StudyArticle
StudyArticle
Correspondence
[email protected]
In Brief
Bastounis et al. show that, during
infection with the intracellular bacterial
pathogen L. monocytogenes, large
infected domains in epithelial cell
monolayers extrude to form three-
dimensional mounds due to the collective
onslaught by their uninfected neighbors.
This mechanical competition between
uninfected and bacterially infected cells
limits pathogen dissemination through
the epithelium.
Highlights
d Epithelial host cells form extruded mounds after infection
with L. monocytogenes
Article
Mechanical competition triggered by innate
immune signaling drives the collective extrusion
of bacterially infected epithelial cells
Effie E. Bastounis,1 Francisco Serrano-Alcalde,2,7 Prathima Radhakrishnan,1,3,7 Patrik Engström,4
Marı́a J. Gómez-Benito,2 Mackenzi S. Oswald,5 Yi-Ting Yeh,6 Jason G. Smith,5 Matthew D. Welch,4
José M. Garcı́a-Aznar,2 and Julie A. Theriot1,8,*
1Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
2Department of Mechanical Engineering, University of Zaragoza, Zaragoza 50009, Spain
3Biophysics Program, Stanford University, Stanford, CA 94305, USA
4Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
5Department of Microbiology, University of Washington School of Medicine, Seattle, WA 98109, USA
6Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA 92093, USA
7These authors contributed equally
8Lead contact
*Correspondence: [email protected]
https://doi.org/10.1016/j.devcel.2021.01.012
SUMMARY
Intracellular pathogens alter their host cells’ mechanics to promote dissemination through tissues.
Conversely, host cells may respond to the presence of pathogens by altering their mechanics to limit
infection. Here, we monitored epithelial cell monolayers infected with intracellular bacterial pathogens,
Listeria monocytogenes or Rickettsia parkeri, over days. Under conditions in which these pathogens
trigger innate immune signaling through NF-kB and use actin-based motility to spread non-lytically inter-
cellularly, we found that infected cell domains formed three-dimensional mounds. These mounds resulted
from uninfected cells moving toward the infection site, collectively squeezing the softer and less contrac-
tile infected cells upward and ejecting them from the monolayer. Bacteria in mounds were less able to
spread laterally in the monolayer, limiting the growth of the infection focus, while extruded infected cells
underwent cell death. Thus, the coordinated forceful action of uninfected cells actively eliminates large
domains of infected cells, consistent with this collective cell response representing an innate immunity-
driven process.
INTRODUCTION et al., 2014; Sellin et al., 2014). Similar single cell extrusion
occurs in human intestinal enteroids infected with S.Tm. or
Mammalian cells communicate through both biochemical and L.m. (Co et al., 2019). The mechanism resembles that which
biomechanical signals. Examples of the latter include the forces normal epithelia use to extrude apoptotic cells (Gudipaty and
that cells transduce to each other and to their extracellular matrix Rosenblatt, 2017). A dying cell contracts and signals its neigh-
(ECM), critical for maintaining tissue integrity and barrier func- bors to assemble a multicellular contractile ‘‘purse string’’ that
tion. Intracellular bacterial pathogens can manipulate host cell moves basally, squeezing out the extruding cell and promoting
functions, including mechanotransduction, to disseminate. For formation of cell-cell junctions among the new neighbors
example, two intracellular bacterial pathogens that generate (Kuipers et al., 2014; Rosenblatt et al., 2001; Tamada et al.,
actin comet tails for propulsion through the cytoplasm of in- 2007). At a low cell density, crawling of neighboring cells also
fected host epithelial cells, Listeria monocytogenes (L.m.) and contributes to apoptotic cell extrusion (Kocgozlu et al., 2016).
Rickettsia parkeri (R.p.), both secrete virulence factors that Some bacterial pathogens resist this innate immune extrusion
reduce intercellular tension, making it easier for bacteria to reaction by secreting virulence factors that stabilize cell-ECM
spread (Lamason et al., 2016). Conversely, host cells may exhibit adhesions (Kim et al., 2009).
biomechanical alterations after infection that could function as Epithelia that are infected by pathogens capable of direct,
innate immune responses limiting bacterial spread. In intestinal non-lytic intercellular spread face a particular challenge. In
epithelial cells infected with Salmonella enterica serovar Typhi- addition to L.m. and R.p., other intracellular bacterial patho-
murium, (S.Tm.) innate immune activation of the inflammasome gens have developed biochemically distinct mechanisms to
triggers apical extrusion of single cells, which are shed into the trigger the rapid assembly of host cell actin filaments at one
intestinal lumen without disrupting barrier function (Knodler pole of the bacterium (Stevens et al., 2006). The pushing
Developmental Cell 56, 443–460, February 22, 2021 ª 2021 Elsevier Inc. 443
ll
Article
Figure 1. Formation of infection mounds is accompannied by changes in the kinematics of L.m.-infected epithelial cell monolayers
(A and B) Orthogonal views of uninfected or L.m.-infected MDCK cells 24 hpi (host nuclei, yellow; L.m., black). In (B) a mound is shown.
(C) Barplots of L.m. mound volume for MDCK (n = 77), A431D (n = 5), and human ileal enteroid-derived cells (n = 9) 24 hpi (mean ± SD).
(D and E) 3D displacements of MDCK cell nuclei from uninfected (D) or L.m.-infected well (E, field of view centered in mound). Nuclei tracked 24–44 hpi.
Trajectories colored by time.
(F) Violin plot of mean nuclear speed of MDCK cells from uninfected or L.m.-infected wells centered in mounds ( n = 271 ± 65 tracks/experiment for 5 independent
experiments).
(G and H) Plots of weighted average for each time delay of all MSD curves of MDCK nuclei shown in examples in (D and E) (gray, weighted SD over all MSD curves;
error bars, SEM; red line, weighted mean MSD curve from which diffusion coefficient D is calculated; see STAR methods). Fit made in first 500 min. (G) shows
zoomed inset.
(I and J) Same as (F) for the average D (I) and scaling exponent a (J). (F, I, and J) dashed line, median; dotted lines, 25% and 75% quartiles of all tracks. For each of
the 5 independent experiments the average value over the field of view and time shown as triangle (mean ± SD, USTT: *p < 0.01). See also Figure S1; Video S1.
Figure 2. MDCK cells in L.m.-infected wells alter their morphology, orientation and cytoskeletal organization
(A) MDCK cells from uninfected and L.m.-infected wells 24 hpi. Columns, brightfield image; L.m., fluorescence, cells color coded by area, and radial alignment q
(see STAR methods). Purple circle surrounds mound. 4th column shows zoomed-in view.
forces generated by the actin network assembly propel the propose that coordinated mechanical forces may be a mech-
bacteria through the host cell cytoplasm and into membrane- anism employed by host epithelial cells to eliminate bacterial
bound protrusions at the host cell surface (Tilney and Portnoy, infection.
1989). In an epithelial monolayer, these protrusions facilitate
bacterial intercellular spread without causing lysis of the orig- RESULTS
inally infected cell (Lamason and Welch, 2017). Actin-driven
spread of L.m. in epithelia gives rise to infected foci Infection with Listeria monocytogenes (L.m.) alters the
comprising hundreds of cells within <24 h after the initial inva- organization and kinematics of host epithelial cells to
sion of a single cell (Ortega et al., 2019). In this situation, extru- form large multicellular mounds
sion of single infected cells using a ‘‘purse string’’ might be To study the effects of infection on the kinematics of host cells,
insufficient to limit infection. we used epithelial Madin-Darby canine kidney (MDCK) cells.
Infection of epithelial cells by many pathogens triggers acti- They form polarized monolayers in culture and are often
vation of the transcription factor, NF-kB (Dev et al., 2011). In used to study L.m. infection (Ortega et al., 2019; Pentecost
cells at rest, NF-kB is inactive and retained within the cyto- et al., 2006; Robbins et al., 1999). We grew MDCK cells to
plasm. During infection, pathogen-associated molecular pat- confluence and infected them with L.m. such that fewer than
terns initiate signal transduction cascades that lead to NF-kB 1 in 103 host cells were invaded. Cells were maintained with
activation and translocation into the nucleus. There, it induces gentamicin so that L.m. could only spread from cell to cell.
expression of genes encoding cytokines and adhesion mole- Over a period of 24–48 h, infected cell foci grew to include
cules, promoting the recruitment of immune cells to the infec- hundreds of host cells. MDCK cells in uninfected monolayers
tion site (Jiang et al., 2011). For several intracellular bacterial exhibited regularly spaced nuclei confined to a single layer
pathogens, including L.m., infection of epithelial cells in a (Figure 1A). In contrast, L.m.-infected foci formed mounds of
monolayer triggers NF-kB activation and cytokine production infected host cells, which piled on top of one another to form
not only in the infected cells but also in nearby uninfected structures 50 mm tall, with a volume of 1.7 3 105 mm3 (Fig-
bystander cells (Dolowschiak et al., 2010; Kasper et al., ures 1B, 1C, and S1A–S1C). We also observed the formation of
2010). The bystanders may play a role in the innate immune mounds in monolayers of human epithelial A431D cells ex-
response to infection by amplifying alarm signals produced pressing E-cadherin and untransformed human enteroid-
by infected cells. derived cells (Figures 1C, S1D, and S1E). Thus, infection
To determine whether epithelial monolayers alter their mound formation may be a widely conserved phenomenon
biomechanics in order to limit pathogen spread, we used for mammalian epithelial monolayers, including both cell lines
time-lapse microscopy to monitor individual infected cell foci and untransformed cells.
for several days after exposure to L.m. or R.p. For both path- Mounds arose due to directional movement of the host cells,
ogens, uninfected neighbor cells undergo a dramatic behav- as evidenced by monitoring the movement of their nuclei via
ioral change reminiscent of the epithelial-to-mesenchymal time-lapse microscopy from 24 h post-infection (hpi) onward.
transition (EMT) and act collectively to squeeze and force the Cells from uninfected wells moved randomly at low speed and
extrusion of large infected cell domains, forming ‘‘mounds.’’ appeared to be confined by their neighbors (Figure 1D). In
Mounds arise due to changes in the mechanics of two battling contrast, L.m.-infected cells within foci and their uninfected
populations—the uninfected ‘‘surrounders’’ and the infected neighbors moved rapidly and persistently toward the center of
‘‘mounders.’’ Innate immune signals, and specifically NF-kB the focus and infected cells also moved upward to form the
activation, are major drivers in this mechanical competition. 3D mound (Figure 1E; Video S1). Net speeds inside and near
Importantly, cells infected with wild-type (WT) R.p. do not acti- L.m.-infected foci increased by 3-fold relative to uninfected
vate NF-kB or form mounds whereas R.p. mutants lacking the monolayers and cell tracks became straighter (Figures 1F and
outer membrane protein B (OmpB), activate NF-kB and do S1F). Over an observation period of 16 h, the average mean
form mounds. Our results demonstrate that the mechanical squared displacement (MSD) for cells from an uninfected well re-
changes in epithelial cells leading to mounds are not simply mained under 50 mm2 (7 mm net displacement). Movement was
a result of cytoskeletal remodeling associated with infection constrained, with the MSD reaching a plateau rather than
but are triggered by innate immune signals. Moreover, our increasing linearly with time as in unconstrained, random move-
findings connect EMT, a conserved cell behavioral transforma- ment (Figure 1G). In contrast, the average MSD for cells in and
tion critical in development and pathologies (e.g., fibrosis and near L.m. foci grew to over 1,000 mm2 over the same time frame
cancer), with innate immune mechanisms. They also underline and increased superdiffusively, indicative of directed motion
the dynamic remodeling capability of epithelia and lead us to (Figures 1H–1J). Thus, cells undergo a behavioral phase
(B) Sketch showing that radial alignment is the angle q between the direction of the cell’s major axis and the axis between the cell centroid and the mound center.
