Background:

The heat-shock response (HSR) protects from insults, such as ischemia–reperfusion injury, by inhibiting signaling pathways activated by sterile inflammation. However, the mechanisms by which the HSR activation would modulate lung damage and host response to a bacterial lung infection remain unknown.

Methods:

HSR was activated with whole-body hyperthermia or by intraperitoneal geldanamycin in mice that had their lungs instilled with Pseudomonas aeruginosa 24 h later (at least six mice per experimental group). Four hours after instillation, lung endothelial and epithelial permeability, bacterial counts, protein levels in bronchoalveolar lavage fluid, and lung myeloperoxidase activity were measured. Mortality rate 24 h after P. aeruginosa instillation was recorded. The HSR effect on the release of interleukin-10 and killing of P. aeruginosa bacteria by a mouse alveolar macrophage cell line and on neutrophil phagocytosis was also examined.

Results:

HSR activation worsened lung endothelial (42%) and epithelial permeability (50%) to protein, decreased lung bacterial clearance (71%), and increased mortality (50%) associated with P. aeruginosa pneumonia, an effect that was not observed in heat-shock protein–72-null mice. HSR-mediated decrease in neutrophil phagocytosis (69%) and bacterial killing (38%) by macrophages was interleukin-10 dependent, a mechanism confirmed by increased lung bacterial clearance and decreased mortality (70%) caused by P. aeruginosa pneumonia in heat-shocked interleukin-10-null mice.

Conclusions:

Prior HSR activation worsens lung injury associated with P. aeruginosa pneumonia in mice via heat-shock protein–72- and interleukin-10-dependent mechanisms. These results provide a novel mechanism for the immunosuppression observed after severe trauma that is known to activate HSR in humans.

What We Already Know about This Topic
  • The heat-shock response, activated during trauma, appears to protect organisms from a variety of injuries

What This Article Tells Us That Is New
  • The heat-shock response was activated 24 h before the experimental development of Pseudomonas aeruginosa pneumonia and led to worsened lung endothelial and epithelial permeability, decreased bacterial clearance, and increased mortality

HEAT-SHOCK proteins are ubiquitous molecular chaperones involved in protein folding, peptide trafficking, and antigen processing under both physiologic and stress conditions. When released actively or passively into the extracellular space, heat-shock proteins function as “danger signal” mediators by mediating the transfer of antigenic peptides from stressed cells to the antigen-presenting cells or by activating toll-like receptors.1  Activation of the heat-shock response (HSR) protects host cells and organs from sterile insults, such as oxidative stress or ischemia–reperfusion injury, via the inhibition of inflammatory cellular pathways.2–5  In humans, we have previously shown that the activation of the HSR correlates with survival after severe trauma.6  Furthermore, other investigators have reported that heat-shock protein (Hsp)-72 genotypes influence plasma cytokine levels and interfere with outcome after major injury in humans.7  Despite the evidence that the activation of the HSR may attenuate the severity of a sterile inflammation, the mechanisms by which HSR activation would modulate lung damage and host response to a bacterial lung infection remain largely unknown.

Pseudomonas aeruginosa is an important cause of nosocomial pneumonia in critically ill patients and is associated with a high mortality rate. Host resistance to P. aeruginosa pneumonia requires an intact innate immune response for the clearance of bacteria from the lungs. This has been demonstrated in an experimental model of P. aeruginosa pneumonia8  and indirectly confirmed in a recent clinical study that reported that patients with large burdens of P. aeruginosa who did not meet clinical criteria for ventilator-associated pneumonia had an increased risk of death when compared with the risk in patients who met ventilator-associated pneumonia criteria.9  Because HSR activation inhibits signaling pathways, such as the nuclear factor-κB pathway,10  that are activated by cell membrane receptors and are critical for the eradication of bacteria from the airspaces of the lung,11  we first tested the hypothesis that prior HSR activation would increase the severity of lung injury in a mouse model of P. aeruginosa pneumonia in wild-type mice and in mice null for the inducible Hsp72, which is one of the most important heat-shock proteins expressed during HSR activation.12 

Experimental studies indicate that the initial response to the endogenous release or exogenous administration of antiinflammatory mediators, such as interleukin (IL)-10, is associated with a more severe lung injury caused by bacterial pneumonia.13–21  Because the heat-shock factor 1 released during HSR activation is a transcriptional activator of IL-10 gene expression in macrophages22  and the fact that extracellular Hsp72 causes the release of IL-10 via the activation of the toll-like receptor-4,23  the second aim of the study was designed to test the hypothesis that release of IL-10 during the HSR activation could be an important mechanism to explain the inhibition of the lung innate immune response after HSR activation in a mouse model of P. aeruginosa lung infection.

Reagents

All cell culture media were prepared by the University of California San Francisco and University of Alabama at Birmingham Cell Culture Facilities using deionized water and analytical-grade reagents. The protein concentration of cell lysates was determined using the Bio-Rad protein assay kit (Bio-Rad, Hercules, CA). The enzyme-linked immunosorbent assay kit for mouse IL-10 was purchased from R&D Systems Inc. (Minneapolis, MN). Myeloperoxidase activity was measured with a mouse myeloperoxidase kit HK210 from Cell Sciences (Canton, MA). Anti-Hsp72 antibody was purchased from Enzo Life Sciences (Farmingdale, NY). Anti-IL-10 monoclonal antibody (JES3-9D7) and isotype immunoglobulin G1 (control Ab) were obtained from Pharmingen (San Diego, CA). Geldanamycin was obtained from InvivoGen (San Diego, CA). 125I-labeled human serum albumin (Jeanatope; ISO-TEX Diagnostics, Friendswood, TX) was used as radioactive tracer. All other reagents were purchased from Sigma-Aldrich (St. Louis, MO).

Cell Culture

A mouse alveolar macrophage cell line (designated as MH-S cells: ATCC no. CRL-2019) was used, as we have previously published.24  Cells were cultured in Roswell Park Memorial Institute 1640 medium supplemented with 10% fetal bovine serum and 1% penicillin and streptomycin and were maintained at 37°C with 5% CO2.

Neutrophil Isolation

Mouse neutrophils were purified from bone marrow cell suspensions as previously described.25  Bone marrow cells were incubated with 30 μl of antibody cocktail specific to the cell surface markers F4/80, CD4, CD45R, CD5, and TER119 for 15 min at 4°C. Anti-biotin tetrameric antibody complexes (100 μl) were then added to the cells and incubated for 15 min at 4°C followed by incubation with 60 μl of colloidal magnetic dextran iron particles for 15 min at 4°C. The cell suspension was then placed into a column surrounded by a magnet. The T cells, B cells, erythrocytes, monocytes, and macrophages were captured in the column, allowing the neutrophils to pass through as a result of negative selection. Cells were then washed with Roswell Park Memorial Institute 1640 with 5% fetal bovine serum. Neutrophil purity, as determined by Wright–Giemsa-stained cytospin preparations, was consistently greater than 98%.

