You are on page 1of 25

Elevated inflammatory response in caveolin-1 deficient mice with P.

aeruginosa infection
is mediated by STAT3 and NF-B

Kefei Yuan*, Canhua Huang*¶, John Fox, Madeleine Gaid, Andrew Weaver, Guoping Li,
Brij B. Singh, Hongwei Gao# and Min Wu¶.

*The State Key Laboratory for Biotherapy, West China Hospital, Sichuan University,
Chengdu 610041, China; #Center for Experimental Therapeutics and Reperfusion Injury,
Department of Anesthesiology, Perioperative & Pain Medicine, Brigham and Women's
Hospital, Harvard Medical School; and Department of Biochemistry and Molecular Biology,
University of North Dakota, Grand Forks, ND58203-9037.
Running head: Cav-1 deficiency impacts P. aeruginosa infection

Address correspondence to: min.wu@med.und.edu, Tel: 701 777-4875, Fax: 701 777-2382;
or hcanhua@hotmail.com, Tel: +86-13258370346, Fax: 86-28-85164060

negative strategy demonstrated that


Caveolin-1 (Cav-1), an important STAT3 activation was essential for the
composition protein within the translocation of NF-B into the nucleus,
flask-shaped membrane invaginations confirming the observations from cav1
termed caveolae, may play a role in host KO mice. Collectively, our studies
defense against infections. However, the indicate that Cav-1 is critical for
phenotype in P. aeruginosa-infected cav1 inflammatory responses regulating the
knockout (KO) mice is still unresolved STAT3-NF-B pathway and thereby
and the mechanism involved is almost impacting P. aeruginosa infection.
entirely unknown. Using a respiratory
infection model, we confirmed a crucial
role played by Cav-1 in host defense P. aeruginosa accounts for 25% of
against this pathogen because Cav-1 KO Gram-negative bacteria isolated from
mice showed increased mortality, severe hospitals associated with high morbidity
lung injury, and systemic dissemination and mortality (1). P. aeruginosa frequently
as compared to wild-type (WT) infects immunocompromised individuals,
littermates. In addition, cav1 KO mice such as those affected by
exhibited elevated inflammatory ventilator-associated infection, severe burns,
cytokines (IL-6, TNF- and IL-12a), and cancer (2). More than 80% of cystic
decreased phagocytic ability of fibrosis patients suffer severe P. aeruginosa
macrophages, and increased superoxide infection (3). This bacterium becomes
release in the lung, liver, and kidney. We increasingly resistant to various antibiotics.
further studied relevant cellular signaling Further understanding the mechanism of
processes and found that STAT3 and host-pathogen interaction may result in
NF-B are markedly activated. Our data effective approaches to preventing this
revealed that the Cav-1-STAT3-NF-κB infection. Thus, P. aeruginosa has been a
axis is responsible for dysregulated focus of airway infectious diseases (1,4-6).
cytokine response, which is attributable Recent studies suggest that P.
to increased mortality and disease aeruginosa also invades the lung epithelial
progression. Moreover, down-regulating cells through lipid raft-mediated
Cav-1 in cell culture with a dominant endocytosis (7-9), which is a possible
  1
reason why this bacterium develops such kinases (JAKs), which in turn lead to
formidable resistance to antibiotics (10). phosphorylation of tyrosine residues on
The invasion process of P. aeruginosa may receptors (24). These receptors serve as
involve certain lipid raft-associated proteins, docking sites for STATs. Suppressors of
including the caveolin family of proteins (9). cytokine signaling (SOCSs) function as
Using caveolin 1 (cav1) KO mice, two negative regulators of STATs and operate by
recent studies investigated the role of Cav-1 binding and inhibiting JAKs or competing
during P. aeruginosa infection (11,12); with STATs for phosphotyrosine binding
however, their observations were very sites on cytokine receptors (25). Recently,
different. Thus, the role of Cav-1 in this STAT3 was found to interact with Cav-1
infection needs to be clearly characterized. and heat shock protein 90 in plasma
Caveolins are a family of integral membrane rafts during E. coli infection (26).
membrane proteins involved in caveolae Cav-1 is also related to a JAK2/STAT5
formation and receptor-dependant pathway since Cav-1 is homologous to the
endocytosis (13-15). Cav-1 and -2 are pseudosubstrate for SOCS. Thus, Cav-1
co-expressed in various cells, such as may down-regulate JAK/STAT pathways,
endothelial cells, airway epithelial cells, and modulating pro-inflammatory response.
type I pneumocytes. Cav-1 is the major Since Cav-1 was found to be associated
component of caveolae, flask-shaped with STAT3 in lipid rafts by a
plasma membrane invaginations. In the co-localization study, it is also possible that
absence of Cav-1, Cav-2 cannot reach the a Cav-1 cascade may impact the PI3K/Akt
plasma membrane and is degraded (16). pathway through molecular interactions,
Through the hetero-oligomeric complex which may serve as a regulator for STAT3
formed between Cav-1 and Cav-2, Cav-2 (27). Thus, Cav-1 may help maintain the
can translocate to lipid rafts of the plasma balance between host response and tissue
membrane. The lipid rafts, also known as damage that may be caused by
membrane microdomains, contain overproduction of inflammatory cytokines.
glycosphingolipids, cholesterol, and To better characterize the role of Cav-1
signaling/receptor proteins (17-19), and in P. aeruginosa infection, we sought to
play a crucial role in many cellular determine the pathogenic mechanism in
activities including airway bacterial cav1 KO mice. We found that cav1 KO
invasion. mice manifested more severe infection,
Signal transducers and activators of including increased bacterial burdens,
transcription (STATs) are SH2 domain inflammation, oxidative stress, and
containing transcription factors involved in susceptibility to infection. Our studies also
inflammatory response in carcinogenesis showed that the STAT3-NF-κB pathway is
and host defense (20-23). STATs are responsible for the dysregulated response
activated through receptors for cytokines during P. aeruginosa infection.
and hormones, and these receptors do not
contain any intrinsic enzymatic activity, Experimental procedures
thus they depend on tyrosine kinases that Mice-cav1 KO and WT control mice
can interact with the intracellular domain of (B6129SF2/J) were obtained from The
the receptor. STAT signaling is initiated by Jackson Laboratory (Bar Harbor, ME) (28).
phosphorylating and activating Janus Mice were housed and bred in the animal