(C) Mean q versus distance from the center of the field of view of cells from uninfected or L.m.-infected wells for the example shown in (A) (solid line, median;
shaded areas, 40, 60 percentiles).
(D) Violin plot of aspect ratio of cells from uninfected wells (n = 5,817), uninfected surrounders (n = 5,163, considered up to ~200 mm away from mound), and
infected mounders (n = 1,248). Dashed line, median; dotted lines, 25%, 75% quartiles. For each of the 3 independent experiments the average value over the field
of view is shown as triangle (mean ± SD, USTT: *p < 0.05).
(E) Orthogonal views of MDCK cells from uninfected or L.m.-infected wells 24 hpi (host nuclei, yellow; L.m., black) and corresponding F-actin or vimentin
localization. See also Figure S2.
no competition
20% infection
100% infection
C Phase contrast L.m. √ux2+uy2 (μm) Stresses (Pa) towards u (μm) away from Overlay of
center r center
0 0.5 1 0 100 200 -1 0 1 ur and L.m.
t=4 hpi
50 μm
t=8 hpi
t=12 hpi
t=16 hpi
t=20 hpi
t=24 hpi
10
cells relative to 8 hpi
20 40 1.5
Time (hpi)
30 *
40
50 1.0
60 20
70 0.5
80
90
0 100 200 300 0 100 200 300 0 0
in ers
ed
nd d
s
un
m c
fe
washes - + - +
f
in
rro
un
su
transition, switching from a caged state to a highly motile state at Video S2). In contrast, the radial deformations, ur, of surround-
late stages of infection. ers at the edge of the mound were large, indicative of them
Changes in kinematics were accompanied by alterations in grabbing the ECM and pulling it away from the mound as
cell morphology. In uninfected monolayers, MDCK cells formed they move directionally toward it. This traction stress orienta-
randomly oriented regularly packed polygons (Figures 2A and tion is not consistent with extrusion generated by a ‘‘purse
S2A). In infected monolayers, infected cells at the mounds’ string’’ but is consistent with lamellipodial protrusion and
base (mounders) had smaller z-projected areas than uninfected directed cell migration (Kocgozlu et al., 2016). Thus, mounds
cells (surrounders), up to 200 mm away from the mounds or cells are not caused by contraction of infected cells but rather by
from uninfected wells (Figures 2A and S2C). Strikingly, the orien- active crawling of uninfected surrounders that migrate toward
tation of the long axis of surrounders pointed toward the mound the focus, squeezing and extruding the infected cells. Kymo-
center, and this persisted over 150 mm away from the mound graphs of ur and L.m. fluorescence as a function of radial dis-
(Figures 2A–2C). Surrounders showed alignment of their long tance from the mound’s center further confirmed that the
axes parallel to that of their immediate neighbors, indicative of largest ur coincided with the edge of the infection focus (Fig-
local nematic order in the monolayer (Figure S2B). The aspect ures 3D, S3A, and S3B). This behavior persisted up to
ratio and shape factor q (perimeter divided by square root of 54 hpi, with the peak of forces moving outward as the infection
area), dimensionless numbers used to characterize cell shapes focus grew. After 54 hpi the peak L.m. fluorescence started to
in EMT (Mitchel et al., 2019), were also increased in cells from decay. This could be due to the extrusion of infected cells and
infected wells relative to uninfected wells (Figures 2D and the inability of the bacteria they carry to spread to new unin-
S2D). To determine whether these changes are due to cytoskel- fected cells in the basal cell layer. To explore this further, we
etal rearrangements associated with infection, we performed infected cells with fluorescent L.m. and measured the effect
immunostaining (Figure 2E). While F-actin localized pericellu- of repetitive washes (to get rid of extruded cells) on the recov-
larly for cells in uninfected wells, in infected wells, cells con- ery of infected cells from the monolayer. We found that late in
tained less pericellular actin. Actin tails were visible in infected infection, half of the infected cells had been extruded into
cells and surrounders showed prominent stress fibers. Microtu- mounds (Figure 3E).
bules were disrupted in infected cells at the mounds’ base, as Infected cells might generate reduced traction because of
were intermediate filaments, vimentin and cytokeratin (Figures secreted L.m. virulence factors that weaken cellular tension,
2E, S2E, and S2F). The cytoskeletal changes of mounders such as the internalins C (InlC) (Rajabian et al., 2009) and P
versus surrounders are consistent with the display of distinct (InlP) (Faralla et al., 2018). To test this, we infected MDCK
mechanical properties of these cells that could contribute to with DinlC or DinlP L.m. but found no difference in resulting
mounding. Alternatively, these changes could be the result of mound volumes (Figures S3C–S3E). Since the listeriolysin O
mounders being squeezed and extruded and not the cause of toxin (LLO) could also modulate mechanotransduction, we in-
mounding. fected cells with LLOG486D L.m. (Rengarajan et al., 2016). This
led to formation of smaller mounds; however, the total L.m.
Mechanical cellular competition drives infected cell load was also decreased comparably to the degree of mound
extrusion en masse volume reduction. These results suggest that the functions of
To test whether mounding requires competition between two known secreted bacterial virulence factors do not drive the
cell populations, we compared MDCK cells infected with cellular changes that lead to mechanical competition and
low (20% infected at 24 hpi) and high (100% infected at mounds.
24 hpi) multiplicity of infection (MOI). Although mounds were The weakening in mounders’ tractions could be due to
visible at low MOI, they were absent in wells infected at high changes in their cytoskeletal integrity, resulting in a reduced
MOI (Figures 3A and 3B). This suggests that mounding may ability to transmit forces. To determine whether cell stiffness
not be a consequence of the loss of cell mechanical integrity changes during infection, we used atomic force microscopy
by infected cells but instead may require the involvement of (AFM). We found that surrounders’ stiffness was 4-fold higher
uninfected surrounders in a form of mechanical competition. than that of mounders (Figure 3F). As compared with cells
To test this, we used traction force microscopy (TFM) to mea- from uninfected wells, surrounders were 2.5-fold stiffer and
sure the forces exerted by cells on a soft 3 kPa hydrogel. We mounders 1.6-fold softer, consistent with the cytoskeletal
found that, as the infection focus grew, infected cells imparted disruption of infected cells at the mounds’ base noted in Fig-
reduced deformations and stresses onto their ECM (Figure 3C; ure 2D. In summary, we found that infected cells became softer
Figure 3. Infection with low bacterial dosage elicits a mechanical competition between uninfected and L.m.-infected cells
(A and B) Phase contrast images 24 hpi of MDCK cells and corresponding L.m. fluorescence for wells that are 20% or 100% infected.
(C) TFM on MDCK cells adherent on 3 kPa hydrogel and infected at low MOI with L.m. Columns, phase contrast image, L.m. fluorescence, deformations (mm),
traction stresses (Pa), radial deformations (ur: positive values indicate deformations pointing away from the focus center), and overlay of ur and L.m. fluorescence.
Rows, p.i. time.
(D) Kymograph of mean ur and radial L.m. fluorescence as a function of time. The infection focus center is considered the center of the polar coordinate system.
(E) Boxplots of fold increase relative to 8 hpi in % of L.m.-infected MDCK cells (flow cytometry). Cells were washed (or not) to get rid of extruded cells (n = 6
samples, mean ± SD, WRST, *p < 0.01).
(F) Violin plots of cell stiffness (kPa) for MDCK cells from uninfected wells (n = 68), surrounders (n = 126), and mounders (n = 130). Cells were indented 2–8 times.
Dashed line: median, dotted lines: 25% and 75% quartiles. For each cell, its average stiffness is shown as triangle (mean ± SD, WRST: * p < 0.01). See also
Figure S3; Video S2.
A B C *
* Nuclei L.m. Nuclei L.m. /Y-27632 3
* *
* 40 μm
2 19 μm
2
1
1
0 0
in
2
eb 52
l
D O
l
tro
M cad
tro
63
at
K
M at K
1
C
st
on
27
on
K
-1
KD E-
D
bi
C
H
C
c
Y-
αE
Bl
D E
Nuclei L.m. αEcat KO nuclei, L.m. 200 ns
100
ns
50
0
time (hpi) 24 24 48 48
washes - + - +
F Phase contrast L.m. √ux2+uy2 (μm) Stresses (Pa) towards ur (μm) away from Overlay of
center center
αEcat KO MDCK 0 0.05 0.1 0 25 50 -0.1 0 0.1 ur and L.m.
t=4 hpi
t=12 hpi
t=20 hpi
t=28 hpi
and less contractile than cells in uninfected monolayers. MDCK cells expressing E-cadherin-RFP (to distinguish popu-
Concurrently, nearby uninfected cells became stiffer and direc- lations) in varying ratios. When 75% of host cells were WT,
tionally polarized, gripped the substrate, and actively pulled on mounds resembled 100% WT monolayers. In contrast, when
it to move themselves directionally toward the focus. This 75% of cells were aEcat KO, mounds were reduced and
competition resulted in infected cells being squeezed and resembled 100% aEcat KO monolayers (data not shown).
ejected from the monolayer plane. Intriguingly, the higher the percentage of aEcat KO in the in-
fected monolayer the lower the correlation length of movement
Cell-cell adhesions and actomyosin contractility between neighboring cells, suggesting that lack of coordi-
contribute to infection mounds nated cell movement may inhibit mounding (Figure S4J). Inter-
As mounders and surrounders differ in their biomechanics, we estingly, when cells were mixed 1:1, we only observed
hypothesized that perturbations of cellular forces should inhibit mounds in foci where infected mounders and immediate
mounds. When L.m.-infected MDCK cells were treated with surrounders were predominately WT, while cells further away
ROCK inhibitors, Y-27632 and H1152, or the myosin II inhibitor, from the mound were of either lineage (Figure S4K). This
blebbistatin, to decrease actomyosin contractility and cellular result, further explored below through simulations, suggests
tractions, we found a reduction in mound volume (Figures 4A, that the presence of functional E-cadherin-based junctions,
4B, and S4A). We then examined the role of intercellular forces at least in the infected mounders and the uninfected cells
by infecting MDCK cells deleted for aEcatenin (aEcat KO) (Or- immediately surrounding the mound, is necessary for mounds
tega et al., 2017) or knocked down for E-cadherin, with L.m. and to emerge.