Preparation of P. aeruginosa

The wild-type PAK strain of P. aeruginosa and the PAK strain labeled with the green fluorescent protein (PAK-GFP) were a kind gift from Dr. Stephen Lory, Ph.D., Professor, Department of Microbiology and Molecular Genetics at Harvard Medical School, Boston Massachusetts. For each experiment, frozen bacteria were inoculated into Luria–Bertani broth (Invitrogen, Carlsbad, CA), incubated for 6 h at 37°C on a rotating platform, and then diluted 1:100 in fresh Luria–Bertani broth. After 16 to 18 h of incubation at 37°C, the stationary-phase bacteria were pelleted, washed three times in phosphate-buffered saline (PBS), and suspended in PBS to a concentration adjusted by optical density at 600 nm, as 1 × 109 colony-forming units (CFU)/ml for in vitro experiments or 2 × 108 CFU/ml for instillation in mice (50 μl preparation per mouse).

Bacterial Killing Assay

The bacterial killing assay was performed, as we have previously published.26  MH-S cells, a murine alveolar macrophage cell line (1 × 106 cells), were heat shocked at 43°C for 30 min, then recovered at 37°C for 1 h before adding 107 CFU/ml of P. aeruginosa PAK for 45 min at 37°C. Gentamicin (150 μg/ml) was added and cells were incubated for 1 or 3 h at 37°C. The media were removed, the cells washed twice with sterile PBS, then lysed by adding 200 μl of hypotonic buffer (pH 7.2), and then incubated on ice for 10 min. After adding 800 μl of sterile water, the cell suspension was serially diluted onto agar plates, incubated for 24 h at 37°C, and the resulting colonies counted. Percent killing of intracellular bacteria was calculated using the following equation: percent killing = (1 − number of bacteria after 3 h/number of bacteria after 1 h) × 100. Preliminary data indicate that lysates from uninfected macrophages did not reveal any bacterial colonies on Luria–Bertani agar plates and that bacteria alone incubated for 1 h with gentamicin were completely killed (data not shown). In some of the experiments, MH-S cells were pretreated with a mouse IL-10-blocking antibody or its isotype control antibody 1 h before undergoing the HSR and subsequent exposure to P. aeruginosa PAK bacteria.

Neutrophil Phagocytosis Assay

The neutrophil phagocytosis assay was performed, as previously published.27  Neutrophils were isolated, as described above. Neutrophils (106 cells) kept in Roswell Park Memorial Institute 1640 were exposed to 42°C for 30 min and recovered at 37°C for 1 h. Control neutrophils were kept at 37°C for 90 min. Next, neutrophils were exposed to P. aeruginosa PAK labeled with PAK-GFP at a multiplicity of infection 80 for 60 min. In some experiments, neutrophils were pretreated with a mouse IL-10-blocking antibody or its isotype control antibody 1 h before undergoing the HSR and subsequent exposure to P. aeruginosa PAK bacteria. Then, the cells were fixed, permeabilized, and stained with phalloidin at a concentration of 1:200 for 10 min. Cells were viewed under a LEICA fluorescent microscope (Leica Microsystems, Inc., Buffalo Grove, IL) and 200 cells were randomly counted three times to determine the percentage of cells containing PAK-GFP (done over three independent experiments).

Measurement of Mouse IL-10

Interleukin-10 levels in cell supernatants were measured by enzyme-linked immunosorbent assay. In brief, MH-S cells were heat shocked at 43°C or kept at 37°C for 30 min, then recovered at 37°C for 1 h. Then, 107 CFU/ml of P. aeruginosa PAK or its vehicle were added to the cell medium and the cells were kept at 37°C for 3 h. Cell supernatant was then collected and the enzyme-linked immunosorbent assay was carried out according to the manufacturer’s protocol (R&D Systems Inc.).

Lung Myeloperoxidase Measurement

Lungs were isolated and quickly frozen into liquid nitrogen. Lungs were kept at −80°C until used. Lung homogenization was performed using a tissue homogenizer (Tissue tearor model 985-370; Biospec Products, Inc., Racine, WI) with the lysis buffer and the protease inhibitor provided by the company (mouse myeloperoxidase kit HK210 from Cell Sciences).

Western Blot

Western blot analyses of frozen lungs were performed as we described previously.28  After equal amounts of protein were loaded in each lane and separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis, the proteins were transferred to Invitrogen iBlot polyvinylidene difluoride membranes (Invitrogen, Grand Island, NY). The membranes were blocked for 1 h with Odyssey blocking buffer (LI-COR Biosciences, Lincoln, NE), which was also used as primary and secondary antibodies incubation buffer. The primary antibody was used at dilutions of 1:1,000, incubated overnight at 4°C. Near-infrared detection was used with the IRDye®-conjugated secondary antibodies (LI-COR Biosciences), which were either goat anti-mouse IRDye® 800CW used at 1:10,000 dilution and imaged at 84-μm resolution with the Odyssey infrared imaging system (LI-COR Biosciences). Quantification was performed with the LI-COR Biosciences analysis software.

Hsp72 Detection in the Mouse Lung

Twenty-four hours after the whole-body hyperthermia or the last administration of geldanamycin (1 μg/g body weight intraperitoneally at 48 and 24 h before) or its vehicle (dimethyl sulfoxide), the mice were sacrificed by aortotomy and the lungs were harvested and frozen at −80°C. The expression of Hsp72 protein in lungs was determined by Western blotting.

Cell Viability Assay

Cell viability (MH-S cells, mouse neutrophils) was measured by the Alamar Blue assay after exposure to the various experimental conditions. Cell media were replaced with medium containing 10% Alamar Blue and placed at 37°C in a cell incubator for 2 h. The media were collected and read on a spectrophotometric plate reader at 530 nm.

Mice Studies

C57BL/6 mice were purchased from Charles River Laboratories (Wilmington, MA) for all experiments except for the inducible Hsp72- and IL-10-null mice. IL-10-null mice and their control C57BL/6J mice were purchased from Jackson Laboratory (Bar Harbor, ME). Inducible hsp72 (Hsp72-1) knockout mice (B6;129S7-Hspa1a/Hspa1btm1Dix/Mmcd) and their control C57BL/6J mice were purchased from the Mutant Mouse Resource Center (University of California, Davis, CA). Mice were housed in either the University of California San Francisco or University of Alabama at Birmingham Animal Care Facilities. All use of animals was according to the protocols approved by the University of California San Francisco (San Francisco, CA) and University of Alabama at Birmingham (Birmingham, AL) Animal Care and Use Committees and were conducted according to the National Institutes of Health Guide for Care and Use of Laboratory Animals.