  2
facility at University of North Dakota bacteria-cell ratio for indicated time points.
(UND), and the animal experiments were Cell transfection-MLE-12 cells were
performed in accordance to the institutional transfected with yellow fluorescent protein
animal care and use committee guidelines. (YFP)-Cav-1, YFP-Cav-1Δ51-169
We anesthetized mice with 45 mg/kg dominant negative (DN) plasmids using
ketamine, and intranasally instilled 0.5×107 LipofectAmine 2000 reagent (Invitrogen
(PAK, 6 mice/group) to 1×107 (PAO1) Life Technologies, Carlsbad, CA) in
colony-forming units (CFUs) of P. serum-free RPMI 1640 medium following
aeruginosa and mice were killed when they the manufacturer’s instruction. The cells
were moribund (29). After were lysed after 24 h transient expression
bronchoalveolar lavage (BAL), the trachea (32).
and lung were excised for homogenization Inflammatory cytokine profiling-Cytokine
or fixed in 10% formalin. concentrations were measured by ELISA kit
Cells-Mouse AM cells were isolated by (eBioscience company, San Diego, CA) in
BAL as described (30). AM cells were samples of cell culture medium, BAL fluid
grown in RPMI 1640 medium and lung homogenates collected at indicated
supplemented with 10% newborn calf times after infection. The MLE-12 cells
serum and penicillin/streptomycin were treated as described above. Culture
antibiotics in 5% CO2 incubator. MLE-12 medium was collected after infection. For
cells were obtained from ATCC and BAL fluid, the trachea was surgically
maintained following the manufacturer’s exposed and cannulated, lungs were
instructions. lavaged 5 times with 1.0 ml volume of
Bacteria strains-P. aeruginosa strain PAO1 lavage fluid, the lavageates were pooled,
wild-type (WT) was a gift from S. Lory and cells were removed by centrifugation.
(Harvard Medical School, Boston, MA). For lung homogenates, excised lungs were
PAK and PAO1-GFP were obtained from G. ground in 500 μl PBS. 96-well plates
Pier (Channing Laboratory, Harvard (Corning Costar 9018) were coated with
Medical School, Boston, MA) (31). 100 μl/well of capture antibody in coating
Infection experiments-Bacteria were grown buffer and incubated overnight at 4°C (33).
overnight in Luria-Bertani (LB) broth at 100ul aliquots of serum samples were
37°C with vigorous shaking. The next day, added to the coated microtiter wells. The
the bacteria were pelleted by centrifugation cytokine concentrations were determined
at 5,000 × g and resuspended in 10 ml of with corresponding detection
fresh LB broth and allowed to grow until HRP-conjugated antibodies. The values
the mid-logarithmic phase. OD 600 nm was were read at 450 nm and analyzed.
measured, density was adjusted to ~0.25 Western blotting-Mouse monoclonal Abs
OD (0.1 OD = 1 × 108 cells/ml). Cells were against Cav-1, phospho-STAT3, IL-6,
washed once with PBS after overnight NF-κB, and rabbit polyclonal Ab against
culture in serum containing medium and phospho-NF-κB and goat polyclonal Abs
changed to serum-free and antibiotic-free against TNF-α, SOCS3 were from Santa
medium immediately before infection (29). Cruz Biotechnology. Rabbit monoclonal Ab
Except dose-determination assays, cells against glyceraldehydes-3-phosphate
were infected by P. aeruginosa in dehydrogenase (GAPDH) was from Cell
multiplicity of infection (MOI) of 10:1 Signaling Technology. The samples derived

  3
from cells and lung homogenates were grown either on coverslips in a 24-well
lysed and quantitated. The lysates were plate or in glass-bottom dishes (MatTek,
boiled for 5 min, and protease inhibitor Ashland, MA). For immunostaining, the
cocktail added. The supernatants were cells were fixed in 3.7% paraformaldehyde,
collected, and an equal amount of each permeabilized with 0.2% Triton X-100 in
sample was loaded onto 10% PBS, and non-specific binding site was
SDS-polyacrylamide mini-gels and blocked with blocking buffer for 30 min.
electrophoresed to resolve proteins. The Cells were incubated with primary Abs at
proteins were then transferred to 1/500 dilution in blocking buffer for 1 h and
polyvinylidine difluoride membranes washed three times with wash buffer. After
(Pierce Biotechnology, Rockford, IL) and incubation with appropriate
blocked 2 h at room temperature using 5% fluorophore-conjugated secondary Abs, the
non-fat milk blocking buffer. Membranes coverslips were mounted on slides with
were incubated overnight at 4°C with Vecta-shield mounting medium (36). DAPI
appropriate first antibodies diluted at (Sigma-Aldrich, St. Louis, MO) was used to
1:1,000 in 5% bovine serum albumin (BSA) stain the nucleus. The images were captured
western antibody buffer. After washing by LSM 510 Meta confocal microscope
three times with washing solution, the (Carl Zeiss MicroImaging, Thornwood,
antigen-antibody complexes were incubated NY), and processed using the software
for 45 min at room-temperature with provided by the manufacturer.
horseradish peroxidase-conjugated Luciferase Assay-Transient transfections
secondary antibody (Rockland were performed with 2×105 cells plated in
Immunochemicals, Gilbertsville, PA) 6-well plates by using 2 μg of DNA and 3
diluted 1:2,000 (34). Signals were μl of Lipofect-Amine 2000 reagent
visualized using enhanced (Invitrogen Life Technologies, Carlsbad,
chemiluminescence detection kit CA) in serum-free RPMI 1640 medium
(SuperSignal West Pico; Pierce). following the manufacturer’s instruction. 24
RT-PCR analysis-RNA was extracted from h after transfection, the cells were infected
Lung homogenates and cells with Trizol with PAK at 10:1 MOI for 1 h (37). Cell
(Invitrogen Life Technologies, Carlsbad, lysates were subjected to luciferase activity
CA) according to the manufacturer’s analysis by using the Dual-Luciferase
instructions. For detected genes, reverse Reporter Assay System (Promega, Madison,
transcription (RT) was performed using 1.5 WI).
g of RNA, RNase ribonuclease inhibitor, 3-(4,5-dimethylthiazol-2-yl)-2,5-dimethyltet
Oligo dT and cloned AMV reverse razolium bromide (MTT) assay-This assay
transcriptase (Invitrogen Life Technologies, measures color change of MTT upon
Carlsbad, CA). PCR products were reduction by enzymes to assess the viability
separated by 1.0% agarose gel of cells. BAL fluid was centrifuged and
electrophoresis containing ethidium cells were cultured in a 96 well plate. After
bromide and visualized under UV light. The incubation with P. aeruginosa, MTT dye
results for each gene were normalized in was added at final concentration of 1 μg/ml
comparison with GAPDH expression (35). per well. The cells were incubated at 37 °C
Confocal microscopy and indirect for 60 min or until color change occurred.
immunofluorescence staining-Cells were The dye is yellow in color and upon

  4
reduction by enzymes, which gave a blue with the serum-free medium for 1 h. Then
formazan product. The reaction was stopped GFP-PAO1 was used to infect the cells at
by adding 100 μl of stop solution (10% 10:1 MOI. After 1 h incubation at 37 °C, the
DMSO; 10% SDS in 50 mM HEPES wells were washed and treated with
buffer). The plate was left at room polymyxin B 100 μg/ml for 1 h to kill any
temperature overnight for complete remaining extracellular bacteria (29). The
dissolution of formazan crystals. Next day, number of phagocytosed bacteria was
the plate was read at 560 nm absorbance counted using Synergy HT fluorimeter
using a plate reader to quantify the dye (BioTek, Winooski, VT) with 485 ± 20 nm
conversion (38). Duplicates were done for excitation and 528 ± 20 nm emission filters.
each sample and control. Background Background correction was done for
correction was done with blanks containing autofluorescence. Duplicates were done for
dye alone. each sample and control. Alternatively,
NBT assay-This assay is based on color classical colony forming unit (CFU) was
change of NBT dye upon reduction by performed to quantitate phagocytosis as
released superoxide. Cells were treated as described previously.
above and same amount of dye added as Myeloperoxidase (MPO) assay-MPO assay
above. The dye is yellow in color and upon was performed as described. Samples were
reduction by superoxide, which gave a blue homogenized in 50 mM
formazan product (39). hexadecyltrimethylammonium bromide
Dihydrodichlorofluorescein diacetate (HTAB) 50 mM KH2PO4, pH 6.0, and 0.5
(H2DCF-DA or H2DCF) assay-H2DCF dye mM EDTA, 1 ml/100 mg tissue and
(Molecular Probes, Carlsbad, CA) does not centrifuged or 15 min at 12,000 rpm at 4°C.
normally fluoresce and emits green Supernatants were decanted and 100 ml of
fluorescence upon reaction with superoxide reaction buffer (0.167 mg/ml O-dianisidine,
inside cells. Cells were treated as above and 50 mM KH2PO4, pH 6.0 and 0.0005% mM
equal amount(s) of dye added. After 10 min H2O2) was added to 100 ml sample.
incubation, fluorescence was measured Absorbance was read at 460 nm at 2 min
using a fluorimeter (BioTek, Winooski, VT) intervals. Duplicates were done for each
(39,40). sample and control (41).
Lipid peroxidation assay-Malondialdehyde Bacteria burden assay-AM cells from BAL
(MDA) is an end product of lipid fluid and ground lung, spleen, liver, kidney
peroxidation process, and was measured in tissues were homogenized with PBS and
a colorimetric assay (Calbiochem, San were spread on LB plates to enumerate
Diego, CA) according to the manufacturer’s bacteria levels. The plates were cultured in
instructions. Homogenized lung tissue in 37°C incubator overnight and colonies were
62.5 mM Tris-HCl (pH=6.8) supplemented counted. Duplicates were done for each
with Complete-Mini Protease Inhibitor sample and control.
(Roche Diagnostics) in equal protein Histopathology analysis-Lung tissues were
amounts were used in the assay. Duplicates fixed in 10% formalin then embedded in
were done for each sample and control (41). paraffin using a routine histologic
Phagocytosis assay-AM cells obtained from procedure. Four-micrometer sections were
BAL fluid were plated in 96 well plates and cut and stained by standard H&E and
grown overnight. The cells were treated examined for differences in morphology