found a dramatic reduction in mound volume (Figures 4C, 4D,
and S4B). Compared to WT MDCK, the speed of aEcat KO cells Simulations suggest that cell stiffness, contractility and
was increased but directional persistence and neighbor coordi- intercellular adhesions are critical for mound formation
nation were decreased (Figure S4C), suggesting that coordina- To explore the mechanism whereby changes in cell mechanics
tion of movement of neighboring cells through robust contacts lead to the collective extrusion of infected cells, we followed a
is important for mounding. Moreover, we found that, as the infec- computational approach that formulated the problem in 3D. In
tion focus grew, differences in traction of infected versus unin- our simplified model, infected versus uninfected cells can have
fected cells were abrogated in aEcat KO MDCK cells (Figure 4F; distinct mechanical properties, but infection is fixed (bacteria
Video S3). These findings confirm that cell-cell and cell-ECM do not spread/replicate). Cells are hexagonal, can deform in all
forces are tightly coupled and that a difference in force transduc- directions, are divided into 3 domains (contractile, adhesive,
tion between the two populations is important for mounds to protruding), and have both a passive stiffness arising from the
emerge. cytoskeleton and an active contractility associated with the acto-
Interestingly, L.m. infection foci for aEcat KO cells myosin contractile apparatus (Figures 5A and S5A–S5C; Sunyer
(compared with WT MDCK) were larger and had increased to- et al., 2016). Cell-ECM interactions are considered through
tal L.m. fluorescence but were less dense in fluorescence in- contact interactions and cell-cell adhesions as a continuous
tensity (Figures S4D–S4F). Consistently, we found that the material, following the mechanism depicted in Figure S5A. If
fold increase in L.m.-infected aEcat KO cells 24 and 48 hpi cell-ECM displacements are large when cells contract, new
relative to 8 hpi was higher as compared with infected WT cell-ECM adhesions are formed. If cell-ECM displacements are
MDCK, and vigorous washing of the L.m.-infected aEcat KO small and there is tensional asymmetry, new protrusions form
monolayers did not reduce the percentage of infected cells at the edge of the cells that are experiencing minimum stress, fol-
(compare Figures 3E and 4E), further supporting the conclu- lowed by another round of cell contraction. If there is no tensional
sion that bacterial spread through the monolayer is more effi- asymmetry, there is no protrusion and the given cell just
cient when mounds cannot form. To confirm the role of adhe- contracts.
rens junctions in L.m. spread and mounds, we infected human We assumed that infected cells decrease their passive stiff-
A431D cells, which mostly do not express E-cadherin, with ness and active contractility, based on our experimental results,
L.m. and found a similar inhibition of mound formation (Figures and examined 4 different cases (Figures 5A and S5D–S5F). In
S4G–S4H). case 1, all cells were uninfected, so during the contraction phase
To examine the organization of cell-cell adhesions in moun- cell displacements and principal stresses were symmetrical and
ders and surrounders, we performed immunostaining for E- thus cells did not create protrusions. However, in case 2, where a
cadherin and ZO-1 to localize adherens and tight junctions. cluster of 7 cells was infected, uninfected cells in close proximity
We found more fluorescent puncta of E-cadherin and moder- developed tensional asymmetry but as they were unable to form
ate disruption of ZO-1 for infected cells at the mounds’ base new cell-ECM adhesions they became displaced away from the
(Figure S4I). To explore the relative contributions of cell-cell focus’s center, which was opposite to what observed in our ex-
junctions in mounders versus surrounders, we then conducted periments. In contrast, in case 3, where we allowed uninfected
infections in monolayers of aEcat KO MDCK mixed with WT cells to form new cell-ECM adhesions during the protrusion
(B) Orthogonal views of L.m.-infected MDCK cells treated with vehicle control or 30 mM of Y27632 24 hpi (host nuclei, yellow; L.m., black).
(C) Barplots of relative L.m. mound volume same as (A) but for WT MDCK, aEcat KO MDCK or MDCK cells treated with siRNA against E-cadherin.
(D) Orthogonal views of WT or aEcat KO MDCK cells infected with L.m. 24 hpi.
(E) Boxplots same as in Figure 3E but for L.m.-infected aEcat KO MDCK cells (n = 6 samples).
(F) Same as Figure 3C but for L.m.-infected aEcat KO MDCK cells. See also Figure S4; Video S3.
phase, we found that uninfected cells in close proximity to in- fected cells (Figures 5B–5D). Surrounders performed less recoil
fected cells developed tensional asymmetry and were displaced than infected mounders on the opposite side of the wound.
toward the infection focus. This led to squeezing of the infected Active crawling to close the wound was observed in both in-
cells and an increase in their height. Finally, in case 4, where fected and uninfected cells. We also used our computational
intercellular adhesions were absent, there was no tensional model to examine predicted responses, which were consistent
asymmetry and thus no protrusions. Cases 3 and 4 were consis- with our experiments (Figure S5I). These findings first show that
tent with our experiments and suggest that surrounders need to the infected cells are capable of mounting a wound-healing
form protrusions and cell-ECM adhesions toward the infection response and that they move away from the focus center to-
focus for mounds to form, and that the lack of intercellular adhe- ward the wound margin. This reinforces that cell kinematics
sions prevents mounding. associated with mounding are specifically due to mechanical
We also examined what would happen if only one of the me- competition between surrounders and mounders, where
chanical parameters was altered in infected cells and found surrounders are stronger, even though mounders retain their
that, for mounds to arise, it is necessary for infected cells to normal ability to mount a directional wound-healing response
decrease their passive stiffness and/or active contractility while despite their infected status. Second, surrounders exhibit
forming robust cell-cell adhesions (Figures 5A and S5G, cases reduced intercellular tension relative to cells from uninfected
5 and 6). We also found that if only infected mounders and monolayers.
surrounders immediately adjacent to them (immediate surround-
ers) form cell-cell adhesions but uninfected cells further away do RNA sequencing reveals distinct transcriptional profiles
not, that is also sufficient to drive mounding (Figure S5H). How- among uninfected cells, infected mounders and
ever, in the opposite case where infected mounders and imme- surrounders
diate surrounders do not form cell-cell adhesions but uninfected To explore how signaling regulates the mechanical changes
cells further away do, mounding is stalled (Figure S5H). In both associated with mounding, we followed a transcriptomics
cases, cells that are unable to form intercellular adhesions create approach. L.m.-infected MDCK cells were flow sorted into
stronger cell-ECM adhesions and tensional asymmetry arises mounders (L.m.-positive) and surrounders (L.m.-negative),
when cells contract. However, the levels of stress and asymme- 24 hpi (Figure 6A). RNA sequencing revealed significant
try are different; thus, when protrusion takes place, mounds numbers of differentially expressed genes (DEGs) when
occur only in the first scenario. Therefore, cell-cell adhesions comparing these two groups to each other and to cells origi-
specifically for mounders and immediate surrounders are crit- nating from uninfected wells (Figures 6B and 6C; Table S1).
ical, since a lack of such adhesions inhibits mounding, consis- Compared to cells not exposed to infection, both mounders
tent with our experiments. and surrounders downregulated genes related to intercellular
and focal adhesions and to the regulation of the actin cytoskel-
Laser ablation demonstrates alterations in tension at eton (Figure 6D; Table S1). Genes whose changes in expres-
cell-cell junctions for infected epithelial monolayers sion are associated with EMT were differentially regulated for
Because simulations indicated the importance of tensional both populations originating from infected wells compared
asymmetries at junctions between infected cells and their unin- with cells not exposed to bacteria (e.g., MMP1, VIM, SNAI1,
fected neighbors, we sought to confirm these predictions by and SNAI2). Surprisingly, mounders showed upregulation in
performing laser ablation (Fernandez-Gonzalez et al., 2009; genes associated with DNA replication and ferroptosis, hinting
Joshi et al., 2010; Kiehart et al., 2000; Kong et al., 2019; at a stress response or death-related process that infected
Smutny et al., 2015). When we ablated rows of cells in unin- cells might undergo, which could be linked to mounding.
fected monolayers, we observed rapid, symmetric recoil of Indeed, using several approaches (see STAR methods) we
cells away from the wound on both sides, over the first few mi- confirmed that cells in mounds undergo death, which rein-
nutes after wounding (Figures 5B–5D). This response is indica- forces the idea that this process is beneficial for the host, since
tive of pre-existing cell-cell tension throughout the monolayer infected cells are being cleared out of the monolayer (Figures
and was followed by active crawling of cells to close the S6A–S6E) and which raises the question of whether cell death
wound. However, the magnitude of the immediate recoil in precedes or follows extrusion.
response to the ablation for cells at the margin of L.m.-mounds Surrounders and to a lesser degree mounders showed upre-
was markedly asymmetric and substantially less than for unin- gulation of genes associated with the IL-17, NF-kB, and TNF
Figure 5. Simulations reveal that changes in cell stiffness, contractility, and intercellular forces drive mound formation, and laser wounding
reveals that tension is reduced in infection
(A) Cases: (1) all cells uninfected, (2–3) infected cells (denoted with asterisk) are softer and uninfected cells cannot (2) or can (3) create new cell-ECM adhesions, (4)
infected cells are softer but cell-cell adhesions are disrupted, (5–6) infected cells reduce only their contractility (5) or passive stiffness (6) and uninfected cells form
new ECM adhesions. Plots show cell displacements in the vertical (Z) direction in two views.
(B) Laser wounding response for MDCK cells from uninfected (top) or L.m.-infected wells (bottom) 24 hpi. Columns, brightfield image superimposed with L.m.
fluorescence (green); corresponding E-cadherin localization prior to wounding (t = 0 min); E-cadherin localization immediately after 1 min of laser illumination
(t = 1 min); cell displacement vectors u (50-fold larger); average uy at t = 1 min (positive/negative values point toward/away from wound); average uy over the time
period 30–40 min.
(C) Kymographs of the average, with respect to the wound, uy as a function of time, t and vertical position, y for the examples in (B) (see single and double crosses).