Pneumonia Model

The mouse pneumonia model was performed, as we have previously reported.26  In brief, mice were anesthetized with tribromoethanol (250 mg/kg, intraperitoneally). The mouse was laid on a board with its head elevated at 45°. Then, 50 μl of PBS containing 1 × 107 CFU of P. aeruginosa PAK was instilled into both lungs through the trachea via the mouth by using a 27-gauge gavage needle. The mouse was allowed to recover for 15 min before being returned to the cage. Mice were active and appeared normal after 30 min. Four hours after the bacterial instillation, mice were euthanized with a larger dose of tribromoethanol (500 mg/kg, intraperitoneally). Blood samples were collected in a sterile manner through puncture of the inferior vena cava after laparotomy and bilateral thoracotomies were performed. The mouse lungs were removed, weighed, and homogenized for lung vascular permeability measurements. Bacterial concentration was determined by quantitative culture of homogenized lung, blood, and spleen tissue. Some mice were injected with geldanamycin or its vehicle (1 μg/g body weight intraperitoneally) 48 and 24 h before the airspace instillation of P. aeruginosa PAK. Pilot studies demonstrated that geldanamycin did not affect the viability of P. aeruginosa PAK (data not shown). For the survival studies, all mice were checked every hour during the 24 h after the instillation P. aeruginosa PAK bacteria into the lung until death or survival at 24 h.

Whole-body Hyperthermia in Mice

Before the treatments, the animals were stabilized at room temperature (25° ± 1°C). Whole-body hyperthermia was performed on a heating pad, with the use of a warming lamp, as we have previously reported.12  Animals were anesthetized and maintained throughout the operation by injection of 2.5% tribromoethanol saline (20 μl/g body mass; Sigma-Aldrich). The animals were taped onto the heating pad and a rectal thermostat probe was inserted. The temperature was maintained at 42° ± 0.2°C for 20 min. During the operation, room temperature was maintained at 25° ± 1°C. The sham control group received tribromoethanol treatment but no preheat treatment. After a 24-h recovery period with free access to food and water, P. aeruginosa PAK bacteria were instilled into the trachea, as described in the Materials and Methods section. We have previously reported in several studies that the inhibition of the innate immunity starts within hours after activation of the HSR10  and is present at least up to 48 h after onset of heat shock.24  In the current study, we used the in vivo protocol (exposure to bacteria 24 h after onset of the HSR) that we have previously shown to be protective effect against the sterile inflammation associated with hemorrhagic shock.3 

Lung Vascular Permeability Measurement

Lung endothelial permeability to protein (%) and excess lung water (μl) was measured, as previously described.26  In brief, 0.5 μCi of 125I-albumin was injected intraperitoneally 2 h before sacrificing the animals to ensure adequate tracer distribution. The blood was collected through puncture of the inferior vena cava. Then, the lungs were removed, counted in a Wizard γ-counter (Perkin-Elmer, Waltham, MA), weighed, and homogenized. The homogenate was weighed and a fraction centrifuged (12,000g, 8 min) to assay the hemoglobin concentration in the supernatant. Another fraction of homogenate, supernatant, and blood were weighed and then dried in an oven (60°C for 48 h) for gravimetric determination of the extravascular lung water. The lung wet-to-dry weight ratio (lung W/D ratio) was determined by the following standard formula:

formula

Endothelial permeability was calculated as the counts of 125I-albumin in the blood-free lung tissue divided by the counts of 125I-albumin in the plasma.

Bacteria Cultures from the Lungs

Lungs were collected in a sterile manner. The lungs were homogenized in sterile containers and the homogenates were serially diluted and plated in triplicate on sheep blood agar plates.

Bronchoalveolar Lavage, Cell Count, and Protein Measurement

Bronchoalveolar lavage fluid was collected by infusing 1 ml of sterile PBS (containing 5 mM EDTA) into the lungs of the mice after tracheal cannulation, as previously described.26  Gentle suction was applied and approximately 85% of the fluid was withdrawn from the lungs. The collected fluid was centrifuged at 6,000 rpm for 5 min. One hundred microliter of the supernatant was immediately used for cytospin preparation. Cytospin preparations were made on glass slides, and differential cell counts were performed by two independent operators using Diff-Quik-stained slides. Mean counts from duplicate slides were obtained and expressed as the number of cells per microliter of bronchoalveolar lavage fluid recovered (×104/ml bronchoalveolar lavage fluid). The remaining supernatant was stored immediately at −80°C for protein measurement.

Statistical Analysis

All data are summarized as mean ± SD. For the statistical analysis, we used Statview 5.0® (SAS Inc., Cary, NC) and MedCalc® 7.2.0.2 (MedCalc Software Inc., Ostend, Belgium). The normal distribution was verified using the Agostino–Pearson test. For normally distributed data, Student t test, one-way ANOVA, and Fisher protected least significant difference for post hoc comparisons were used to determine differences between experimental and control groups. To compare data that were not normally distributed, the Wilcoxon–Mann–Whitney two-sample rank sum test was used. All statistical comparison of means was bilateral (two-tailed tests). A Kaplan–Meier analysis followed by a log rank (Mantel-Cox) test was used to compare the survival between the four experimental groups of mice at 24 h. A P value of less than 0.05 was considered statistically significant.

Prior activation of the HSR by whole-body hyperthermia or geldanamycin increases lung injury in a mouse model of P. aeruginosa pneumonia. We first verified that both whole-body hyperthermia and geldanamycin, which is a benzoquinone ansamycin antibiotic and a pharmacologic activator of the HSR,29  cause a significant increase in the expression of the inducible heat-shock protein Hsp72 in the mouse lung compared with controls without whole-body hyperthermia or vehicle (fig. 1, A and B). This result confirms that the Hsp72 response is due to a HSR and not merely a response to hyperthermia. Then, we tested the hypothesis that prior activation of the HSR would worsen lung injury caused by P. aeruginosa PAK. Airspace instillation of P. aeruginosa caused the development of pulmonary edema in mice which was significantly increased (as evidenced by an increase in excess lung water) by prior activation of the HSR with whole-body hyperthermia or geldanamycin (fig. 2, A–D). The increase in severity of pulmonary edema was associated with a decrease in pulmonary bacterial clearance (fig. 3A) and an increase in alveolar epithelial permeability to protein (fig. 3B), despite an increase in total cells and neutrophil recruitment into the airspaces (fig. 4, A and B). In accordance with these results, whole-lung myeloperoxidase activity (as marker of polymorphonuclear leukocytes recruitment) was strongly increased after PAK-induced pneumonia and heat shock (fig. 4C). Mice that underwent HSR and airspace instillation with vehicle 24 h later did not have any bacterial growth from their lung homogenates (data not shown). Because it has previously been demonstrated that activation of the HSR inhibits phagocytosis by neutrophils,30  we next examined whether prior heat shock would decrease phagocytosis of P. aeruginosa PAK labeled with GFP. Our results reveal that phagocytosis of PAK-GFP was decreased by 70% in heat-shocked neutrophils compared with that in control cells (fig. 5, A and B). Furthermore, heat shock did not cause mouse neutrophil death as measured by the Alamar Blue assay (data not shown). Taken together, these results demonstrate that activation of the HSR increases lung damage associated with PAK-induced pneumonia secondary to a decrease in the clearance of P. aeruginosa bacteria from the alveolar space.