  5
post-infection (42). analyzed for the percentage of PMN cells.
Statistical analysis-All experiments were PMN penetration to the lung was increased
performed in triplicate and repeated at least in the BAL fluid and serum of cav1 KO
three times. Data were presented as percent mice when compared to WT mice (Fig.
changes compared with the controls ± SD 2B-C). 
from the three independent experiments. P. aeruginosa infection was previously
Group means were compared by One-way shown to induce the release of reactive
ANOVA [Turkey’s post-hoc], using SPSS oxygen species (ROS) in lung, which may
software, and difference was accepted at p < accumulate and eventually cause lung
0.05 (39). Survival test was represented by injury (43). To measure this oxidative stress,
Kaplan-Meier survival curves, using SPSS cav1 KO and WT mice were infected by
software. PAK and AM cells were obtained.
Compared to WT mice, AM cells of cav1
Results KO mice showed increased oxidative stress
Decreased survival in cav1 KO mice-To 18 h post infection as determined by NBT
determine the role of Cav-1 in P. assays (Fig. 2D). These results were further
aeruginosa infection, we intranasally confirmed using the H2DCF assay (Fig. 2E),
instilled PAK (0.5×107 CFU/mouse) to a sensitive fluorescence method for
cav1 KO and WT mice (with otherwise quantifying superoxide. Moreover,
similar genetic backgrounds). As shown decreased mitochondrial membrane
in Fig. 1, cav1 KO mice exhibited increased potential was also observed in cav1 KO
lethality (50% cav1 KO mice died within 24 mice compared to WT mice using JC-1
h post infection). At 60 h, all KO mice died fluorescence assays (Fig. 2F), indicating
while 50% of the wt control mice remained that increased oxidation resulted in
alive. The result is represented by apoptotic cell death. These data collectively
Kaplan-Meier survival curves (p = 0.017, suggest that increased ROS may hamper
Log-rank test). These findings suggest that cell survival by increasing apoptosis and
Cav-1 is required for host defense in P. may significantly damage the lung and
aeruginosa infection in acute pneumonia other organs. These results are consistent
models. with the increased PMN penetration in the
Increased bacterial burdens and oxidation lung and serum in cav1 KO mice (Fig.
in cav1 KO lungs-To fully analyze the cause 2B-C).
of the lethality, moribund mice were killed Under P. aeruginosa infection, PMN are
and organs were aseptically removed for expected to infiltrate into the lung to clear
various assays. Lung homogenates were bacteria through ROS and proteases.
used to measure bacterial burdens. Cav-1 Although bacterial clearance is dependent
KO mice showed significantly increased in part on the production of ROS, excessive
CFUs of PAK in the lung tissue and AM ROS accumulation may cause lung injury.
cells when compared with WT mice (Fig. As a direct indicator of lung injury, lung
2A; p < 0.001, one-way ANOVA), histology was examined 18 h post PAK
indicating severe lung injury and infection. Although both cav1 KO mice and
pneumonia. To quantitatively determine WT control mice showed signs of
polymorphonuclear neutrophil (PMN) pneumonia, significant histological
infiltration, BAL fluid and serum were alterations and increased PMN infiltration

  6
were only observed in the lungs of cav1 KO The lung showed a marked increase in lipid
mice, indicating more severe lung injury in peroxidation compared to the liver and
these animals (Fig. 2G). The insets kidney, suggesting that the lung was the
demonstrate the typical regions with serious main target. These data were consistent with
tissue damage or inflammatory response. the severity of lung injury determined using
Infection dissemination in cav1 KO mice-A MPO assay and superoxide detection,
logical question is whether or not the suggesting that the increased lipid
intranasal inoculation only caused a local peroxidation in KO mice is relevant to the
infection within the lung. Using tissue progression of lung injury by oxidation.
homogenates, we assessed bacterial burdens Altered AM function in cav1 KO mice-AM
in several other organs. CFUs of PAK were cells play a crucial role in bacterial
also increased in the spleen, liver, and clearance by phagocytosis (43). To explore
kidneys, indicating that bacteria were able this relationship, we also measured the
to spread from the original inoculation site ingested PAK bacteria in AM cells.
(lungs) (Fig. 3A-C). The spleen, which is Increased bacterial burdens were found in
the most sensitive indicator for bacterial the AM cells of cav1 KO mice compared to
spread, displayed marked increase in those of WT mice as assessed by CFU (Fig.
bacterial burdens (Fig. 3A). Consistent with 4A). However, this result still cannot
the PMN penetration into the serum (Fig. address whether the bacteria were dealt
2C), these results also suggest that the with through active phagocytosis or passive
severe lung injury and bacterial spread into uptake. To determine the function of AM
other organs are the possible cause of cells, equal amounts of PAO1-GFP, a strain
mortality in these mice. of P. aeruginosa with green fluorescence
To confirm the dissemination results, we (31,39), were added into cultured AM cells
next determined myeloperoxidase (MPO) isolated from cav1 KO mice or WT mice
activity in the lung and other organs to for 2 h incubation. This allowed us to
again probe neutrophil penetration, since conveniently quantify phagocytosis by
MPO is a recognized influx for oxidation in measuring fluorescence intensity using a
tissue. As expected, similar MPO activity fluorescent plate reader (BioTek) (32,44).
was observed in the lung, liver, and kidney Phagocytosis of cav1 KO AM cells was
(Fig. 3D-F). Increased MPO in organs other significantly lower than that of WT AM,
than the lung (e.g., liver and kidney) indicating that Cav-1 may play a role in
suggest that the superoxide release may phagocytosis by AM cells (Fig. 4B). These
result from systemic spread of the invading results also indicate that increased
PAK bacteria. pre-ingestion of the pathogen resulted in
As increased oxidation can also cause reduced phagocytic capacity in AM
tissue injury by oxidative degradation of following infection.
lipids, we used a thiobarbituric acid reactive Survival of AM cells post infection is
substances (TBARS) assay to detect lipid also an essential factor for maintaining their
peroxidation in the lung, liver, and kidney bacterial clearance function. Thus, to
tissue. Our results show that lipid determine the AM viability, AM cells were
peroxidation was significantly increased in infected with PAK for 1 h at MOI 10:1.
all of the PAK infected organs of cav1 KO After adding polymyxin B (to remove the
mice as compared to WT mice (Fig. 3G-I). surface bacteria, Sigma-Aldrich), survival