(C) Barplots of the mean uy (along a distance of 100 mm away from the wound, calculated immediately after ablation) for cells originating from uninfected wells,
surrounders and infected mounders (results from 3 independent experiments, mean ± SD, USTT: *p < 0.01). See also Figure S5; Video S4.
innate immune signaling pathways, and NF-kB activation in have been implicated in the regulation of EMT genes such as
infection was further confirmed through western blotting (Fig- SNAI1 (Li et al., 2017; Pires et al., 2017; Strippoli et al.,
ure S6F). This indicates that, although surrounders are not them- 2008; Tian et al., 2018; Weichhart et al., 2015) and given the
selves infected with L.m., their distinct mechanical behavior 6- and 9-fold increase in SNAI1 expression in surrounders
might arise from cytokines released in infection, which influence and mounders, respectively, as compared with cells from unin-
their phenotype, gene expression, and mechanics. Indeed, we fected cultures, we also knocked down SNAI1 in MDCK cells
found that the supernatant of cells infected for 24 h with L.m. (Figure S7F) and found a 50% decrease in mound volume
had enriched GM-CSF, IL-8, and MCP-1 compared with the su- (Figures 7D, 7E, and S7F). Since reactive oxygen species
pernatant of uninfected cells (Figure 6E). Interestingly, gene (ROS) and nitric oxide species (NOS) could be generated in
expression of these cytokines was upregulated to a greater cells in L.m.-infected wells and trigger NF-kB activation (Gouin
extent in surrounders compared with mounders (Figure 6F; Table et al., 2019; McFarland et al., 2018; Morgan and Liu, 2011), we
S1). All three cytokines are associated with NF-kB signaling, treated cells with diphenyleneiodonium (DPI) and L-NIL that
either being NF-kB activators or expressed and secreted in inhibit ROS and NOS generation, respectively, and found a sig-
response to it, or both (Hoesel and Schmid, 2013; Kang et al., nificant reduction in mound volume only for cells where ROS
2007; Manna and Ramesh, 2005). To assess whether cytokine production was inhibited (Figures S7G and S7H). We also
signaling alone, without actual infection, can induce changes in used the DCFHA assay to assess ROS generation and found
cell polarization and nematic order as observed in surrounders, a small increase in DCF fluorescence, indicative of ROS gener-
we exposed MDCK cells to conditioned media from L.m.-in- ation, for cells originating from L.m.-infected wells compared
fected wells for 24 h and found that the area and aspect ratio with uninfected wells (Figure S7I). Overall, these results sug-
of these cells were both greater than that of control cells (Figures gest that innate immune signaling through NF-kB activation,
S6G and S6H). Thus, at least part of the response and collective possibly driven by ROS production, and the subsequent cyto-
behavioral change of the surrounders is due to paracrine kine signaling play a role in promoting mounds, contributing to
signaling. loss of mechanical stiffness in mounders, and triggering cell
shape changes and motility responses similar to EMT in
Apoptosis inhibition does not attenuate mounds, but surrounders.
perturbations in NF-kB signaling do If our conclusion that NF-kB activation contributes to
Given that infected cells undergo cell death, we tested whether mounds is correct, then we would also observe mounds if we
this was required for mounding by treating infected monolayers were to infect MDCK cells with a different intracellular bacterial
with either Z-VAD-FMK or GSK0 872 3, which inhibit apoptosis pathogen that activates NF-kB. To test this, we infected MDCK
or necroptosis, respectively, associated with L.m. infection cells with WT R.p. or the ompB mutant (Figures 7F and 7G).
(Zhang and Balachandran, 2019). We found that mounds OmpB protects the rickettsial surface from ubiquitination in
were similar in volume to control mounds (Figures 7A and the cytoplasm of infected host cells (Engström et al., 2019),
7B), suggesting that cell death occurs after infected cell extru- and when S.Tm. become ubiquitinated, NF-kB is activated
sion and not before. (Noad et al., 2017). We thus hypothesized that ompB R.p.
Since NF-kB-related pathways were upregulated in cells would trigger NF-kB activation, whereas WT R.p. would not.
originating from infected wells, we tested whether inhibiting Observation of NF-kB nuclear translocation and RT-PCR on
NF-kB with the NF-kB essential modulator (NEMO)-binding NF-kB target genes confirmed this hypothesis (Figures 7H,
peptide would affect mound volume, and found a 25% S7A, and S7B). Importantly, host cells infected with ompB
reduction compared with control mounds (Figures 7C and R.p. formed mounds, while those infected with WT R.p. did
7E). Inhibition of NF-kB was confirmed through western blot- not, and ompB R.p. mounds were inhibited by treatment
ting and immunostaining (Figures S6F, S7A, and S7B). More- with the NEMO-binding peptide (Figures 7F and 7G). Similar
over, treatment of infected cells with the NEMO-binding pep- to L.m. mounds, we found that ZO-1 localization was perturbed
tide suppressed the loss of vimentin from the cells at the for ompB R.p. mounds but not for WT R.p.-infected MDCK
mound’s base, to a comparable extent as the loss of cell-cell cells which resembled cells from uninfected wells (Figure S7J).
junctions (Figures S7C–S7E). This is consistent with the idea Also, similar to L.m.-infected MDCK cells, the supernatant
that cytoskeletal changes associated with the decrease in pas- coming from ompB R.p.-infected MDCK was 2-fold richer in
sive stiffness of infected cells might be downstream conse- MCP-1 compared with supernatant from WT R.p.-infected
quences of NF-kB activation. As both MCP-1 and NF-kB MDCK and uninfected cells (Figure 7K). Together, these results
Figure 6. Infected mounders, surrounders, and cells from uninfected wells display distinct transcriptional profiles
(A) Sketch of the RNA-sequenced MDCK cell populations (n = 4 replicates): cells from uninfected well, L.m.-positive and L.m.-negative cells from infected well.
(B) Volcano plots of DEGs. The log10 p values are plotted against the average log2-fold changes in expression. For each pair of conditions compared, upre-
gulated genes of each group are shown in the corresponding color.
(C) PCA on top genes that have ANOVA p value % 0.05 on FPKM abundance estimations. PC1 versus PC3.
(D) Pathway enrichment analysis. Infected mounders or surrounders are compared with uninfected cells based on their enrichment score (log10p).
(E) Barplots of cytokine concentration in the supernatant of cells originating from an uninfected or L.m.-infected well 4 or 24 hpi (n = 8 samples originating from
2 independent experiments, mean ± SD, WRST: *p < 0.01).
(F) Barplots of gene expression of CSF2, CXCL8, and CCL2 for infected mounders or surrounders compared with cells from uninfected wells (n = 4, mean ± SD,
WRST: *p < 0.01). See also Figure S6.
3 33 μm 30 μm 30 μm
0
72
l
K
tro
M
'8
on
-F
30 μm
KS
D
C
VA
Z-
C D E Nuclei L.m.
NEMO binding peptide SNAI1 KD nuclei L.m.
*
Relative mound volume
2.0 * 3
20 μm 19 μm
1.5
2
1.0
1
0.5
0.0 0
g
C KD
pe O b ol
l
id din
tro
tr
EM on
M AI1
30 μm
on
pt n
K
i
e
C
SN
D
N
0
R.p. WT ompB- WT ompB-
15 μm NEMO-binding
- - + +
peptide
* 80 2.0
* *
* 3 *
3 * 1.5 * 1.5
* 60
ns ns ns
2 1.0 2 * 1.0
40 * *
ns *
1 0.5 20 1 0.5
ns
0 0.0 0 0 0.0
bacteria - L.m. - R.p. ompB- - L.m. - R.p. ompB- - L.m. - R.p. ompB -
- L.m. - R.p. ompB- - L.m. - R.p. ompB-
time (hpi) 24 72 24 72 24 72 24 72 24 72
Figure 7. NF-kB activation contributes to mounding for two unrelated bacterial pathogens
(A) Barplots of L.m. mound volume 24 hpi for MDCK cells treated with 5 mM GSK0 872, or 100 mM Z-VAD-FMK relative to control mound volumes (mean ± SD,
WRST: *p < 0.01).
(B) Orthogonal views of L.m.-infected MDCK cells treated with vehicle control, 5 mM GSK0 872, or 100 mM Z-VAD-FMK (host nuclei, yellow; L.m., black).
support that cellular changes in mechanics associated with nessed in the context of bacterial infection to contribute to
mounds are not simply a result of cytoskeletal alterations clearance of infected cells and re-epithelization. Mechanical
driven by pathogens such as L.m. and R.p. that use actin competition between two cell populations, where one of
comet tails for intercellular spread. Rather, NF-kB signaling is them loses and gets eliminated, have been studied during
key to eliciting the mechanical competition between infected oncogenesis and various processes are thought to lead to
and uninfected cells that leads to mounds and collective clear- the elimination of the losers (Matamoro-Vidal and Levayer,
ance of infected cells in epithelial monolayers. 2019)Hogan et al. (2009); Wagstaff et al. (2016). Here, we
found that paracrine signaling that arises due to infection is a
DISCUSSION major driver, consistent with previous studies on viral infection
of epithelia (Abaitua et al., 2013; Beerli et al., 2019). Differential
Mechanical forces drive the remodeling of tissues during sensitivity to mechanical stress is also key, since both cell stiff-
morphogenesis and homeostasis (Ohsawa et al., 2018). In ness and tractions are dramatically different between moun-
the epithelium, these forces promote cell extrusion to prevent ders and surrounders. Notably though, mounders remain
accumulation of excess or unfit cells (Grieve and Rabouille, capable of initiating a migratory wound-healing response,
2014; Gudipaty and Rosenblatt, 2017). Infection of epithelial demonstrating that they are not mechanically passive but
monolayers with intracellular bacterial pathogens also triggers rather that they cannot resist the coordinated onslaught by
extrusion of single infected cells, which represents an innate surrounders. Other signals may also contribute to coordinated
immune protective mechanism of the host (Knodler et al., behavior of surrounders. For example, the MEK-ERK axis has
2014; Sellin et al., 2014), although in some cases it contributes been implicated in the collective extrusion of UV-treated cells
to bacterial spread (Knodler et al., 2010). Here, we describe a (Aikin et al., 2019) and of virally infected cells (Beerli
collective mechanical response of epithelial monolayers, et al., 2019).
actively forming mounds that can contribute to the clearance In this work, we are intrigued by the central role of NF-kB in
of domains comprising hundreds of infected cells. We argue mound formation for both L.m. and R.p. because of its involve-
that this is beneficial for the host, since bacteria carried by ment in the host’s innate immune response to infection by many
extruded cells cannot spread along the basal monolayer, pathogens (Dev et al., 2011). Studies have shown that L.m.