Fig. 1.

Activation of the heat-shock response by whole-body hyperthermia or geldanamycin leads to an increased expression of the inducible Hsp72 protein in the mouse lung. Lungs from C57BL/6 mice were harvested 24 h in mice that underwent (A) a 20-min whole-body hyperthermia at 42° ± 0.2°C induced by exposure heating pad and warming lamp or were kept at room temperature and (B) a pretreatment with geldanamycin or its vehicle (1 μg/g body weight given intraperitoneally at 48 and 24 h before lung harvesting). One representative lung is shown for each experimental condition.

Fig. 1.

Activation of the heat-shock response by whole-body hyperthermia or geldanamycin leads to an increased expression of the inducible Hsp72 protein in the mouse lung. Lungs from C57BL/6 mice were harvested 24 h in mice that underwent (A) a 20-min whole-body hyperthermia at 42° ± 0.2°C induced by exposure heating pad and warming lamp or were kept at room temperature and (B) a pretreatment with geldanamycin or its vehicle (1 μg/g body weight given intraperitoneally at 48 and 24 h before lung harvesting). One representative lung is shown for each experimental condition.

Close modal
Fig. 2.

Prior activation of the heat-shock response by whole-body hyperthermia or geldanamycin increases lung injury in a mouse model of Pseudomonas aeruginosa pneumonia. (AD) C57BL/6 mice were left untreated or underwent a heat-shock response with whole-body hyperthermia or two intraperitoneal injections of intraperitoneal geldanamycin or its vehicle (1 μg/g body weight). Twenty-four hours later, mice had their airspaces instilled with P. aeruginosa bacteria (PAK strain, 1 × 107 colony-forming units [CFU]) or its vehicle. Mice were euthanized 4 h later and excess lung water (ELW) and lung vascular permeability (extravascular pulmonary edema [EVPE]) were measured, as described in the Materials and Methods section. For all experiments, results are shown as mean ± SD (n = 6 mice per group); *P ≤ 0.05 from controls; **P ≤ 0.05 from mice with PAK alone.

Fig. 2.

Prior activation of the heat-shock response by whole-body hyperthermia or geldanamycin increases lung injury in a mouse model of Pseudomonas aeruginosa pneumonia. (AD) C57BL/6 mice were left untreated or underwent a heat-shock response with whole-body hyperthermia or two intraperitoneal injections of intraperitoneal geldanamycin or its vehicle (1 μg/g body weight). Twenty-four hours later, mice had their airspaces instilled with P. aeruginosa bacteria (PAK strain, 1 × 107 colony-forming units [CFU]) or its vehicle. Mice were euthanized 4 h later and excess lung water (ELW) and lung vascular permeability (extravascular pulmonary edema [EVPE]) were measured, as described in the Materials and Methods section. For all experiments, results are shown as mean ± SD (n = 6 mice per group); *P ≤ 0.05 from controls; **P ≤ 0.05 from mice with PAK alone.

Close modal
Fig. 3.

Prior activation of the heat-shock response by whole-body hyperthermia decreases lung bacterial clearance and increases lung epithelial protein permeability in a mouse model of Pseudomonas aeruginosa pneumonia. (A and B) C57BL/6 mice were left untreated or underwent a heat-shock response with whole-body hyperthermia that was followed 24 h later by airspace instillation of P. aeruginosa (PAK strain, 1 × 107 colony-forming units [CFU]) or its vehicle. Mice were euthanized 4 h later and either bacteria were counted in the lung homogenates or protein concentration was measured in the bronchial alveolar lavage (BAL) fluid. For all experiments, results are shown as mean ± SD (n = 6 mice per group); *P ≤ 0.05 from controls; **P ≤ 0.05 from mice with PAK alone.

Fig. 3.

Prior activation of the heat-shock response by whole-body hyperthermia decreases lung bacterial clearance and increases lung epithelial protein permeability in a mouse model of Pseudomonas aeruginosa pneumonia. (A and B) C57BL/6 mice were left untreated or underwent a heat-shock response with whole-body hyperthermia that was followed 24 h later by airspace instillation of P. aeruginosa (PAK strain, 1 × 107 colony-forming units [CFU]) or its vehicle. Mice were euthanized 4 h later and either bacteria were counted in the lung homogenates or protein concentration was measured in the bronchial alveolar lavage (BAL) fluid. For all experiments, results are shown as mean ± SD (n = 6 mice per group); *P ≤ 0.05 from controls; **P ≤ 0.05 from mice with PAK alone.

Close modal
Fig. 4.

Prior activation of the heat-shock response by whole-body hyperthermia increases total cell and neutrophil recruitment into the airspaces in a mouse model of Pseudomonas aeruginosa pneumonia. (AC) C57BL/6 mice were left untreated or underwent a heat-shock response with whole-body hyperthermia that was followed 24 h later by airspace instillation of P. aeruginosa (PAK strain, 1 × 107 colony-forming units [CFU]) or its vehicle. Mice were euthanized 4 h later and bronchial alveolar lavage (BAL) was performed and cells counted. In separate experiments, 4 h after P. aeruginosa instillation, lungs were removed and frozen in liquid nitrogen for measurement of myeloperoxidase (MPO) activity as described in the Materials and Methods section. For all experiments, results are shown as mean ± SD (n = 6 mice per group); *P ≤ 0.05 from controls; **P ≤ 0.05 from mice with PAK alone. PMN = polymorphonuclear leukocytes.

Fig. 4.