  7
levels of AM were measured, which again we assessed several signaling proteins in
showed a decrease in the survival of AM lung tissues by western blotting (Fig. 5G).
cells in cav1 KO mice after PAK infection We found that NF-B was highly activated
(Fig. 4C). These results indicate that Cav-1 in protein expression (2-fold) and
may play a critical role in AM-mediated phosphorylation (1.4-fold). To further
host defense against PAK infection. dissect the pathway of cytokine response,
Altered inflammatory response in cav1 KO we also analyzed STAT activation. STAT3,
mice-Cav-1 has been implicated in a transcription factor associated with
bacteria-induced inflammatory responses. NF-B activation in various cellular
Cytokine concentrations of BAL fluid were processes, showed significant expression in
measured 18 h post PAK infection to probe cav1 KO mouse lungs (4-fold) and
various cytokines for reflecting phospho-STAT3 was drastically increased
inflammatory response. The BAL fluid of (24-fold, Fig. 5G). Since STAT5 was not
cav1 KO mice showed no significant significantly increased (data not shown),
difference in inflammatory response our data suggest that STAT3 might play a
compared to that of WT mice. However, in specific regulatory role in this infection (20).
cav1 mouse lungs, IL-2 and IL-10 levels We then evaluated the immediate up-stream
were significantly elevated compared with regulator of STAT3, and found that
WT mice as assayed by ELISA (p < 0.05). phospho-JAK2 was dramatically increased
Furthermore, TNF- and IL-6 levels were in cav1 KO mice (34-fold) (Fig. 5G). We
increased by approximately 4-fold in cav1 also analyzed roles of SOCS3, a negative
KO mice (p < 0.01) (Fig. 5A-D). This regulator of STAT3. A significant increase
indicates that the lung epithelium may be in SOCS3 was seen in cav1 KO mice as
the main source of increased cytokines. To compared to WT mice by western blotting
validate the above data, mRNA levels of the analysis (data not shown), suggesting that
above-quantified cytokines were also SOCS3 was responsive to the
measured by semi-quantitative RT-PCR. infection-induced inflammatory response.
mRNA expressions of TNF-α, IL-6, IFN-, These results strongly indicate the
and IL-12a were all up-regulated by activation of STAT3 in cav1 KO mice as
approximately 2 to 4 fold as determined by compared to WT mice.
densitometry (Fig. 5E). Similarly, levels of PAO1 infection displays similar phenotypes
TNF-, IFN-, IL-1, IL-12a, and IL-6 in cav1 KO mice-To determine whether the
were increased in cav1 KO lung tissues as infection response and its mechanism are
assessed by western blotting with similar across different bacterial strains, we
densitometrical analysis, particularly IL-12a studied the immune response to another
and IL-6 (Fig. 5F). Collectively, these data well-studied P. aeruginosa strain - PAO1 in
indicate that cav1 KO mice showed more cav1 KO mice. A similar infection condition
intense pro-inflammatory response (TNF-, was used, except for a higher CFU count
IL-1 and IL-6) following PAK infection because PAO1 is less cytotoxic than PAK.
as compared to WT mice. Consistent with observations in PAK
Activation of NF-B pathway and STAT3 by infection, cav1 KO mice also showed
PAK infection in cav1 KO mice-To illustrate elevated bacterial burdens in lungs and in
the mechanism that causes the dysregulated other major organs (the liver, spleen, and
response to PAK infection in cav1 KO mice, kidney) after PAO1 infection (Fig. 6A-D).

  8
Our findings demonstrate that lung tissues PAK for 1 h at MOI 10:1 and lysed for CFU
were severely injured in cav1 KO mice as assay. As expected, increased bacterial
compared to WT mice. In addition, sepsis burdens were observed in cav1 DN
was also observed in cav1 KO mice versus MLE-12 cells (Fig. 7A). Next, we assessed
WT mice by PAO1 infection. Furthermore, the mRNA levels for NF-B, JAK2, and
inflammatory response indicated by STAT3, which were not significantly
cytokines and superoxide was also activated at an early phase (1 h post
increased in cav1 KO mice following PAO1 infection). However, mRNA levels of IL-6,
infection (data not shown). We then and IL-12a significantly increased. To
investigated the NF-B pathway and ascertain the role of STAT3, WP1066 (a
STAT3 activation using western blotting STAT3 inhibitor, Sigma-Aldrich) was used
analysis. Similar to PAK strain, PAO1 strain to pre-treat the cav1 DN cells. WP1066 has
also induced significant inflammatory been demonstrated to inhibit the
responses (pNF-B 6.4-fold, pSTAT3 phosphorylation of STAT3 at low M range,
5.4-fold, and pJAK2 5.8-fold) in cav1 KO thereby effectively blocking STAT3
mice as compared to WT mice. These signaling (47). After treatment with the
results indicate that PAO1 infection also STAT3 inhibitor, activation of IL-6 and
activated the STAT3 pathway along with TNF- was abolished, while the total
NF-B activation (Fig. 6E). In addition, mRNA levels were not altered (Fig. 7B).
levels of inflammatory cytokines, IL-6 (7.3 These data support the view that STAT3
fold) and TNF- (5.6 fold), were also plays a crucial role in cytokine production.
increased in cav1 KO mice as compared to Furthermore, we determined the protein
WT mice. Interestingly, the SOCS3 level levels of the aforementioned cytokines and
was much higher in cav1 KO mice than WT their regulators. Consistent with the mRNA
mice (data not shown), which was also expression, the protein levels of IL-6 and
more significantly activated than in the TNF- were increased in cav1 DN
PAK-infected mice (Fig. 5G). These data transfected cells when compared to WT
indicate that PAO1 infection with cav1 KO controls by semi-quantitative RT-PCR (Fig.
mice exhibited a similar phenotype to the 7C), which showed that cav1 DN
PAK strain and that a more rigorous transfection resulted in a similar profile as
inflammatory response was induced by the observed in cav1 KO mice. By contrast,
lower-cytotoxicity strain PAO1, as protein levels of IL-6 and TNF-α were
compared to PAK. suppressed by WP1066 pretreatment.
NF-B pathway is altered by mutating Importantly, phosphorylation of NF-B,
Cav-1 in MLE-12 cells and by STAT3 JAK2, and STAT3 was all significantly
inhibitor-To better understand the role of increased in cav1 DN transfected cells
STAT3 in the NF-B pathway, we versus the control. Similarly, WP1066
transfected murine alveolar epithelial pre-treatment abolished the phosphorylation
MLE-12 cells with either WT cav1 or a of NF-B, JAK2, and STAT3 in cav1 DN
dominant negative (DN) cav1 expressing cells (Fig. 7C). These results confirm our
plasmid (45). MLE-12 cells have been findings observed in cav1 KO mice,
widely used as a model for analyzing indicating that the typical phenotypes may
murine lung epithelial function (39,41,46). be attributable to a dysregulated
Transfected cells were then infected with pro-inflammatory cytokine response

  9
through the Cav-1-STAT3-NF-B axis. because both phagocytosis of the bacteria
Inactive (resting) NF-B and STAT3 by alveolar macrophages and cytokine
are normally retained in the cytosol of production by alveolar epithelial cells were
MLE-12 cells (48). We hypothesized that Cav-1-dependent (50). Similar phenotypes
these factors can be activated by infection, have been observed with other pathogens
which may induce their translocation to the such as S. Typhimurium (51), showing
nucleus (49). We examined the localization increased susceptibility, increased
of NF-B by confocal microscopy, and production of cytokines, and elevated
found that both NF-B and STAT3 were bacterial burdens in cav1 KO mice. Thus,
activated and translocated into the nucleus Cav-1 may render critical immune defense
in cav1 DN MLE-12 cells, but not in against a wide range of microorganisms.
vector-treated MLE-12 cells (Fig. 8A-B). Previous studies have demonstrated
To further probe the activation of NF-B, that caveolins are widely expressed in lung
we transfected MLE-12 cells with luciferase epithelial cells (52,53). Because caveolins
expression of NF-B. Consistent with the are associated with lipid rafts (54), they are
imaging data, luciferase promoter assay thought to be an important part of host
defined the activation of NF-B (Fig. 8C). response to P. aeruginosa infection in lung
Importantly, we reaffirmed that the epithelial cells (7). The majority of previous
activation of NF-B was also associated studies have focused on illustrating the role
with the STAT3 pathway, because the of caveolins in regulating the cell responses
elevation in luciferase activity was to pathogen virulence factors like LPS
significantly inhibited by WP1066. A (28,55-57). However, limited reports have
simplified model represents the pathway actually investigated the role of Cav-1 in
implicated in cav1 KO mice and cav1 DN the innate immunity of the lung against P.
cells (Fig. 8D). aeruginosa infection. Two previous reports
showed different phenotypes in cav1 KO
Discussion mice with a single pathogen P. aeruginosa
In this study, we investigated the (11,12). This discrepancy must be addressed
phenotype of P. aeruginosa infection in in order to understand the real role of this
cav1 KO mice. Severe disease phenotypes critical protein. Our timely studies reveal
in cav1 KO mice were observed in our that Cav-1 is indispensable in host defense
experiments including decreased survival, against P. aeruginosa infection, consistent
increased inflammatory response, and more with the observation made by Gadjeva et al.
severe lung injury compared to WT mice. Furthermore, our studies reveal
We reasoned that dysregulated cytokine mechanistically that Cav-1 is a powerful
response and increased superoxide release negative regulator in pro-inflammatory
contribute to this exaggerated pathogenesis. cytokines during this infection.
Importantly, we have identified a novel We have demonstrated that ROS levels
mechanism involved in the phenotype, in were significantly increased and that
which the STAT3 pathway is responsible for mitochondrial potential was decreased in
the dysregulated inflammatory response. AM cells of cav1 KO mice as compared to
Our results are consistent with the that found in WT mice. These data showed
hypothesis that Cav-1 plays an essential an elevated oxidative stress in cav1 KO
role in the clearance of P. aeruginosa, mice, which might cause lung injury and