limiting the growth of the infection focus. Additionally, extruded infection of certain cell types leads to NF-kB activation (Rahman
cells undergo death, probably due to their forcible separation and McFadden, 2011), although the specific trigger(s) from the
from survival signals communicated to normal epithelia by bacterial side may vary (Drolia et al., 2018; Gouin et al., 2010;
direct contact with their basement membrane. In the geometry Mansell et al., 2000). ROS promotes intercellular communica-
associated with the primary site of L.m. infection in mammalian tion in infection (Dolowschiak et al., 2010) and signaling that
hosts, the small intestine, the extrusion of infected cells would leads to NF-kB activation in L.m.-infected cells (Herb et al.,
drive their shedding into the intestinal lumen, where they could 2019). We found that ROS are involved in L.m.-mounding but
be eliminated from the body via the fecal route. Intriguingly, whether they act upstream of NF-kB is yet to be determined.
bacterial infections in the intestine of Drosophila melanogaster, In our experiments, we did not observe NF-kB activation in
a model for human intestinal infection (Apidianakis and Rahme, MDCK cells infected with WT R.p., which correlated with lack
2011), trigger massive remodeling of the intestinal epithelium, of mounds, in contrast to L.m.- and ompB R.p.-infected cells.
featuring elimination of infected cells and replacement of This provides one rationale as to why many pathogens,
damaged tissue by remodeling (Buchon et al., 2010). Likewise, including viruses, that are highly adapted to an intracellular life-
infection of epithelia with intracellular viral pathogens increases style and specifically those that are capable of non-lytic intercel-
polarization and motility of uninfected cells, leading to mounds lular spread, may have evolved mechanisms to suppress activa-
of infected cells in vitro and in vivo (Abaitua et al., 2013; Beerli tion of immune signaling pathways that otherwise would lead
et al., 2019). to mounds (Rahman and McFadden, 2011; Reddick and
In our system, mound extrusion depends on active crawling Alto, 2014).
of uninfected cells surrounding the infection focus, with those Our discovery underscores the importance of mechanics in
uninfected cells becoming morphologically polarized and nem- regulating infection and the relationship between innate im-
atically ordered. Both the softer and less contractile infected mune signals and cellular biomechanics, specifically EMT.
cells of the mound and their uninfected neighbors undergo a Intriguingly, NF-kB activation is linked to EMT in contexts not
biomechanical transition reminiscent of EMT (compare Yang involving infection (Strippoli et al., 2008; Tian et al., 2018).
et al., 2020), suggesting that EMT can be specifically har- Studying the dynamics of these signals using live-cell
(C) Barplots similar to (A) of relative L.m. mound volume for cells treated with 50 mM of NEMO-binding peptide.
(D) Barplots similar to (A) of relative L.m. mound volume for MDCK cells treated with siRNA against SNAI1 compared to control non-targeting siRNA.
(E) Orthogonal views of L.m.-infected MDCK cells treated with 50 mM of NEMO-binding peptide (left) or siRNA against SNAI1 (right).
(F) Orthogonal views of WT R.p.- or ompB R.p.-infected MDCK cells 72 hpi.
(G) Barplots of mound volume 72 hpi for MDCK cells infected with WT R.p. or ompB R.p. (n = 2 experiments) treated (or not) with the NEMO-binding peptide
relative to control mound volume (n = 1 experiment) (mean ± SD, WRST, *p < 0.01).
(H) Boxplots of relative levels of NF-kB target genes obtained by RT-qPCR for MDCK cells originating from uninfected or infected wells with L.m., WT R.p., or
ompB R.p. For infection with L.m. or R.p, expression levels are normalized relative to expression of control uninfected cells (n = 4, mean ± SD, t test: *p < 0.01).
See also Figure S7.
biosensors will further reveal the spatiotemporal crosstalk be- SUPPLEMENTAL INFORMATION
tween innate immunity and cell biomechanics. In this work, we
Supplemental Information can be found online at https://doi.org/10.1016/j.
propose a general cooperative epithelial extrusion mechanism,
devcel.2021.01.012.
that could quickly help limit the local spread of infection in
epithelia, particularly in the vulnerable surfaces of the lung
ACKNOWLEDGMENTS
and intestine, common sites of pathogen invasion where the
outside is separated from the inside of the body by a single We are grateful to Nigel Orme and Matthew Footer for preparation of the
epithelial monolayer. Whether these results can be recapitu- graphical abstract. We thank M. Footer, M. Walkiewicz, K. Kirkegaard, D. Kie-
lated in vivo in the presence of additional cells, varying ECM hart, J. Woodward, and members of the Theriot Lab for discussions and exper-
topography, and mechanical cues has not yet been explored. imental support. We thank A. Hayer for sharing code. We thank the Stanford
Continued investigation using more complex assays will un- Cell Sciences Imaging Facility for use of the Atomic Force Microscope as
well as National Center for Research Resources (NCRR), award number
cover the precise biomechanical signals that regulate infection
1S10OD021514-01. This research was supported by the Cell Analysis Facility
and how host cells modulate those by orchestrating an innate Flow Cytometry and Imaging Core in the Department of Immunology at the
immune response to clear infection. University of Washington. RNA-seq and RT-qPCR were performed by Arrays-
tar Inc. and multiplex immunoassay by PBL Assay Sciences. This work was
supported in part by NIH R01AI036929 (J.A.T.), NIH R01AI109044 (M.D.W),
STAR+METHODS
NIH R01AI104920 (J.G.S.), HHMI (J.A.T.), Spanish Government, award num-
ber RTI2018- 094494-B-C21 (J.G.A. and M.G.B.), and the American Heart As-
Detailed methods are provided in the online version of this paper sociation, award number: 18CDA34070047 (E.E.B.).
and include the following:
AUTHOR CONTRIBUTIONS
d KEY RESOURCES TABLE
d RESOURCE AVAILABILITY Conceptualization, E.E.B. and J.A.T.; methodology, E.E.B., J.A.T., M.D.W.,
B Lead contact P.E., M.G.B., F.S.-A., P.R., J.M.G.-A., M.S.O., and J.G.S.; software,
E.E.B., F.S.-A., and P.R.; investigation, E.E.B., P.E., M.J.G.-B., F.S.-A., P.R.,
B Materials availability
Y.-T.Y., and M.S.O.; writing–original draft, E.E.B., J.A.T., M.J.G.-B., F.S.A.,
B Data and code availability
and J.M.G.-A.; writing–review & editing, E.E.B., J.A.T., M.D.W., P.E.,
d EXPERIMENTAL MODEL AND SUBJECT DETAILS M.J.G.-B., F.S.-A., P.R., J.M.G.-A., M.S.O., J.G.S., and Y.-T.Y.; resources,
B Cell culture J.A.T., J.M.G.-A., M.D.W., and J.G.S.; supervision, J.A.T., J.M.G.-A.,
B Human ileal enteroid cells M.D.W., and J.G.S.
B Bacterial strains used in this study
d METHOD DETAILS DECLARATIONS OF INTEREST
B Bacterial growth conditions and infections
The authors declare no competing interests.
B Flow cytometry of MDCK epithelial cells infected with
L.m. Received: June 15, 2020
B MDCK cell transfection with siRNA Revised: November 2, 2020
B RT-PCR Accepted: January 20, 2021
B Fabrication of polyacrylamide hydrogels and traction Published: February 22, 2021
force microscopy (TFM)
B Atomic Force Microscopy (AFM) for determination of REFERENCES
cell stiffness
Abaitua, F., Zia, F.R., Hollinshead, M., and O’Hare, P. (2013). Polarized cell
B RNA isolation and RNA sequencing
migration during cell-to-cell transmission of herpes simplex virus in human
B Principal component analysis (PCA) and mRNA func- skin keratinocytes. J. Virol. 87, 7921–7932.
tion enrichment analysis Aigouy, B., Umetsu, D., and Eaton, S. (2016). Segmentation and quantitative
B Immunofluorescence microscopy analysis of epithelial tissues. Methods Mol. Biol. 1478, 227–239.
B Multiplexed magnetic Luminex cytokine immunoassay Aikin, T.J., Peterson, A.F., Pokrass, M.J., Clark, H.R., and Regot, S. (2019).
and data analysis Collective MAPK signaling dynamics coordinates epithelial homeostasis.
B Western blotting bioRxiv https://www.biorxiv.org/content/10.1101/826917v1#::text=ABST
B Measurement of reactive oxygen species (ROS) pro- RACT,decisions%20while%20maintaining%20barrier%20function.
duction &text=Overall%2C%20we%20show%20that%20the,behaviors%20to%
20maintain%20tissue%20homeostasis.
B Nuclear segmentation, tracking and characterization of
Apidianakis, Y., and Rahme, L.G. (2011). Drosophila melanogaster as a model
dynamics of motion
for human intestinal infection and pathology. Dis. Model. Mech. 4, 21–30.
B Cell segmentation and extraction of cell shape charac-
Bastounis, E., Meili, R., Álvarez-González, B., Francois, J., del Álamo, J.C.,
teristics
Firtel, R.A., and Lasheras, J.C. (2014). Both contractile axial and lateral traction
B Characterization of mound volume and infection
force dynamics drive amoeboid cell motility. J. Cell Biol. 204, 1045–1061.
focus area
Bastounis, E.E., Ortega, F.E., Serrano, R., and Theriot, J.A. (2018). A multi-
B Live/dead staining, TUNEL and BrdU assays well format polyacrylamide-based assay for studying the effect of extracel-
B Laser ablation experiments lular matrix stiffness on the bacterial infection of adherent cells. J. Vis.
B Computational modeling of cell monolayers during Exp. 137, 57361.
infection €schner, G., Mu
Beerli, C., Yakimovich, A., Kilcher, S., Reynoso, G.V., Fla €ller,
d QUANTIFICATION AND STATISTICAL ANALYSIS D.J., Hickman, H.D., and Mercer, J. (2019). Vaccinia virus hijacks EGFR
Buchon, N., Broderick, N.A., Kuraishi, T., and Lemaitre, B. (2010). Herb, M., Gluschko, A., Wiegmann, K., Farid, A., Wolf, A., Utermöhlen, O.,
DrosophilaEGFR pathway coordinates stem cell proliferation and gut remod- Krut, O., Krönke, M., and Schramm, M. (2019). Mitochondrial reactive oxygen
eling following infection. BMC Biol. 8, 152. species enable proinflammatory signaling through disulfide linkage of NEMO.
Sci. Signal. 12, eaar5926.
Co, J.Y., Margalef-Català, M., Li, X., Mah, A.T., Kuo, C.J., Monack, D.M., and
Amieva, M.R. (2019). Controlling epithelial polarity: a human enteroid model for Hoesel, B., and Schmid, J.A. (2013). The complexity of NF-kB signaling in
host-pathogen interactions. Cell Rep. 26, 2509–2520.e4. inflammation and cancer. Mol. Cancer 12, 86.
Crane, A.M., and Bhattacharya, S.K. (2013). The use of bromodeoxyuridine Hogan, C., Dupré-Crochet, S., Norman, M., Kajita, M., Zimmermann, C.,
incorporation assays to assess corneal stem cell proliferation. Methods Mol. Pelling, A.E., Piddini, E., Baena-López, L.A., Vincent, J.P., Itoh, Y., et al.