Prior activation of the heat-shock response by whole-body hyperthermia increases total cell and neutrophil recruitment into the airspaces in a mouse model of Pseudomonas aeruginosa pneumonia. (AC) C57BL/6 mice were left untreated or underwent a heat-shock response with whole-body hyperthermia that was followed 24 h later by airspace instillation of P. aeruginosa (PAK strain, 1 × 107 colony-forming units [CFU]) or its vehicle. Mice were euthanized 4 h later and bronchial alveolar lavage (BAL) was performed and cells counted. In separate experiments, 4 h after P. aeruginosa instillation, lungs were removed and frozen in liquid nitrogen for measurement of myeloperoxidase (MPO) activity as described in the Materials and Methods section. For all experiments, results are shown as mean ± SD (n = 6 mice per group); *P ≤ 0.05 from controls; **P ≤ 0.05 from mice with PAK alone. PMN = polymorphonuclear leukocytes.

Close modal
Fig. 5.

Activation of the heat-shock response inhibits phagocytosis of Pseudomonas aeruginosa by mouse neutrophils. (A) Primary mouse neutrophils were isolated, heat-shocked at 42°C for 30 min, and then exposed 60 min later to P. aeruginosa expressing green fluorescent protein (PAK-GFP) for 30 min. After cell fixation, permeabilization, and staining with phalloidin, the number of intracellular PAK-GFP was counted, as described in the Materials and Methods section. Results are shown as mean ± SD (n = 3 experiments repeated in triplicate); *P ≤ 0.05 from non–heat-shocked cells. (B) This provides an example of the inhibition of PAK-GFP bacteria phagocytosis by primary mouse neutrophils that underwent prior heat shock activation. Please note that although PAK-GFP bacteria are stuck to the outside of heat-shocked neutrophils, they have not been endocytosed.

Fig. 5.

Activation of the heat-shock response inhibits phagocytosis of Pseudomonas aeruginosa by mouse neutrophils. (A) Primary mouse neutrophils were isolated, heat-shocked at 42°C for 30 min, and then exposed 60 min later to P. aeruginosa expressing green fluorescent protein (PAK-GFP) for 30 min. After cell fixation, permeabilization, and staining with phalloidin, the number of intracellular PAK-GFP was counted, as described in the Materials and Methods section. Results are shown as mean ± SD (n = 3 experiments repeated in triplicate); *P ≤ 0.05 from non–heat-shocked cells. (B) This provides an example of the inhibition of PAK-GFP bacteria phagocytosis by primary mouse neutrophils that underwent prior heat shock activation. Please note that although PAK-GFP bacteria are stuck to the outside of heat-shocked neutrophils, they have not been endocytosed.

Close modal

Exacerbation of P. aeruginosa–induced lung injury and mortality by prior activation of the HSR is reduced by the genetic deletion of the inducible Hsp72 protein. Prior experimental work indicates that inducible Hsp72 is one of the most important heat-shock proteins expressed during HSR activation12  and plays an important role in the attenuation of the inflammatory response induced by immune cells.24  Therefore, we tested the hypothesis that genetic deletion of Hsp72 would attenuate the exacerbation of P. aeruginosa–induced lung injury and mortality caused by prior HSR activation. Genetic deletion of Hsp72 was associated with suppression of the heat-shock–induced increase in lung vascular permeability (fig. 6, A and B), decrease in lung bacterial clearance (fig. 6C), and development of alveolar edema (fig. 6D). In addition, this strategy was associated with a significant increase in survival of mice subjected to heat shock followed 24 h later by airspace instillation with P. aeruginosa PAK although it did not affect the survival of sham mice that had similar airspace instillation with P. aeruginosa PAK (fig. 7).

Fig. 6.

Genetic deletion of Hsp72 attenuates the effect of the heat-shock response on lung vascular permeability and bacterial clearance in a mouse model of Pseudomonas aeruginosa pneumonia. (AD) C57BL/6 wild-type (WT) or Hsp72-null mice were left untreated or underwent a heat-shock response with whole-body hyperthermia that was followed 24 h later by airspace instillation of P. aeruginosa (PAK strain, 1 × 107 colony-forming units [CFU]) or its vehicle. Mice were euthanized 4 h later and excess lung water (ELW) and lung vascular protein permeability (EVPE) were measured, as described in the Materials and Methods section. In separate experiments, bronchial alveolar lavage (BAL) was performed and cells counted and BAL fluid protein concentration measured. For all experiments, results are shown as mean ± SD (n = 6 mice per group); *P ≤ 0.05 from controls, **P≤ 0.05 from mice with PAK alone.

Fig. 6.

Genetic deletion of Hsp72 attenuates the effect of the heat-shock response on lung vascular permeability and bacterial clearance in a mouse model of Pseudomonas aeruginosa pneumonia. (AD) C57BL/6 wild-type (WT) or Hsp72-null mice were left untreated or underwent a heat-shock response with whole-body hyperthermia that was followed 24 h later by airspace instillation of P. aeruginosa (PAK strain, 1 × 107 colony-forming units [CFU]) or its vehicle. Mice were euthanized 4 h later and excess lung water (ELW) and lung vascular protein permeability (EVPE) were measured, as described in the Materials and Methods section. In separate experiments, bronchial alveolar lavage (BAL) was performed and cells counted and BAL fluid protein concentration measured. For all experiments, results are shown as mean ± SD (n = 6 mice per group); *P ≤ 0.05 from controls, **P≤ 0.05 from mice with PAK alone.

Close modal
Fig. 7.

Genetic deletion of Hsp72 attenuates the increased mortality associated with a prior heat-shock response in a mouse model of Pseudomonas aeruginosa pneumonia. C57BL/6 wild-type (WT) or Hsp72-null mice were left untreated or underwent a heat-shock response with whole-body hyperthermia that was followed 24 h later by airspace instillation of P. aeruginosa (PAK strain, 1 × 107 colony-forming units [CFU]) or its vehicle. Kaplan–Meier survival analysis was performed (n = 10 mice per group). WT but not Hsp72-null mice that underwent a heat-shock response and had their airspaces instilled with P. aeruginosa PAK died significantly earlier (*P = 0.05) than the WT mice that did not undergo a heat-shock response before the onset of P. aeruginosa pneumonia.

Fig. 7.

Genetic deletion of Hsp72 attenuates the increased mortality associated with a prior heat-shock response in a mouse model of Pseudomonas aeruginosa pneumonia. C57BL/6 wild-type (WT) or Hsp72-null mice were left untreated or underwent a heat-shock response with whole-body hyperthermia that was followed 24 h later by airspace instillation of P. aeruginosa (PAK strain, 1 × 107 colony-forming units [CFU]) or its vehicle. Kaplan–Meier survival analysis was performed (n = 10 mice per group). WT but not Hsp72-null mice that underwent a heat-shock response and had their airspaces instilled with P. aeruginosa PAK died significantly earlier (*P = 0.05) than the WT mice that did not undergo a heat-shock response before the onset of P. aeruginosa pneumonia.