  10
systemic bacterial infection. modifications of the cell signaling pathway
Over-production of superoxide can impair in the lungs were examined. In particular,
innate immunity, including the phagocytosis we found NF-B, a major transcription
function of AM, the cells that form the front factor for cytokine production in alveolar
line of innate defense in the lung. Indeed, epithelial cells, to be highly activated (61).
AM cells have been shown to be To investigate the molecular events
dysfunctional in cav1 KO mice, exhibiting associated with this activation, we studied
reduced phagocytosis and increased the JAK/STAT pathway. Interestingly,
susceptibility to cell death after P. STAT3, but not STAT5 was found to be
aeruginosa infection. Although similar highly activated in cav1 KO mice following
methods were utilized in cross-pathogen infection. The interaction between NF-B
comparison study by Lisanti et al., no and STAT3 has been discussed extensively
difference in bacterial ingestion was found in various experimental models, particularly
between cav1 KO mice and WT mice with in cancer research, whereas the
S. Typhimurium infection (51). This STAT3-NF-B axis is still relatively
discrepancy may be due to inherent undefined in Gram-negative infections.
differences in the two pathways, or the Among the cytokines whose expression was
distinct experimental settings. In addition, altered, TNF-α and IL-6 were most
PMN cells, another important cell type significantly up-regulated. Considering that
responsible for bacterial clearance, show TNF-α and IL-6 are able to induce the
lowered phagocytosis against P. aeruginosa activation of NF-B and STAT3,
under Cav-1 deficiency (12). Together, respectively, these significant changes
these observations demonstrate that the might be partially attributable to positive
bacterial clearance mechanism is impaired feedback (22,62). Interestingly, we also
in Cav-1 deficient mice, at least in the case observed significantly increased expression
of P. aeruginosa infection. Furthermore, the and phosphorylation of Akt and
sustained recruitment of PMN cells into the GSK3data not shown, which may be the
lung necessary for bacterial clearance may other pathways responding to or resulting
release excess superoxide and proteases, from the dysregulated cytokine production,
resulting in more severe lung injury and and may interact with the JAK2/STAT3
systemic bacterial infection. pathway in Cav-1 deficient animals.
Another major contributing factor to Consistent with our observations, previous
lung injury and mortality is inflammatory reports have showed the relevance of the
response. We found that the lungs of cav1 PI3K/Akt axis (27) and GSK3to a
KO mice exhibit increases in JAK/STAT pathway (63). These indicate
pro-inflammatory cytokines, but a similar that in cav1 KO mice, infection by various
increase was not found in the BAL fluid of bacteria may activate several different
these mice. Thus, lung epithelial cells might pathways to compensate for the loss of this
also contribute to the production of critical protein. Indeed, there is little doubt
cytokines (41,58,59), while macrophages that Cav-1 is important for regulating lipid
and neutrophils are traditionally regarded as raft-mediated endocytosis and various other
the main players in inflammatory responses cellular events (32,64,65). Despite such
(60). To further explore the dysregulation of activation in various pathways, loss of
inflammatory response in KO mice, Cav-1 cannot be completely rescued to

  11
combat infections and usually leads to interesting question for future studies.
lethal consequences, because crucial In conclusion, we have demonstrated a
cellular signaling systems may have been typical phenotype of P. aeruginosa infection
ill-regulated. For example, Cav-1 has been in cav1 KO mice, and our data indicate an
previously reported to interact with STAT3 important role for Cav-1 in innate immunity
directly through molecular interactions (51). in mice. Cav-1 deficiency impaired the
Deficiency in Cav-1 may impact STAT5 phagocytic ability and other immune
activity (66), while the ultimate effect on defense mechanisms, resulting in sustained
STAT5 activity is cell-dependent (51). infiltration of PMN cells into the lung and
Furthermore, no previous studies have intense inflammatory response. Along with
linked Cav-1 with the STAT3 pathway in the bacterial infection-induced oxidative
response to a respiratory pathogen. Thus, stress, the accumulation of PMN cells and
linking Cav-1 with STAT3-NF-B is a failure of bacterial clearance may cause
novel finding for this host response. Finally, severe lung injury and systemic bacterial
the role of the STAT3 pathway in AM is dissemination, which finally results in host
currently unclear and is worth studying. mortality. In addition, we revealed a novel
Thus, we hypothesized that STAT3 Cav-1-STAT-3-NF-B axis directly
might play a vital role in regulating NF-B contributing to a dysregulated cytokine
activation. We used MLE-12 cells to profile in cav1 KO mice. More importantly,
establish a Cav-1 deficient lung epithelial we confirmed this Cav-1-STAT-3-NF-B
culture model. Our data indicate that pathway in cav1 knockdown MLE-12 cells
transfection of a cav1 DN plasmid in during P. aeruginosa infection. Taken
MLE-12 cells showed a similar together, these observations have provided
inflammatory response and cell signaling an insight into the role of Cav-1 in innate
activation as seen in cav1 KO mice. STAT3 immunity against P. aeruginosa and might
inhibitor WP1066 was then used to confirm indicate novel targets for clinical therapy.
the role of STAT3 in MLE-12 cells. As
expected, STAT3 inhibition decreased
NF-B activation and reduced the
inflammatory response, indicating that
STAT3 activation is essential for NF-B
activation and down-stream inflammatory
response modifications. Taken together, our
data indicate that Cav-1 initiates a negative
regulation in the STAT3-NF-B pathway.
Although SOCS3, a chief negative regulator
of SOCS3, was also increased in cav1 KO
mice (data not shown), the over-activation
of STAT3 and associated inflammatory
response was still not contained. Thus, our
study suggests that Cav-1 may potentiate
the negative regulatory effects of SOCS3 on
inflammatory response, while loss of Cav-1
significantly dampens this role, an