Biol. 1014, 65–70. (2009). Characterization of the interface between normal and transformed
epithelial cells. Nat. Cell Biol. 11, 460–467.
Darzynkiewicz, Z., and Zhao, H. (2011). Detection of DNA strand breaks in
apoptotic cells by flow- and image-cytometry. Methods Mol. Biol. Holly, M.K., and Smith, J.G. (2018). Adenovirus infection of human enteroids
682, 91–101. reveals interferon sensitivity and preferential infection of goblet cells. J. Virol.
92, e00250–00218.
del Álamo, J.C., Meili, R., Alonso-Latorre, B., Rodrı́guez-Rodrı́guez, J.,
Aliseda, A., Firtel, R.A., and Lasheras, J.C. (2007). Spatio-temporal analysis Jiang, T., Grabiner, B., Zhu, Y., Jiang, C., Li, H., You, Y., Lang, J., Hung, M.C.,
of eukaryotic cell motility by improved force cytometry. Proc. Natl. Acad. and Lin, X. (2011). CARMA3 is crucial for EGFR-Induced activation of NF-kB
Sci. USA 104, 13343–13348. and tumor progression. Cancer Res. 71, 2183–2192.
Dev, A., Iyer, S., Razani, B., and Cheng, G. (2011). NF-kB and innate immunity. Joshi, S.D., von Dassow, M., and Davidson, L.A. (2010). Experimental control
Curr. Top. Microbiol. Immunol. 349, 115–143. of excitable embryonic tissues: three stimuli induce rapid epithelial contrac-
tion. Exp. Cell Res. 316, 103–114.
Dolowschiak, T., Chassin, C., Ben Mkaddem, S., Fuchs, T.M., Weiss, S.,
Vandewalle, A., and Hornef, M.W. (2010). Potentiation of epithelial innate Kang, H.B., Kim, Y.E., Kwon, H.J., Sok, D.E., and Lee, Y. (2007). Enhancement
host responses by intercellular communication. PLoS Pathog. 6, of NF-kappaB expression and activity upon differentiation of human embry-
e1001194. onic stem cell line SNUhES3. Stem Cells Dev. 16, 615–623.
Drolia, R., Tenguria, S., Durkes, A.C., Turner, J.R., and Bhunia, A.K. (2018). Kasper, C.A., Sorg, I., Schmutz, C., Tschon, T., Wischnewski, H., Kim, M.L.,
Listeria adhesion protein induces intestinal epithelial barrier dysfunction for and Arrieumerlou, C. (2010). Cell-cell propagation of NF-kB transcription fac-
bacterial translocation. Cell Host Microbe 23, 470–484.e7. tor and MAP kinase activation amplifies innate immunity against bacterial
infection. Immunity 33, 804–816.
Edelstein, A.D., Tsuchida, M.A., Amodaj, N., Pinkard, H., Vale, R.D., and
Stuurman, N. (2014). Advanced methods of microscope control using Kiehart, D.P., Galbraith, C.G., Edwards, K.A., Rickoll, W.L., and Montague,
mManager software. J. Biol. Methods 1, e10. R.A. (2000). Multiple forces contribute to cell sheet morphogenesis for dorsal
closure in Drosophila. J. Cell Biol. 149, 471–490.
Engström, P., Burke, T.P., Mitchell, G., Ingabire, N., Mark, K.G., Golovkine, G.,
Iavarone, A.T., Rape, M., Cox, J.S., and Welch, M.D. (2019). Evasion of auto- Kim, D., Langmead, B., and Salzberg, S.L. (2015). HISAT: a fast spliced aligner
phagy mediated by Rickettsia surface protein OmpB is critical for virulence. with low memory requirements. Nat. Methods 12, 357–360.
Nat. Microbiol. 4, 2538–2551. Kim, M., Ogawa, M., Fujita, Y., Yoshikawa, Y., Nagai, T., Koyama, T., Nagai, S.,
Escribano, J., Chen, M.B., Moeendarbary, E., Cao, X., Shenoy, V., Garcia- €ssler, R., and Sasakawa, C. (2009). Bacteria hijack integrin-linked
Lange, A., Fa
Aznar, J.M., Kamm, R.D., and Spill, F. (2019). Balance of mechanical forces kinase to stabilize focal adhesions and block cell detachment. Nature 459,
drives endothelial gap formation and may facilitate cancer and immune-cell 578–582.
extravasation. PLoS Comp. Biol. 15, e1006395. Knodler, L.A., Crowley, S.M., Sham, H.P., Yang, H., Wrande, M., Ma, C.,
Escribano, J., Sunyer, R., Sánchez, M.T., Trepat, X., Roca-Cusachs, P., and Ernst, R.K., Steele-Mortimer, O., Celli, J., and Vallance, B.A. (2014).
Garcı́a-Aznar, J.M. (2018). A hybrid computational model for collective cell Noncanonical inflammasome activation of caspase-4/caspase-11 mediates
durotaxis. Biomech. Model. Mechanobiol. 17, 1037–1052. epithelial defenses against enteric bacterial pathogens. Cell Host Microbe
16, 249–256.
Faralla, C., Bastounis, E.E., Ortega, F.E., Light, S.H., Rizzuto, G., Gao, L.,
Marciano, D.K., Nocadello, S., Anderson, W.F., Robbins, J.R., et al. (2018). Knodler, L.A., Vallance, B.A., Celli, J., Winfree, S., Hansen, B., Montero, M.,
Listeria monocytogenes InlP interacts with afadin and facilitates basement and Steele-Mortimer, O. (2010). Dissemination of invasive Salmonella via bac-
membrane crossing. PLoS Pathog. 14, e1007094. terial-induced extrusion of mucosal epithelia. Proc. Natl. Acad. Sci. USA 107,
Fernandez-Gonzalez, R., Simoes, Sde M., Röper, J.C., Eaton, S., and Zallen, 17733–17738.
J.A. (2009). Myosin II dynamics are regulated by tension in intercalating cells. Kocgozlu, L., Saw, T.B., Le, A.P., Yow, I., Shagirov, M., Wong, E., Mège, R.M.,
Dev. Cell 17, 736–743. Lim, C.T., Toyama, Y., and Ladoux, B. (2016). Epithelial cell packing induces
Gouin, E., Adib-Conquy, M., Balestrino, D., Nahori, M.A., Villiers, V., Colland, distinct modes of cell extrusions. Curr. Biol. 26, 2942–2950.
F., Dramsi, S., Dussurget, O., and Cossart, P. (2010). The Listeria monocyto- Kong, W., Loison, O., Chavadimane Shivakumar, P., Chan, E.H., Saadaoui,
genes InlC protein interferes with innate immune responses by targeting the M., Collinet, C., Lenne, P.F., and Clément, R. (2019). Experimental valida-
I{kappa}B kinase subunit IKK{alpha}. Proc. Natl. Acad. Sci. USA 107, tion of force inference in epithelia from cell to tissue scale. Sci. Rep.
17333–17338. 9, 14647.
Gouin, E., Balestrino, D., Rasid, O., Nahori, M.-A., Villiers, V., Impens, F., Kuipers, D., Mehonic, A., Kajita, M., Peter, L., Fujita, Y., Duke, T., Charras, G.,
Volant, S., Vogl, T., Jacob, Y., Dussurget, O., and Cossart, P. (2019). and Gale, J.E. (2014). Epithelial repair is a two-stage process driven first by
Ubiquitination of Listeria virulence factor InlC contributes to the host response dying cells and then by their neighbours. J. Cell Sci. 127, 1229–1241.
to infection. mBio 10, e02778–19. Lamason, R.L., Bastounis, E., Kafai, N.M., Serrano, R., Del Álamo, J.C.,
Grieve, A.G., and Rabouille, C. (2014). Extracellular cleavage of E-cadherin Theriot, J.A., and Welch, M.D. (2016). Rickettsia Sca4 reduces vinculin-medi-
promotes epithelial cell extrusion. J. Cell Sci. 127, 3331–3346. ated intercellular tension to promote spread. Cell 167, 670–683.e10.
Gudipaty, S.A., and Rosenblatt, J. (2017). Epithelial cell extrusion: pathways Lamason, R.L., and Welch, M.D. (2017). Actin-based motility and cell-to-cell
and pathologies. Semin. Cell Dev. Biol. 67, 132–140. spread of bacterial pathogens. Curr. Opin. Microbiol. 35, 48–57.
STAR+METHODS
Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
L. monocytogenes: L.m.-DinlC F. Ortega (Ortega et al., 2017) N/A
ActAp::mTagRFP
L. monocytogenes: L.m.-DinlP A. Bakardjiev (Faralla et al., 2018) N/A
L. monocytogenes: L.m.-LLOG486D M. Rengarajan (Rengarajan et al., 2016) N/A
ActAp::mTagRFP
R. parkeri Portsmouth strain Chris Paddock N/A
R. parkeri: R.p.- ompBSTOP::tn P. Engstrom (Engström et al., 2019) N/A
L. monocytogenes: L.m.-Dhly M. Rengarajan (Rengarajan et al., 2016) N/A
Oligonucleotides
siRNA targeting sequence for CDH1: GE Healthcare Dharmacon, Inc. CTM-521910 and CTM-521911
Custom CDH1 duplex
siRNA targeting sequence for SNAi1: GE Healthcare Dharmacon, Inc. CTM-544153 and CTM-544153
Custom SNAI1 duplex
Forward Primer for CDH1 (forward: This paper N/A
5’ AGACCCAGTAACTAACGACGG 3’)
Reverse Primer for CDH1 (reverse: This paper N/A
5’ ACACCAAAGTCTTCAGGGATT 3’)
Forward Primer for SNAI1 (forward: This paper N/A
5’ CCCCCATTTGGCTGTGTTG 3’)
Reverse Primer for SNAI1 (reverse: This paper N/A
5’ ATCAGTCTGTCGGCTTTTATCCT 3’)
Forward Primer for b-actin (forward: This paper N/A
5’ CCCAGCACAATGAAGATCAAGAT 3’)
Reverse Primer for b-actin (reverse: This paper N/A
5’ CAAGAAAGGGTGTAACGCAACT 3’)
Forward Primer for SNAI2 (forward: This paper N/A
5’ GAAGCATTTCAACGCCTCC 3’)
Reverse Primer for SNAI2 (reverse: This paper N/A
5’ ACTCACTCGCCCCAAGGAT 3’)
Forward Primer for NFKBIA (forward: This paper N/A
5’ CACATTCCCTAGCCAGAAACATT 3’)
Reverse Primer for NFKBIA (reverse: This paper N/A
5’ TACACCTGGTTGTCACGCATC 3’)
Forward Primer for ICAM1 (forward: This paper N/A
5’ ACCGAGGGTTGGGATTGTT 3’)
Reverse Primer for ICAM1 (reverse: This paper N/A
5’ GTCTCTGGCAGGACAAAGGTT 3’)
Forward Primer for CXCL8 (forward: This paper N/A
5’ GAAAACTCAGAAATCATTGTAAAGC 3’)
Reverse Primer for CXCL8 (reverse: This paper N/A
5’ ATCTTGTTTCTCAGCCTTCTTTAG 3’)
Software and Algorithms
ImageJ Schneider et al., 2012 https://imagej.nih.gov/ij/
MicroManager Open Imaging https://www.micro-manager.org/
MATLAB MathWorks http://www.mathworks.com/products/
matlab/?requestedDomain=www.