Close modal

Exacerbation of P. aeruginosa–induced lung injury and mortality by prior activation of the HSR is mediated by an IL-10-dependent mechanism. Previous studies have reported that IL-10 inhibits bacterial clearance from the lung in a mouse model of P. aeruginosa–induced bacterial pneumonia.13–21  Furthermore, IL-10 protein expression can be induced in response to activation of the HSR.22,23  Thus, in the next series of experiments, we tested the hypothesis that prior activation of the HSR would exacerbate P. aeruginosa–induced lung injury and mortality via an IL-10-dependent mechanism. We first found that prior heat shock increased by 50% the release of IL-10 by MH-S cells, a mouse alveolar macrophage cell line (fig. 8A). Heat shock did not cause MH-S cell death as measured by the Alamar Blue assay (data not shown). Furthermore, heat-shocked MH-S cells that were subsequently exposed to P. aeruginosa PAK bacteria released significantly more IL-10 compared with IL-10 released by the cells exposed to the P. aeruginosa but kept at 37°C (fig. 8A). We next demonstrated that the decrease in intracellular bacterial killing observed in heat-shocked MH-S cells was dependent on IL-10, because it was blocked by pretreatment with a specific blocking mouse IL-10 antibody, but not by its isotype control antibody (fig. 8B). In the next series of experiments, we found that exposure to increasing doses of mouse IL-10 (0.1 to 10 ng/ml) caused a progressive decrease in the phagocytosis of P. aeruginosa labeled with the PAK-GFP by mouse neutrophils (fig. 8C). Furthermore, HSR-mediated inhibition of neutrophil phagocytosis of P. aeruginosa bacteria was completely inhibited by pretreatment with IL-10-blocking antibody, but not with an isotype control antibody (fig. 8D). Interestingly, pretreatment with IL-10 (10 ng/ml) did not further decrease the uptake of P. aeruginosa bacteria by heat-shocked neutrophils (fig. 8D). Finally, the results of these in vitro experiments were confirmed in our in vivo mouse model of heat shock and subsequent P. aeruginosa pneumonia. Decreased lung bacterial clearance and excess of 24-h mortality caused by P. aeruginosa pneumonia in heat-shocked wild-type mice were significantly attenuated in IL-10-null mice (fig. 8, E and F). Taking together, these results demonstrate an important mechanistic role for IL-10 in decreasing lung bacterial clearance by inhibiting intracellular P. aeruginosa killing by macrophages and bacterial phagocytosis by neutrophils and thus increasing mortality after activation of the HSR in a mouse model of P. aeruginosa pneumonia.

Fig. 8.

Increased interleukin (IL)-10 release induced by the activation of the heat-shock response is associated with a decreased intracellular killing by alveolar macrophages, decreased bacterial phagocytosis by neutrophils, and increased mortality in a mouse model of Pseudomonas aeruginosa pneumonia. (A) MH-S cells, a mouse alveolar macrophage cell line (1 × 106 cells), were left untreated or heat shocked with heat for 30 min at 43°C, then exposed 1 h later to P. aeruginosa PAK (2 × 107 colony-forming units [CFU]/ml; multiplicity of infection [MOI] = 10:1) or its vehicle for 4 h. Enzyme-linked immunosorbent assay of IL-10 was carried out according to the manufacturer’s protocol. Results are shown as mean ± SD (n = 5 experiments repeated in triplicate); *P ≤ 0.05 from control cells; **P ≤ 0.05 from cells treated with PAK alone. (B) MH-S cells were heat shocked at 43°C for 30 min or kept at 37°C, then recovered at 37°C for 1 h before adding 107 CFU/ml of P. aeruginosa PAK for 45 min at 37°C. Gentamicin (150 μg/ml) was added and cells were incubated for 1 or 3 h at 37°C. After cell lysis, percent of intracellular bacterial killing was calculated, as described in the Materials and Methods section. In some experiments, MH-S cells were pretreated with a mouse IL-10-blocking antibody or its isotype control antibody 1 h before undergoing the heat-shock response and subsequent exposure to P. aeruginosa bacteria. Results are shown as mean ± SD (n = 5 experiments repeated in triplicate); *P ≤ 0.05 from non–heat-shocked cells; **P ≤ 0.05 from heat-shocked cells pretreated with an isotype control antibody. (C) Primary mouse neutrophils were isolated, exposed to increasing concentrations of mouse IL-10 (0.1–10 ng/ml), then exposed 60 min later to P. aeruginosa expressing green fluorescent protein (PAK-GFP) for 30 min. After cell fixation, permeabilization, and staining with phalloidin, the number of intracellular PAK-GFP was counted, as described in the Materials and Methods section. Results are shown as mean ± SD (n = 3 experiments repeated in triplicate); *P ≤ 0.05 from neutrophils exposed to IL-10 vehicle. (D) Primary mouse neutrophils were isolated as described in the Materials and Method section. Cells were heat shocked at 43°C for 30 min or kept at 37°C, then exposed 60 min later to P. aeruginosa PAK-GFP for 30 min. After cell fixation, permeabilization, and staining with phalloidin, the number of intracellular PAK-GFP was counted, as described in the Materials and Methods section. Results are shown as mean ± SD (n = 3 experiments repeated in triplicate); *P ≤ 0.05 from non heat-shocked cells; **P ≤ 0.05 from heat-shocked cells pretreated with an isotype control antibody. (E and F) C57BL/6 wild-type (WT) or IL-10-null mice underwent a heat-shock response with whole-body hyperthermia that was followed 24 h later by airspace instillation of P. aeruginosa (PAK strain, 1 × 107 CFU) or its vehicle, as described in the Materials and Method section. Number of live bacteria in the lung homogenates and mortality at 24 h are reported. Results are shown as mean ± SD (n = 6 mice per group for lung bacterial clearance and n = 8 mice per group for mortality); *P ≤ 0.05 from non–heat-shocked mice, **P ≤ 0.05 from wild-type mice.

Fig. 8.