  12
References:

1.  Driscoll, J. A., S. L. Brody, and M. H. Kollef. (2007) Drugs 67, 351‐368 
2.  Lyczak, J. B., Cannon, C. L., and Pier, G. B. (2000) Microbes Infect 2, 1051‐1060 
3.  Heijerman, H. (2005) J Cyst Fibros 4 Suppl 2, 3‐5 
4.  Fagon, J. Y., J. Chastre, Y. Domart, J.L. Trouillet, and C. Gibert. (1996) Clin. Infect. Dis. 23, 538‐542 
5.  Dunn, M., and R. G. Wunderink. (1995) Clin. Chest Med. 16, 95‐109 
6.  Crouch, B. S., R. G. Wunderink, C. B. Jones, and K. V., Jr. Leeper. (1996) Chest 109, 1019‐1029 
7.  Zaas, D. W., M. J. Duncan, G. Li, J. R. Wright, and S. N. Abraham. (2005) J. Biol. Chem. 280, 4864‐4872 
8.  Grassme, H., V. Jendrossek, A. Riehle, G. VonKurthy, J. Berger, H. Schwarz, M. Weller, R.    Kolesnick, 
and E. Gulbins. (2003) Nat. Med. 9, 322‐330 
9.  Garcia‐Medina, R., W. M. Dunne, P. K. Singh, and S. L. Brody. (2005) Infect. Immun. 73, 8298‐8305 
10.  Chastre, J., and Fagon, J. Y. (2002) Am J Respir Crit Care Med 165, 867‐903 
11.  Zaas, D. W., Z. D. Swan, B. J. Brown, G. Li, S. H. Randell, S. Degan, M. E. Sunday, J. R. Wright, and S. N. 
Abraham. (2009) J. Biol. Chem. 284, 9955‐9964 
12.  Gadjeva, M., C. Paradis‐Bleau, G. P. Priebe, R. Fichorova, and G. B. Pier. (2010) J. Immunol. 184, 
296‐302 
13.  Scherer, P. E., T. Okamoto, M. Chun, I. Nishimoto, H. F. Lodish, and M. P. Lisanti. (1996) Proc. Natl. 
Acad. Sci. U.S.A. 93, 131‐135 
14.  Tang, Z., P. E. Scherer, T. Okamoto, K. Song, C. Chu, D. S. Kohtz, I. Nishimoto, H. F. Lodish, and M. P. 
Lisanti. (1996) J. Biol. Chem. 271, 2255‐2261 
15.  Williams, T. M., and M. P. Lisanti. (2004) Genome Biol. 5, 214 
16.  Mora, R., V. L. Bonilha, A. Marmorstein, P. E. Scherer, D. Brown, M. P. Lisanti, and E. Rodriguez‐Boulan. 
(1999) J. Biol. Chem. 274, 25708‐25717 
17.  Thomas, S., A. P. Pais, S. Casares, and T. D. Brumeanu. (2004) Molecular Immunology 41, 399‐409 
18.  Thomas, S., R. S. Kumar, and T. D. Brumeanu. (2004) AITE 52, 215‐224 
19.  Korade, Z. (2008) Neuropharmacology 55, 1265‐1273 
20.  Grivennikov, S. I. a. M. K. (2010) Cytokine & Growth Factor Reviews 21, 11‐19 
21.  Ivashkiv, L. B. (2010) Cell Host Microbe 8, 132‐135 
22.  Takeda, K., T. Kaisho, N. Yoshida, J. Takeda, T. Kishimoto, and S. Akira. (1998) J. Immunol. 161, 
4652‐4660 
23.  Jones, M. R., Quinton, L. J., Simms, B. T., Lupa, M. M., Kogan, M. S., and Mizgerd, J. P. (2006) J Infect 
Dis 193, 360‐369 
24.  Wang, L., Kurosaki, T., and Corey, S. J. (2007) Oncogene 26, 2851‐2859 
25.  Norkina, O., Dolganiuc, A., Catalano, D., Kodys, K., Mandrekar, P., Syed, A., Efros, M., and Szabo, G. 
(2008) Alcohol Clin Exp Res 32, 1565‐1573 
26.  Maruvada, R., Argon, Y., and Prasadarao, N. V. (2008) Cell Microbiol 10, 2326‐2338 
27.  Shen‐Tu, G., Schauer, D. B., Jones, N. L., and Sherman, P. M. (2010) Lab Invest 90, 266‐281 
28.  Drab, M., P. Verkade, M. Elger, M. Kasper, M. Lohn, B. Lauterbach, J. Menne, C. Lindschau, F. Mende, 
and F. C. Luft. (2001) Science 293, 2449‐2452 
29.  Kannan, S., Audet, A., Huang, H., Chen, L. J., and Wu, M. (2008) J Immunol 180, 2396‐2408 
30.  Wisniowski, P. E., Spech, R. W., Wu, M., Doyle, N. A., Pasula, R., and Martin II, W. J. (2000) Am J Respir 

  13
Crit Care Med 162, 733‐739 
31.  Priebe, G. P., Brinig, M. M., Hatano, K., Grout, M., Coleman, F. T., Pier, G. B., and Goldberg, J. B. (2002) 
Infect Immun 70, 1507‐1517 
32.  Kannan, S., Audet, A., Knittel, J., Mullegama, S., Gao, G. F., and Wu, M. (2006) European J Immunol 36, 
1739‐1752 
33.  Kannan, K., Huang, H., Seeger, D., Audet, A., Chen, Y., Huang, C., Gao, H., Li, S., and Wu, M. (2009) 
PLoS One 4, e4891 
34.  Wu, M., Stockley, P. G., and Martin II, W. J. (2002) Electrophoresis 23, 2373‐2376 
35.  Wu, M., Harvey, K. A., Ruzmetov, N., Welch, Z. R., Sech, L., Jackson, K., Stillwell, W., Zaloga, G. P., and 
Siddiqui, R. A. (2005) Int J Cancer 117, 340‐348 
36.  Kannan, S., Pang, H., Foster, D., Rao, Z., and Wu, M. (2006) Cell Death Differ 13, 311‐323 
37.  Yan, C., Cao, J., Wu, M., Jiang, T., Yoshimura, A., and Gao, H. (2010) J Biol Chem 285, 37227‐37239 
38.  Wu, M., Brown, W. L., and Stockley, P. G. (1995) Bioconjugate Chem 6, 587‐595 
39.  Wu, M., Huang, H., Zhang, W., Kannan, S., Weaver, A., Mckibben, M., Herington, D., Zeng, H., and Gao, 
H. (2011) Infect Immun 79, 75‐87 
40.  Wu, M., Audet, A., Cusic, J., Seeger, D., Cochran, R., and Ghribi, O. (2009) J Neurochem 111, 
1011‐1021 
41.  Kannan, S., H. Huang, D. Seeger, A. Audet, Y. Chen, C. Huang, H. Gao, S. Li, and M. Wu. (2009) Plos 
One 4, e4891 
42.  Wu, M., Hussain, S., He, H. Y., Pasula, R., Smith, P. A., and Martin II, W. J. (2001) Proc Natl Acad Sci U S 
A 98, 14589‐14594 
43.  Suntres, Z. E., O. Abdelwahab, and P. N. Shek. (2002) Microbial Pathogenesis 32, 27‐34 
44.  Kannan, S., A. Audet, H. Huang, L. Chen, and M.    Wu. (2008) J. Immunol. 180, 2396‐2408 
45.  Brazer, S. C., Singh, B. B., Liu, X., Swaim, W., and Ambudkar, I. S. (2003) J Biol Chem 278, 27208‐27215 
46.  Renier, M., Tamanini, A., Nicolis, E., Rolfini, R., Imler, J. L., Pavirani, A., and Cabrini, G. (1995) Hum 
Gene Ther 6, 1275‐1283 
47.  Kenneth, R. F., A. H. Charles, and L. C. Timothy. (2005) Clin. Cancer Res. 11, A277 
48.  Sugahara, K., A. Mizutani and H. Yamamoto. (2010) Am. J. Respir. Crit. Care Med. 181, A3613 
49.  Arita, Y., T. Ito, T. Oono, K. Kawabe, T. Hisano, and R. Takayanagi. (2008) World J. Gastroenterol. 14, 
4473‐4479 
50.  Kowalski, M. P., A. Dubouix‐Bourandy, M. Bajmoczi, D. E. Golan, T. Zaidi, Y. S. Coutinho‐Sledge, M. P. 
Gygi, S. P. Gygi, E. A. Wiemer, and G. B. Pier. (2007) Science 317, 130‐132 
51.  Medina, F. A., C. J. de Almeida, E. Dew, J. Li, G. Bonuccelli, T. M. Williams, A. W. Cohen, R. G. Pestell, P. 
G. Frank, H. B. Tanowitz, and M. P. Lisanti. (2006) Infect. Immun. 74, 6665‐6674 
52.  Barth, K., K. Weinhold, A. Guenther, M. T. Young, H. Schnittler, and M. Kasper. (2007) FEBS Journal 274, 
3021‐3033 
53.  Barar, J., L. Campbell, A. J. Hollins, N. P. B. Thomas, M. W. Smith, C. J. Morris, and M. Gumbleton. 
(2007) Biochemical and Biophysical Research Communications 359, 360‐366 
54.  Jacobson, K., O. G. Mouritsen and R. G. Anderson. (2007) Nat. Cell Biol. 9, 7‐14 
55.  Shaykhiev, R., J. Sierigk, C. Herr, G. Krasteva, W. Kummer, and R. Bals. (2010) The FASEB Journal 24, 
4756‐4766 
56.  Garrean, S., X. P. Gao, V. Brovkovych, J. Shimizu, Y. Y. Zhao, S. M. Vogel, and A. B. Malik. (2006) J. 
Immunol. 177, 4853‐4860 
57.  Cohen, A. W., R. Hnasko, W. Schubert, and M. P. Lisanti. (2004) Physiol. Rev. 84, 1341‐1379 