mathworks.com
GraphPad Prism v6 GraphPad http://www.graphpad.com/scientific-
software/prism/
Hisat 2 (Kim et al., 2015) https://ccb.jhu.edu/software/hisat2/
index.shtml)
Cutadapt (Martin, 2011) https://cutadapt.readthedocs.io/en/stable/
(Continued on next page)
Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
R package GAGE (Luo et al., 2009) https://bioconductor.org/packages/
release/bioc/html/gage.html
R package ‘‘Pathview’’ (Luo et al., 2017) https://www.bioconductor.org/packages/
release/bioc/html/pathview.html
ABAQUS Dassault systèmes https://www.3ds.com/products-services/
simulia/products/abaqus/
Imaris Bitplane https://imaris.oxinst.com/
RESOURCE AVAILABILITY
Lead contact
Further information and requests for reagents may be directed to and will be fulfilled by the Lead Contact Julie Theriot jtheriot@uw.
edu (J.A.T.).
Materials availability
Materials developed in this study are available on request to the corresponding author.
Cell culture
Type II MDCK cells (generous gift from the Bakardjiev lab, University of California, San Francisco) were cultured in high glucose
DMEM medium (Thermofisher; 11965092) containing 4.5 g/L glucose and supplemented with 10% fetal bovine serum (GemBio;
900-108) (Ortega et al., 2017). Passages were between P10-P40. MDCK cells expressing E-cadherin-RFP were a generous gift
from the Nelson lab, Stanford University (Perez et al., 2008). a-catenin knockout (aEcat KO) MDCK cells were also a generous gift
from the Nelson lab, Stanford University (Ortega et al., 2017). Human epithelial carcinoma A431D cells were a generous gift from
Cara Gottardi, Northwestern University and were grown in DMEM with high glucose (4.5 g/L) in the presence of 10% FBS and
1% penicillin-streptomycin. Transduced cell lines that express full length E-cadherin were cultured in media supplemented with
800 mg/mL geneticin and were generated as described previously (Ortega et al., 2017).
(Species: L.m. 1043S, Genotype/Description: DinlC ActAp::mTagRFP), DinlP (Species: L.m. 1043S, Genotype/Description: DinlP,
obtained from the Bakardjiev lab) (Faralla et al., 2018), JAT983 (Species: L.m. 1043S, Genotype/Description: LLOG486D ActAp::
mTagRFP) (Rengarajan et al., 2016), Dhly (Species: L.m. 1043S, Genotype/Description: Dhly) (Rengarajan et al., 2016). Note that
we used the LLOG486D L.m. to test the role of LLO in induction of mounds and in activating signaling pathways that affect mechano-
transduction., LLOG486D L.m. is a strain that carries a point mutation in the gene encoding LLO and thus exhibits 100x less hemolytic
activity than the WT protein, permitting a moderate degree of bacterial proliferation and intercellular spread (unlike strains completely
lacking LLO which are unable to proliferate in host cells) (Rengarajan et al., 2016).
WT Rickettia parkeri (R.p.) strain Portsmouth was originally obtained from Dr. Chris Paddock (Center for Disease Control and Pre-
vention, NCBI accession no. NC_017044.1), and the ompB- mutant (ompBSTOP::tn) was isolated and validated in a recent study (Eng-
ström et al., 2019). R.p. propagation and purifications were from five T175 flasks of Vero cells growing in DMEM (Gibco, 11965) with
high glucose (4.5 g l-1) and 2% FBS (Benchmark) that after 5-6 days of infection were harvested in the media using a cell scraper.
Infected cells were then centrifuged 12000g for 15 min at 4 C, resuspended in cold K-36 buffer (0.05 M KH2PO4, 0.05 M
K2HPO4, pH 7, 100 mM KCl and 15 mM NaCl), and bacteria were then released using a dounce-homogenizer. The homogenate
was centrifuged at 200g for 5 min at 4 C and the supernatant was then overlaid onto cold 30% v/v MD-76R (Mallinckrodt Inc.,
1317-07) diluted in K-36, and centrifuged at 58300g for 20 min at 4 C in an SW-28 swinging-bucket rotor. The bacterial pellet
was resuspended in cold 1x BHI broth, aliquoted and frozen at -80 C.
METHOD DETAILS
EDTA. Trypsin-EDTA solutions in each well were then pipetted up and down 6 times to ensure single cell suspensions and 200 mL of
complete media was added to inactivate trypsin in each well. Suspensions were transferred into 35-mm cell strainers, (Falcon,
352235) and spun through at 500 x g followed by fixation in 1% paraformaldehyde. Samples were then washed once in PBS and
stored in PBS with 1% BSA. Flow cytometry analysis was performed on a BD FACS Canto RUO analyzer (University of Washington
Cell Analysis Facility). 10,000-20,000 cells were analyzed per each replicate. To ensure analysis of single cells, the bulk of the dis-
tribution of cell counts was gated using the forward versus side scatter plot. This gating strategy ensures that single cells are analyzed
and debris or cell doublets or triplets are eliminated from the analysis. A second gating step was then performed to exclude cells that
autofluorescence by measuring the fluorescence of control uninfected cells and gating the population of infected cells to exclude
autofluorescence.
RT-PCR
To confirm the knockdowns, MDCK cells treated with control (non-targeting) or experimental siRNA 72 h after transfection (approx-
imate cell concentration was 1.6x105 cells/well) were harvested and lyzed using the QIAshredder Kit (Qiagen, 79656). mRNA was
harvested using the RNeasy Plus Micro Kit (Qiagen, 74004) and eluted in 30 mL RNAase free water RNA concentrations were
measured spectrophotometrically (NanoDrop) and were comparable between conditions. cDNA was prepared using the Superscript
III First-strand Synthesis SuperMix (Invitrogen, 18080085). RT-qPCR was performed using the SYBR qPCR Master mix by Arraystar
Inc. Genes of interest were amplified using the appropriate primers: for CDH1 forward:5’ AGACCCAGTAACTAACGACGG 3’ and
reverse:5’ ACACCAAAGTCTTCAGGGATT 3’; for SNAI1 forward:5’ CCCCCATTTGGCTGTGTTG 3’ and reverse:5’ AT-
CAGTCTGTCGGCTTTTATCCT 3’; for b-actin forward:5’ CCCAGCACAATGAAGATCAAGAT 3’ and reverse:5’ CAAGAAAGGGTG-
TAACGCAACT 3’. Briefly the steps followed were: (1) perform RT-qPCR for each target gene and the housekeeping gene GAPDH;
(2) determine gene concentration using the standard curve with Rotor-Gene Real-Time Analysis Software 6.0; (3) calculate relative
amount of the target gene relative to GAPDH.
Similar procedure was followed to confirm expression of NF-kB target genes. MDCK cells were infected or not for 24 h with L.m..
MDCK cells were also infected or not or for 72 h with WT R.p. or the ompB- mutant. For each condition 4 replicates were prepared.
Cells were harvested and lyzed, and their mRNA was harvested as described above. RT-qPCR was performed as above. NF-kB
target genes tested were NFKBIA, CXCL8, ICAM1, SNAI1 and SNAI2. Genes of interest were amplified using the appropriate primers:
for NFKBIA forward:5’ CACATTCCCTAGCCAGAAACATT 3’ and reverse:5’ TACACCTGGTTGTCACGCATC 3’; for CXCL8 forward:5’
GAAAACTCAGAAATCATTGTAAAGC 3’ and reverse:5’ ATCTTGTTTCTCAGCCTTCTTTAG 3’; for ICAM1 forward:5’ AC-
CGAGGGTTGGGATTGTT 3’ and reverse:5’ GTCTCTGGCAGGACAAAGGTT 3’; for SNAI1 forward:5’ CCCCCATTTGGCTGTGTTG
3’ and reverse:5’ ATCAGTCTGTCGGCTTTTATCCT 3’; for SNAI2 forward:5’ GAAGCATTTCAACGCCTCC 3’ and reverse:5’ ACT-
CACTCGCCCCAAGGAT 3’; for b-actin forward:5’ CCCAGCACAATGAAGATCAAGAT 3’ and reverse:5’ CAAGAAAGGGTGTAACG-
CAACT 3’.
dimethyl sulfoxide (DMSO) and 50 mM HEPES, pH 7.5, on the upper surface of the hydrogels and exposing them to UV light for
10 min. Hydrogels were washed with 50 mM HEPES at pH 7.5 and were coated with 200 mL of 0.25 mg/ml rat tail collagen I
(Sigma-Aldrich; C3867) in 50 mM HEPES at pH 7.5 overnight at room temperature. Next morning, the collagen coated surfaces
were washed with HEPES and gels were stored in HEPES.
TFM was performed as previously described (del Álamo et al., 2007; Lamason et al., 2016). Briefly, in TFM, cells actively pull on their
ECM depending on how well their focal adhesions are organized and connected to the underlying cytoskeleton, and cellular force
generation can be inferred from displacement of fluorescent tracer particles embedded in the deformable ECM (Bastounis et al.,
2014; del Álamo et al., 2007). Prior to seeding cells hydrogels were equilibrated with cell media for 30 min at 37 C. Cells were
then seeded to a concentration of 2x105 cells per well directly onto the hydrogels 24 h prior to infection. Cells are then either infected
or not with low dosage of L.m. and imaging started 4 hpi. Multi-channel time-lapse sequences of fluorescence (to image the beads
within the upper portion of the hydrogels) and phase contrast images (to image the cells) were acquired using an inverted Nikon
Eclipse Ti2 with an EMCCD camera (Andor Technologies) using a 40X 0.60NA Plan Fluor air objective or a 20x 0.75 NA Plan Apo
air objective and MicroManager software package (Edelstein et al., 2014). The microscope was surrounded by a box type incubator
(Haison) maintained at 37 C and 5% CO2. Images were acquired every 10 min for approximately 8 h. Subsequently, at each time
interval we measured the 2D deformation of the substrate at each point using an image correlation technique similar to particle image
velocimetry. We calculated the local deformation vector by performing image correlation between each image and an undeformed
reference image which we acquired by adding 10% SDS at the end of each recording to detach the cells from the hydrogels. We used
interrogation windows of 32 x 8 pixels (window size x overlap). Calculations of the two-dimensional traction stresses that cell mono-
layers exert on the hydrogel are described elsewhere (Bastounis et al., 2014; Lamason et al., 2016).
flow cell at 8 pM concentration, and amplified in situ using TruSeq SR Cluster Kit v3-cBot-HS (#GD-401-3001, Illumina). Sequencing
was carried out using the Illumina X- ten/NovaSeq according to the manufacturer’s instructions. Sequencing was carried out by
running 150 cycles.