Increased interleukin (IL)-10 release induced by the activation of the heat-shock response is associated with a decreased intracellular killing by alveolar macrophages, decreased bacterial phagocytosis by neutrophils, and increased mortality in a mouse model of Pseudomonas aeruginosa pneumonia. (A) MH-S cells, a mouse alveolar macrophage cell line (1 × 106 cells), were left untreated or heat shocked with heat for 30 min at 43°C, then exposed 1 h later to P. aeruginosa PAK (2 × 107 colony-forming units [CFU]/ml; multiplicity of infection [MOI] = 10:1) or its vehicle for 4 h. Enzyme-linked immunosorbent assay of IL-10 was carried out according to the manufacturer’s protocol. Results are shown as mean ± SD (n = 5 experiments repeated in triplicate); *P ≤ 0.05 from control cells; **P ≤ 0.05 from cells treated with PAK alone. (B) MH-S cells were heat shocked at 43°C for 30 min or kept at 37°C, then recovered at 37°C for 1 h before adding 107 CFU/ml of P. aeruginosa PAK for 45 min at 37°C. Gentamicin (150 μg/ml) was added and cells were incubated for 1 or 3 h at 37°C. After cell lysis, percent of intracellular bacterial killing was calculated, as described in the Materials and Methods section. In some experiments, MH-S cells were pretreated with a mouse IL-10-blocking antibody or its isotype control antibody 1 h before undergoing the heat-shock response and subsequent exposure to P. aeruginosa bacteria. Results are shown as mean ± SD (n = 5 experiments repeated in triplicate); *P ≤ 0.05 from non–heat-shocked cells; **P ≤ 0.05 from heat-shocked cells pretreated with an isotype control antibody. (C) Primary mouse neutrophils were isolated, exposed to increasing concentrations of mouse IL-10 (0.1–10 ng/ml), then exposed 60 min later to P. aeruginosa expressing green fluorescent protein (PAK-GFP) for 30 min. After cell fixation, permeabilization, and staining with phalloidin, the number of intracellular PAK-GFP was counted, as described in the Materials and Methods section. Results are shown as mean ± SD (n = 3 experiments repeated in triplicate); *P ≤ 0.05 from neutrophils exposed to IL-10 vehicle. (D) Primary mouse neutrophils were isolated as described in the Materials and Method section. Cells were heat shocked at 43°C for 30 min or kept at 37°C, then exposed 60 min later to P. aeruginosa PAK-GFP for 30 min. After cell fixation, permeabilization, and staining with phalloidin, the number of intracellular PAK-GFP was counted, as described in the Materials and Methods section. Results are shown as mean ± SD (n = 3 experiments repeated in triplicate); *P ≤ 0.05 from non heat-shocked cells; **P ≤ 0.05 from heat-shocked cells pretreated with an isotype control antibody. (E and F) C57BL/6 wild-type (WT) or IL-10-null mice underwent a heat-shock response with whole-body hyperthermia that was followed 24 h later by airspace instillation of P. aeruginosa (PAK strain, 1 × 107 CFU) or its vehicle, as described in the Materials and Method section. Number of live bacteria in the lung homogenates and mortality at 24 h are reported. Results are shown as mean ± SD (n = 6 mice per group for lung bacterial clearance and n = 8 mice per group for mortality); *P ≤ 0.05 from non–heat-shocked mice, **P ≤ 0.05 from wild-type mice.

Close modal

In this study, we demonstrate that (1) prior HSR activation worsens the development of protein-rich pulmonary edema, decreased lung bacterial clearance, and increased mortality associated with P. aeruginosa pneumonia; (2) this HSR effect was not observed in mice null for the inducible Hsp72, one of the most important heat-shock proteins expressed during HSR activation,12  thus confirming the specificity of this observation; and (3) IL-10 plays an important mechanistic role in mediating HSR-mediated decrease in bacterial killing by MH-S cells, which is a mouse lung macrophage cell line, and decrease in neutrophil bacterial phagocytosis. These in vitro results were confirmed by the increased lung bacterial clearance and decreased mortality caused by P. aeruginosa pneumonia in heat-shocked IL-10-null mice.

Heat-shock response activation has been shown to attenuate cell and organ response to inflammatory stimuli induced by several pathological conditions, particularly associated with sterile inflammation.2–5  However, the effect of HSR activation on the innate and adaptive immune response to lung bacterial infection remains unknown. We found in the current study that HSR activation by heat or a pharmacologic inhibitor of Hsp90 increased lung injury caused by P. aeruginosa bacteria in mice. These results are in accordance with the fact that HSR activation inhibits cellular pathways, such as the nuclear factor-κB pathway,10  that are activated by cell membrane receptors and are critical for the eradication of bacteria from the airspaces of the lung.11  Interestingly, HSR activation inhibited lung bacterial clearance despite an increase in the number of neutrophils migrating into the airspaces of the lungs in response to the bacterial challenge. Previous work has demonstrated that there are functional heat-shock elements on the promoter of IL-8, a critical neutrophil chemokine, that result in an increased protein expression of this chemokine.31,32  However, the increased number of airspace neutrophils in the heat-shocked mice that had their airspaces instilled with P. aeruginosa was not associated with an increase, but with a decrease in bacterial clearance from the lung of these animals. It has been previously reported that HSR activation inhibits neutrophil phagocytosis.30  We also found in our study that HSR activation decreased the ability of neutrophils to phagocyte P. aeruginosa bacteria by 70% (fig. 5).

The next set of experiments demonstrated that the HSR inhibition of the bacterial clearance after a lung challenge with P. aeruginosa was specific to the HSR because Hsp72-null mice did not show the increase in lung injury and inhibition of lung bacterial clearance associated with HSR in their wild-type littermates. These in vivo results are in accordance with previous experimental work that has shown that intracellular Hsp72 directly inhibits the nuclear factor-κB pathway33–35  which is critical for the eradication of P. aeruginosa bacteria.11  It should be pointed out that HSR activation might not be detrimental in established sepsis. Indeed, if a brisk inflammatory response to infecting organism is essential for pathogen containment and eradication. However, systemic spread of inflammation beyond the focus of bacterial infection leads to organ injury and mortality.36  Thus, it is not surprising that HSR activation with Hsp90 inhibitor, such as a derivate of the geldanamycin used in our study prolonged survival, attenuate systemic inflammation and reduce lung injury in murine sepsis37  In addition, the results of clinical studies confirm that patients with a genetic variation on Hsp72 that causes low expression of that protein in mononuclear cells have higher risk for septic shock.38,39  Taken together, our data and the results of previous studies indicate that prior induction of the HSR increases lung damage caused by bacterial pneumonia because of the HSR-mediated decrease in bacterial clearance from the airspaces of the lung. Furthermore, genetic deletion of Hsp72 completely reverses the deleterious effect of HSR activation on lung injury caused by P. aeruginosa bacteria. This result demonstrates that the increased lung expression of Hsp72 associated with HSR activation may regulate the intensity of the inflammatory response to bacterial products within the lung. On the basis of the results of previous work, it also suggests that in the presence of established systemic inflammatory response secondary to sepsis, Hsp72 may attenuate a systemic exuberant inflammatory response by the host that may cause end-organ damage.