  14
58.  Amano, H., K. Morimoto, M. Senba, H. Wang, and Y. Ishida. (2004) J. Immunol. 172, 398‐409 
59.  Thorley, A. J., P. Goldstraw, A. Young, and T. D. Tetley. (2005) Am. J. Respir. Cell Mol. Biol. 32, 262‐267 
60.  Ozaki, T., Maeda, M., Hayashi, H., Nakamura, Y., Moriguchi, H., Kamei, T., Yasuoka, S., and Ogura, T. 
(1989) Am Rev Respir Dis 140, 1595‐1601 
61.  Moodie, F. M., Marwick, J. A., Anderson, C. S., Szulakowski, P., Biswas, S. K., Bauter, M. R., Kilty, L., and 
Rahman, I. (2004) FASEB J 10, 1897‐1899 
62.  Wang, C., M. M. Mayo, and A. S. Baldwin. (1996) Science 274, 784‐787 
63.  Nakayama, M., Hisatsune, J., Yamasaki, E., Isomoto, H., Kurazono, H., Hatakeyama, M., Azuma, T., 
Yamaoka, Y., Yahiro, K., Moss, J., and Hirayama, T. (2009) J Biol Chem 284, 1612‐1619 
64.  Anderson, R. G. (1998) Annu. Rev. Biochem. 67, 199‐225. 
65.  Anderson, R. G., and K. Jacobson. (2002) Science 296, 1821‐1825 
66.  Park, D. S., Lee, H., Frank, P. G., Razani, B., Nguyen, A. V., Parlow, A. F., Russell, R. G., Hulit, J., Pestell, R. 
G., and Lisanti, M. P. (2002) Mol Biol Cell 13, 3416‐3430 

FOOTNOTES
This project was supported by NIH ES014690 and American Heart Association Scientist
Development Grant (MW); and by NIH 5R01HL092905-04 and 3R01HL092905-02S1 (HG).
We thank S. Rolling of UND imaging core for help with confocal imaging.

The abbreviations used are: AM, alveolar macrophage; BAL, bronchoalveolar lavage; Cav-1,
caveolin-1; DN, dominant negative; MOI, multiplicity of infection; MPO,
myeloperoxidase; YFP, yellow fluorescent protein; PA, Pseudomonas Aeruginosa; JAK,
Janus kinase; SOCS, suppressors of cytokine signaling; H2DCF, Dihydrodichlorofluorescein
diacetate; TBARS, thiobarbituric acid reactive substances.
Competing interests’ statement: The authors declare that they have no competing financial
interests.

  15
FIGURE LEGENDS
Figure 1. cav1 KO mice displayed increased susceptibility to P. aeruginosa infection.
cav1 KO mice and WT mice were intranasally challenged with 0.5×107 CFU/mouse PAK.
Then, the mice were maintained up to 72 h. Survival test is represented by Kaplan-Meier
survival curves (p = 0.017, 95% confidence interval: 18.4–44.5, log-rank test).

Figure 2. Increased bacterial burdens, PMN, and oxidation injury in the lung of cav1
KO mice. A, The lungs showed significantly increased bacterial burdens after infection with
PAK in cav1 KO mice compared to WT mice. cav1 KO mice and WT mice were infected
with 0.5×107 CFU/mouse PAK. Frozen tissues were homogenized in PBS. The same
amounts of tissue were evaluated for bacterial colonies for CFUs/gram tissue. B-C, Increased
PMN infiltration and acute inflammatory response were observed in the lung and serum of
cav1 KO mice compared to WT mice. After HEMA-3 staining (Fisher Scientific), PMN cell
percentages were calculated versus total nuclear cells. D, Superoxide production of AM cells
was significantly increased in cav1 KO mice compared to WT mice using NBT assay (1
g/ml). The same amounts of AM cells were infected with PAK at MOI 10:1 for 1 h. The
optical density for NBT was quantified at 560 nm. E, Oxidative stress was increased in cav1
KO mice determined by H2DCF assay (5 M). The fluorescence was quantified at 488 nm.
F, cav1 KO mice showed significantly lower mitochondrial potential than WT mice assessed
by JC-1 fluorescent assay (0.1 g/ml). The fluorescence was quantified at 532 nm. G.
Increased lung injury and inflammatory assessed by histology. cav1 KO mice and WT mice
were infected with 0.5×107 CFU/mouse of PAK for 24 h. Mice were dissected and lungs
were embedded in formalin. Sections were analyzed by H&E staining. The data are
representative of three experiments (One-way ANOVA [Turkey’s post-hoc], * p < 0.05, ** p
< 0.01).

Figure 3. Increased dissemination and oxidation in cav1 KO mice. A-C. The spleen, liver,
and kidneys showed significantly increased bacterial burdens in cav1 KO mice compared
with WT mice after PAK infection. D-F, Increased MPO activity in cav1 KO mouse lung,
liver, and kidney tissue compared to WT mice. G-I, Increased levels of lipid peroxidation
were observed in the lung, liver, and kidney tissues of cav1 KO mice compared to WT mice
by TBARS assay. The data are representative of three experiments (One-way ANOVA
[Turkey’s post-hoc], * p < 0.05, ** p < 0.01).

Figure 4. AM cell function is altered in cav1 KO mice. A. Increased bacterial burdens in


AM cells after infection with PAK in cav1 KO mice compared with WT mice. B, cav1 KO
mice and WT control mice were infected with 0.5×107 CFU/mouse of PAK for 18 h. The
same amounts of AM cells were infected with PAO1-GFP at MOI 10:1 for 1 h. Fluorescence
intensity showed decreased phagocytosis in cav1 KO mice. C, Survival percentages of AM
cells were measured by MTT assay. The data are representative of three experiments
(One-way ANOVA [Turkey’s post-hoc], * p < 0.05, ** p < 0.01).

Figure 5. PAK infection induced intense inflammatory response in cav1 KO mice. A-D,
Increased inflammatory cytokines in BALF of cav1 KO mice compared to WT mice

  16
assessed by ELISA. Mice (6 mice/group) were infected with 0.5×107 CFU/mouse of PAK for
18 h. BAL fluid was collected and cytokines measured. E-F, Increased inflammatory
cytokines in BAL fluid of cav1 KO mice compared to WT mice assessed by RT-PCR (E) and
western blotting (F). Frozen lung tissue of cav1 KO mice and WT mice at 18 h
post-infection was lysed for protein and mRNA assays. G, PAK infection activated the
NF-B pathway and STAT3 in cav1 KO mice. cav1 KO mice and WT mice were infected
with 0.5×107 CFU/mouse of PAK for 18 h. Gel data were quantified using densitometry with
Quantity one, and data are representative of three experiments (One-way ANOVA [Turkey’s
post-hoc], * p < 0.05, ** p < 0.01).

Figure 6. PAO1 infection caused similar phenotypes in cav1 KO mice as seen in PAK
infection. A-D, Increased bacterial burdens were observed in cav1 KO lung, spleen, liver,
and kidney tissues after PAO1 infection. E, Lung tissues of PAO1 infected cav1 KO mice
and WT mice (5 mice/group) were lysed for measuring cytokines and cell signaling protein
levels by western blotting (One-way ANOVA [Turkey’s post-hoc], ** p < 0.01). Gel data
were quantified using densitometry with Quantity one, and data are representative of three
experiments.