Raw sequencing data generated from Illumina X-ten/NovaSeq that pass the Illumina chastity filter were used for following analysis.
Trimmed reads (trimmed 5’, 3’-adaptor bases) were aligned to the reference genome (CanFam3). Based on alignment statistical
analysis (mapp.i.ng ratio, rRNA/mtRNA content, fragment sequence bias), we determined whether the results can be used for
subsequent data analysis. To examine the sequencing quality, the quality score plot of each sample was plotted. Quality score Q
is logarithmically related to the base calling error probability (P): Q = 10log10(P). For example, Q30 means the incorrect base calling
probability to be 0.001 or 99.9% base calling accuracy. After quality control, the fragments were 5’, 3’-adaptor trimmed and filtered
% 20 bp reads with Cutadapt software. The trimmed reads were aligned to the reference genome with Hisat 2 software. In a typical
experiment, it is possible to align 40 90% of the fragments to the reference genome. However, this percentage depends on multiple
factors, including sample quality, library quality and sequencing quality. Sequencing reads are classified into the following classes: (1)
Mapped : reads aligned to the reference genome (including mRNA, pre-mRNA, poly-A tailed lncRNA and pri-miRNA); (2) mtRNA and
rRNA: fragments aligned to rRNA, mtRNA; and (3) Unmapped: Reads that are not aligned.
Differentially expressed genes and differentially expressed transcripts are calculated. The novel genes and transcripts are also pre-
dicted. The expression level (fragments per kilobase of transcript per million mapped reads, FPKM) of known genes and transcripts
were calculated using ballgown through the transcript abundances estimated with StringTie. The number of identified genes
and transcripts per group was calculated based on the mean of FPKM in group R 0.5. FPKM is calculated with the formula:
FPMK = CL3N 3106
, where C is the number of fragments that map to a certain gene/ transcript, L is the length of the gene/transcript
in Kb and N is the fragments number that map to all genes/transcripts. Differentially expressed gene and transcript analyses were
performed with R package ballgown. Fold change (cutoff 1.5), p-value (% 0.05) and FPKM (R 0.5 mean in one group) were used
for filtering differentially expressed genes and transcripts.
Immunofluorescence microscopy
Uninfected or L.m.-infected MDCK cells residing on glass substrates were incubated with 1 mg/mL Hoechst to stain the cells’ nuclei
for 10 min at 37 C prior to fixation. The following steps were carried out at room temperature. Cells were washed with PBS and fixed
with 4% EM grade formaldehyde in PBS for 10 min. Following a wash with PBS, samples were permeabilized for 5 min in 0.2% Triton
X-100 in PBS and washed again with PBS. Samples were then blocked for 30 min with 5% BSA in PBS and then incubated with pri-
mary antibodies (anti-vimentin: Abcam, ab8069; anti-pan cytokeratin: ThermoFisher, 53-9003-80; anti-tubulin: Abcam, ab52866;
anti-NF-kB: Abcam, ab190589; anti-E-cadherin: Cell Signaling, 3195; anti-ZO-1: Thermofisher, ZO1-1A12; anti-R.p., I7205 (gift
from Dr Ted Hackstadt, NIH Rocky Mountain Laboratories) diluted 1:100 in PBS containing 2% BSA for 1 h for all cases other
than anti-R.p. where dilution was 1:1000. Samples were washed in PBS three times and then incubated with the appropriate sec-
ondary fluorescent antibodies (Invitrogen) diluted 1:250 in PBS containing 2% BSA for 1 h. For actin staining we used 0.2 mM Alexa-
Fluor488 phalloidin (Thermo Fisher, A12379). Samples were washed three times in PBS and stored in 1 mL PBS for imaging. N > 8
mounds were analyzed per condition (N> 1200 cells). For epifluorescence imaging, we used an inverted Nikon Eclipse Ti2 with an
EMCCD camera (Andor Technologies) and a 40x 0.60NA Plan Fluor air or a 20x 0.75NA Plan Apo air objective and MicroManager
software. For confocal imaging we used a Yokogawa W1 Spinning Disk Confocal with Borealis upgrade on a Leica DMi6 inverted
microscope with a 50um Disk pattern, a 60x 1.4NA Plan Apo oil objective and MicroManager software.
well. The plate was again sealed and covered with foil and incubated with agitation for 1 h. After incubation, 25 mL Streptavidin-
Phycoerythrin was added to each well containing the detection antibodies. (Plate was not washed after incubation with detection
antibodies as per manufacturer’s protocol). The plate was sealed and covered with foil and incubated with agitation for 30 min.
Then, well contents were gently aspirated out, and the plate was washed two times. Finally, 150 mL of sheath fluid was added to
all wells, and the beads were resuspended on a plate shaker for 5 min at room temperature. The plate was run on Bio-Plex 200
flow cytometer. Data analysis was performed using the Bio-Plex Manager 5.0. We used a 5-parameter log fit curve against the signal
intensities for the analytes present in the Canine Cytokine Magnetic bead Panel (GM-CSF, IFN-g, IL-2, IL-6, IL-7, IL-8, IL-15, IP-10,
KC-like, IL-10, IL-18, MCP-1, TNF-A) to generate a standard curve for the assay. Sample signal intensities were then back interpo-
lated to the standard curve to determine the analyte concentrations. The upper limit of quantitation (ULOQ) was determined as the
highest point at which the interpolated data of the standard curve are within 25% (±) of the expected recovery. ULOQ represents the
highest concentration of an analyte that can be accurately quantified. Similarly, the lower limit of quantitation (LLOQ) was determined
as the lowest point at which the interpolated data of the standard curve are within 25% (±) of the expected recovery. LLOQ represents
the lowest concentration of an analyte that can be accurately quantified.
Western blotting
To assess phosphorylation of the p65 subunit of NF-kB and expression levels of NF-kB, MDCK cells were seeded at a concentration
of 2 3 105 cells/well (24-well plates) for 24 h and then infected or not for 24 h with intracellular L.m. at a MOI200 bacteria/cell. Cells
in infected wells were treated with vehicle control or the NEMO binding peptide (50 mM) starting 4 hpi. Cells were then lysed with a
buffer containing 0.5M Tris-HCl, pH 7.4, 1.5M NaCl, 2.5% deoxycholic acid, 10% NP-40, 10mM EDTA and a protease inhibitor
mixture (phenylmethylsulfonyl fluoride [PMSF], leupeptin, aprotinin, and sodium orthovanadate). The total cell lysate was separated
by SDS–PAGE (10% running, 4% stacking) and transferred onto a nitrocellulose membrane (Immobilon P, 0.45-mm pore size). The
membrane was then incubated with the designated antibodies. Immunodetection was performed using the Western-Light chemilu-
minescent detection system (Applied Biosystems). Membrane blots from 76 kDa to 52 kDa were imaged for phospho-p65 and p65
(65 kDa) and 52 kDa to 33 kDa were imaged for the corresponding b-actin (42 kDa) (Figure S6F).
IMARIS software (Bitplane). The x, y and z positions of all identified objects and tracks were exported for import into MATLAB (Math-
Works) for further analysis of cellular speed, net/total distance travelled and coordination scores as described previously (Hayer et al.,
2016). Regarding coordination score calculation we considered a radius of 65 mm around each given cell for the calculation of the
pairwise velocity correlations (Hayer et al., 2016).
The correlation length shown in Figure S4J is calculated as previously documented (Tambe et al., 2011) and it is defined as in the
next equation:
P
j dðrj Þ$dðrj + RÞ
CðRÞ = P t;f
j dðrj Þ$dðrj Þ
where C is the correlation length for all vectors separated by a distance R, dðrj Þ is the modified displacement vector for a given po-
sition rj and the angle brackets signify an average over all directions and time. To avoid the dependence of the correlation value with
the absolute value of the displacement, we substrate the mean displacement of each frame (x) to the displacement vector (xðrj Þ),
dðrj Þ = xðrj Þ x. We fit the correlation over the radius using the function CðRÞ = expð R =R0 Þ and determine the correlation length
as R0 .
the cell-ECM adhesions are removed (stretching-based interactions). However, during cell protrusion cells are in contact with the
ablated area (compression-based contact).
To simulate how host cells behave when certain cells are able to form cell-cell adhesions while others not, and to better understand
in which scenarios an infection mound will be formed, we consider two cases. In the first one, all the infected cells and their immediate
surrounders are able to form cell-cell adhesions, but all the rest uninfected cells are not. In the second one, the infected cells and their
immediate surrounders are not able to form cell-cell adhesions, but uninfected cells far from the infection mound are able to. As in
previous simulations, when one edge of the cell cannot adhere to another cell’s edge, the cell-ECM interaction forces increase in
these edges of the cell. Thus, a cell without cell-cell adhesions in any direction would be an isolated cell with high cell-ECM traction
forces in all directions.
We implement the model into a commercial finite element model (FE-based) ABAQUS70. To simulate cells two meshes are over-
lapping sharing the nodes to represent the active and passive components of the cell. We discretise the model with tetrahedral and
hexahedral linear elements of average size 2 mm.
Statistical parameters and significance are reported in the Figures and the Figure Legends. Data are determined to be statistically
significant when p < 0.05 or p<0.01 by an unpaired Student’s T-Test (USTT), or Wilcoxon Rank Sum Test (WRST), where indicated.
As such, asterisk denotes statistical significance as compared to indicated controls. For statistical analysis of the kinematics and
shape morphometrics of large number of cells characterized in each experiment, we used violin plots considering all nuclei tracked
(Figures 1F, 1I, 1J, S4C, and S4J) or all cells characterized (Figures 2D, S2C, and S2D). In the violin plots dashed line represents the
median of the distribution and dotted lines the 25 and 75% quartiles considering data from all experiments. For each independent
experiment (replicate) the mean value was also calculated and indicated as a colored inverted triangle. Those means where then used
to calculate their average (horizontal bar) and standard deviation (vertical bars) and their p-value using an USTT (Lord et al., 2020). For
the rest of the data represented using barplots (e.g. relative infection mound volumes) midlines denote mean value and whiskers the
standard deviation (vertical bars). P-value was calculated using the non-parametric WRST and asterisk denotes p < 0.05. Statistical
analysis was performed in GraphPad PRISM 8.