizing enterocolitis. Med Hypotheses 65:478–482.
14. Boddicker JD, Jones BD (2004) Lon protease activity causes down-regulation of 37. Hoiseth SK, Stocker BA (1981) Aromatic-dependent Salmonella typhimurium are non-
Salmonella pathogenicity island 1 invasion gene expression after infection of epithelial virulent and effective as live vaccines. Nature 291:238–239.
cells. Infect Immun 72:2002–2013. 38. Knodler LA, et al. (2003) Salmonella type III effectors PipB and PipB2 are targeted to
15. Drecktrah D, Knodler LA, Ireland R, Steele-Mortimer O (2006) The mechanism of detergent-resistant microdomains on internal host cell membranes. Mol Microbiol 49:
Salmonella entry determines the vacuolar environment and intracellular gene 685–704.
expression. Traffic 7:39–51. 39. Valdivia RH, Falkow S (1996) Bacterial genetics by flow cytometry: Rapid isolation of
16. Cirillo DM, Valdivia RH, Monack DM, Falkow S (1998) Macrophage-dependent Salmonella typhimurium acid-inducible promoters by differential fluorescence induc-
induction of the Salmonella pathogenicity island 2 type III secretion system and its
tion. Mol Microbiol 22:367–378.
role in intracellular survival. Mol Microbiol 30:175–188.
40. Drecktrah D, et al. (2008) Dynamic behavior of Salmonella-induced membrane
17. Minamino T, Imae Y, Oosawa F, Kobayashi Y, Oosawa K (2003) Effect of intracellular
tubules in epithelial cells. Traffic 9:2117–2129.
pH on rotational speed of bacterial flagellar motors. J Bacteriol 185:1190–1194.
41. Andersen JB, et al. (1998) New unstable variants of green fluorescent protein for studies
18. Menendez A, et al. (2009) Salmonella infection of gallbladder epithelial cells drives
of transient gene expression in bacteria. Appl Environ Microbiol 64:2240–2246.
local inflammation and injury in a model of acute typhoid fever. J Infect Dis 200:
42. Knodler LA, Bertero M, Yip C, Strynadka NC, Steele-Mortimer O (2006) Structure-
1703–1713.
19. Madara JL (1990) Maintenance of the macromolecular barrier at cell extrusion sites in based mutagenesis of SigE verifies the importance of hydrophobic and electrostatic
intestinal epithelium: Physiological rearrangement of tight junctions. J Membr Biol residues in type III chaperone function. Mol Microbiol 62:928–940.
116:177–184. 43. Chong A, et al. (2008) The early phagosomal stage of Francisella tularensis determines
20. Mayhew TM, Myklebust R, Whybrow A, Jenkins R (1999) Epithelial integrity, cell optimal phagosomal escape and Francisella pathogenicity island protein expression.
death and cell loss in mammalian small intestine. Histol Histopathol 14:257–267. Infect Immun 76:5488–5499.
21. Rosenblatt J, Raff MC, Cramer LP (2001) An epithelial cell destined for apoptosis 44. Khan MA, et al. (2006) Toll-like receptor 4 contributes to colitis development but not
signals its neighbors to extrude it by an actin- and myosin-dependent mechanism. to host defense during Citrobacter rodentium infection in mice. Infect Immun 74:
Curr Biol 11:1847–1857. 2522–2536.

17738 | www.pnas.org/cgi/doi/10.1073/pnas.1006098107 Knodler et al.

You might also like