Figure 7. cav1 mutation in MLE-12 cells activated the STAT3-NF-κB pathway by PAK
infection. A, MLE-12 cells were transfected with a cav1 DN or cav1 WT plasmid. After 24 h,
cells were infected with PAK at MOI 10:1 for 1 h. The STAT3-NF-B pathway was
determined by western blotting. Cells were treated by STAT3 inhibitor WP1066 (2 M) for
0.5 h before infection. B, mRNA expression levels of IL-6 and TNF- were measured with
RT-PCR. C, After PAK infection, cav1 DN MLE-12 or WT MLE-12 cells were lysed to
determine expression and phosphorylation of cytokines and cell signaling proteins by
western blotting (three samples/each treatment). Gel data were quantified using densitometry
with Quantity one, and data are representative of three experiments in triplicate (One-way
ANOVA [Turkey’s post-hoc], * p < 0.05).

Figure 8. Elevated nuclear translocation of NF-B and STAT3 induced in cav1 DN


MLE-12 cells. A-B, WT and cav1 DN MLE-12 cells were infected with PAK for 1 h.
Localization of NF-B and STAT3 were visualized by indirect immunofluorescence staining.
Nuclear translocation of NF-B and STAT3 were detected in both WT and cav1 DN
MLE-12 cells (arrows showing the nuclear translocation). WP1066 (2 M) was used to
pre-treat the cells for 0.5 h before infection. C, Activation of NF-B was detected in both
WT and cav1 DN MLE-12 cells using Luciferase promoter assay. Transient transfections
were performed with 2×105 cells plated in 6-well plates using 2 g of DNA and 3 l of
LipofectAmine 2000 reagent for 24 h. After 1 h PAK infection at 10:1 MOI, cell lysates were
subjected to luciferase activity analysis using the Dual-Luciferase Reporter Assay System.
The data are representative of three experiments in triplicate (One-way ANOVA [Turkey’s
post-hoc], * p < 0.05). D, Diagram delineates a pathway involved in Cav-1 KO cells against
P. aeruginosa infection. Cav-1 deficiency impacts the phosphorylation of both NF-B and
STAT3 in cytoplasm by P. aeruginosa infection. The translocation of both factors to the
nucleus continuously induces cytokine production and aggravates disease progression.

  17
100%
Cav1 KO
WT

80%
Survival

60%

40%

20%

Time post infection (hours)


Figure 1
Lung BAL 800 Serum
A B 350 ** C *
3 ** 700

106/g tissue
300 μ

l
μ 600

l
250

PMN cells/
500
2

PMN cells/
200 400
150 300
× 1 100 200
CFU

50 100
0 0 0
WT Cav1 WT Cav1 WT Cav1

D 0.35 * E 70 0 * F 180

Mitochondrial Potential
160
0.3 60 0

H2DCF Assay (RFU)
NBT Assay (RLU)

140
0.25 50 0 120
0.2 40 0 100 *
0.15 30 0 80
60
0.1 20 0
40
0.05 10 0
20
0 0 0
WT Cav1 WT Ca v1 WT Cav1
G WT Cav1
Normal                                 PAK Normal                                 PAK

50 μ m Figure 2
A Spleen
B Liver C Kidney
**
* *
2 2 2

106/g tissue
106/g tissue

106/g tissue
1 1 1
× × ×

CFU
CFU

CFU
0 0 0
WT Cav1 WT Cav1 WT Cav1

D Lung
E Liver
F Kidney
0.05

MPO Assay (RLU)
** *

MPO Assay (RLU)
MPO Assay (RLU)

0.04 0.03
0.04
0.03
0.02 0.03
0.02 0.02
0.01 0.01
0.01
0 0 0
WT Cav1 WT Cav1 WT Cav1

G Lung H I
Liver Kidney
Lipid peroxidation (RLU)

Lipid peroxidation (RLU)
Lipid peroxidation (RLU)

0.5 0.25 *
** 0.2

0.4 0.2
0.15
0.3 0.15
0.1
0.2 0.1

0.1 0.05 0.05

0 0 0
WT Cav1 WT Cav1 WT Cav1
Figure 3
4000
A B C

Phagocytosis (RFU)
140%
3500
8 120%
** 3000 *
2500 100%
CFU/cell

6 *

Survival 
2000 80%
4 1500 60%
1000 40%
2
500 20%
0 0 0%
WT Cav1 WT Cav1 WT Cav1 WT+PAK Cav1+PAK

Figure 4
A  B  C  6 **
D
2.5 6 12 **

TNF‐ (ng/ml)
5

IL‐10   (ng/ml)

IL‐6 (ng/ml)
IL‐2  (ng/ml)
2 5 10
4
4 8
1.5 3
3 6
1 2 2 4
0.5 1
1 2
0
0 0 0
WT         Cav1 WT           Cav1 WT          Cav1 WT          Cav1

Control PAK
E Control PAK
F  Control PAK G 
WT Cav1 WT Cav1
WT Cav1 WT Cav1 WT Cav1 WT Cav1
Cav‐1
IFN‐ Cav‐1
pNF‐B
IL‐1 IFN‐ NF‐B
IL‐6 IL‐1
pJAK2
IL‐12a IL‐6 JAK2
TNF‐ IL‐12a pSTAT3
GAPDH TNF‐ STAT3
GAPDH GAPDH
4.5 **
4 IFN -γ 12 30
p N F-κB **
Relative Density

Relative Density
Relative Density

IL -1 β IFN -γ ** **
3.5 10 25 N F-κB
IL -6 IL -1 β
3 * 8 IL -6 p JAK2
IL -1 2 a 20
2.5 TN F-α * * IL -1 2 a p STAT3
2 6 TN F-α * 15 STAT3
1.5 4 * 10
1 *
2 * *
5 *
0.5
0 0 0
(-) PAK (-) PAK (-) PAK
Figure 5
A B C D
Lung Spleen Liver Kidney
106/g tissue

106/g tissue

106/g tissue
106/g tissue
55 **
** 4 3
** ** 2 **
44 3
33 2
2 1
× 22 × × ×
1
11 1
CFU

CFU

CFU
00 CFU 0 0 0
WTWT Cav1
Cav1 WT Cav1 WT Cav1 WT Cav1

E WT                  Cav1 WT                      Cav1
10 **
Cav‐1 pJAK2

Relative Density
p N F-κB *
8 p STAT3 *
pNF‐B JAK2 * *
p JAK2
6 IL -6
NF‐B IL‐6 4 TN F-α

pSTAT3 TNF‐ 2

STAT3 GAPDH 0
WT                      Cav1

Figure 6
A B Control PAK
120 (—)       Cav1      DN   WP1066 (—)      Cav1       DN  WP1066

100 NF‐B
CFU/104 cells

80 JAK2

60 STAT3

40 IL‐6
TNF‐
20
GAPDH
0
(-) Cav1 DN WP1066 (-) Cav1 DN WP1066

Relative Density
IL -6 *
2
TN F-α
Control PAK
1.5

C 1
Control PAK
0.5
(—)       Cav1      DN   WP1066 (—)      Cav1      DN   WP1066
Cav1‐YFP 0
△Cav1‐YFP (-) DN WP1066
pNF‐B
NF‐B
5 **
pJAK2 Relative Density
p N F-κB
JAK2 4 p JAK2 *
p STAT3
pSTAT3 *
3 IL -6 *
STAT3 TN F-α *
2
IL‐6
TNF‐ 1

GAPDH 0
Figure 7 (-) DN WP1066
A NF‐B DAPI           Merge with DIC  B STAT3                    DAPI            Merge with DIC 

(—) (—)

PAK PAK

Cav1 DN Cav1 DN
+ PAK + PAK

WP1066 WP1066
+ PAK + PAK
Relative Luciferase Activity 

C 3000 *
D
2500

2000

1500

1000

500

0
(-) Cav1 DN WP1066 (-) Cav1 DN WP1066

Control       PAK
Figure 8

You might also like