The second important aim of the study was to determine potential mechanisms that could explain the inhibitory effect of HSR activation on the bacterial clearance by immune cells. For example, previous work has shown that the initial response to the endogenous release or exogenous administration of antiinflammatory mediators, such as IL-10, is associated with more severe lung injury caused by bacterial pneumonia.13–21  Furthermore, the heat-shock factor 1 released during HSR activation is a transcriptional activator of the IL-10 gene expression in macrophages22  and the fact that extracellular Hsp70 causes the release of IL-10 via the activation of the toll-like receptor-423  and enhances immunosuppressive function of CD4+CD25+FoxP3+ T regulatory cells that release IL-10.40  In the current study, heat-shocked alveolar macrophages released more IL-10 than that released by the cells kept at 37°C. Furthermore, HSR activation inhibited intracellular killing of P. aeruginosa bacteria by alveolar macrophages via an IL-10-dependent mechanism despite the fact that it is known that HSR activation and Hsp70 both increase phagocytosis by these cells.41–43  These results are in accordance with previous work from our laboratory that has shown that macrophages play an important role in the airspace clearance of P. aeruginosa.44,45  Furthermore, exposure of alveolar macrophages to P. aeruginosa caused the release of IL-10 that impaired the ability to kill this bacterium, and this defect was partially reversed by neutralization of that cytokine.15 

Does HSR activation affect phagocytosis and intracellular killing of P. aeruginosa by neutrophils? Neutrophils are critical for the eradication of P. aeruginosa from the lungs.8  Furthermore, a low airspace bacterial inoculum is cleared more efficiently than a larger one, suggesting that granulocyte killing of P. aeruginosa is saturable within the airspaces of the lungs.46  Interestingly, despite an increase in neutrophil migration into the lungs induced by HSR activation, lung bacterial clearance was reduced in heat-shocked mice. This apparent paradox may in part be explained by the fact that HSR activation has been shown to inhibit neutrophil phagocytosis.30  We found a significant 70% inhibition of P. aeruginosa phagocytosis caused by HSR activation in neutrophils, an effect blocked by pretreatment with an IL-10-blocking antibody. Furthermore, we also found that exposure of mouse neutrophils to low concentrations of IL-10 (5 to 10 ng/ml) resulted in the comparable inhibition of the phagocytosis of live P. aeruginosa bacteria. These results are in accordance with previous work that has shown that IL-10 inhibits neutrophil phagocytic activity of Escherichia coli and Staphylococcus aureus bacteria in vitro.47,48  Our in vitro results were confirmed in our in vivo mouse model of heat shock and subsequent P. aeruginosa pneumonia. Decreased lung bacterial clearance and excess of 24 h mortality caused by P. aeruginosa pneumonia in heat-shocked wild-type mice were attenuated in IL-10-null mice. Taken together, our data and the results of previous studies indicate an important mechanistic role for IL-10 in mediating the inhibitory effect of HSR activation on the bacterial clearance by immune cells.

The current study has several limitations. First, we examined here the HSR effect on bacterial pneumonia caused by one bacterium, P. aeruginosa. Previous work has shown that the host response to bacterial pneumonia is organism dependent.49  Thus, additional work will be needed to determine whether the effect of HSR is comparable in experimental models of bacterial pneumonia induced by other microorganisms. Second, we only examined the effect of HSR on bacterial eradication by alveolar macrophages and neutrophils. We did not examine the effect of HSR activation on several other immune cells that participate in the elimination of P. aeruginosa from the distal airspaces of the lungs. In particular, intratracheal dendritic cells appear to modulate the balance between IL-12 and IL-10 expression in response to P. aeruginosa pneumonia in mice.50  Furthermore, Rag2-null mice deficient in lymphocytes showed impaired bacterial clearance in response to P. aeruginosa pneumonia51  although it is known that HSR activation enhances immunosuppressive function of CD4+CD25+FoxP3+ T regulatory cells that release IL-10.40  Finally, although our data indicate an important mechanistic role of IL-10 to explain the impaired lung bacterial clearance associated with HSR activation in our experimental model of P. aeruginosa pneumonia, IL-10 release after the onset of bacterial infection may play an important function to attenuate parenchymal tissue destruction when the pathogen elicits a strong local and systemic inflammatory response (reviewed in the study by Cyktor and Turner52 ).

In summary, our studies demonstrate that HSR activation significantly increases lung injury caused by P. aeruginosa in mice. This effect is mediated in large part by IL-10-dependent mechanisms that inhibit airspace clearance of P. aeruginosa bacteria by alveolar macrophages and neutrophils. These results also suggest that if HSR activation may have a protective effect early after the onset of sterile inflammation, such as severe trauma,3,12  it may also facilitate the later development of immunosuppression and bacterial lung infection in these patients.

The authors kindly thank Marybeth Howard, Ph.D. (Department of Anesthesiology, University of California San Francisco, San Francisco, California), for her technical assistance on this project.

This study was supported in part by a grant (no. RO1 GM-62188) from the National Institutes of Health (Bethesda, Maryland) (to Dr. Pittet) and a Research Fellowship Grant from the Foundation for Anesthesia Education and Research (Rochester, Minnesota) (to Dr. Wagener).

The authors declare no competing interests.

1.
Asea
A
:
Heat shock proteins and toll-like receptors.
Handb Exp Pharmacol
2008
, pp
111
27
2.
Fernandes
TR
,
Pontieri
V
,
Moretti
AI
,
Teixeira
DO
,
Abatepaulo
F
,
Soriano
FG
,
Negri
EM
,
Velasco
IT
,
Souza
HP
:
Hypertonic saline solution increases the expression of heat shock protein 70 and improves lung inflammation early after reperfusion in a rodent model of controlled hemorrhage.
Shock
2007
;
27
:
172
8
3.
Pittet
JF
,
Lu
LN
,
Geiser
T
,
Lee
H
,
Matthay
MA
,
Welch
WJ
:
Stress preconditioning attenuates oxidative injury to the alveolar epithelium of the lung following haemorrhage in rats.
J Physiol
2002
;
538
(
Pt 2
):
583
97
4.
Lee
H
,
Pespeni
M
,
Roux
J
,
Dennery
PA
,
Matthay
MA
,
Pittet
JF
:
HO-1 induction restores c-AMP-dependent lung epithelial fluid transport following severe hemorrhage in rats.
FASEB J
2005
;
19
:
287
9
5.
Godzich
M
,
Hodnett
M
,
Frank
JA
,
Su
G
,
Pespeni
M
,
Angel
A
,
Howard
MB
,
Matthay
MA
,
Pittet
JF
:
Activation of the stress protein response prevents the development of pulmonary edema by inhibiting VEGF cell signaling in a model of lung ischemia-reperfusion injury in rats.
FASEB J
2006
;
20
:
1519
21
6.
Pittet
JF
,
Lee
H
,
Morabito
D
,
Howard
MB
,
Welch
WJ
,
Mackersie
RC
:
Serum levels of Hsp 72 measured early after trauma correlate with survival.
J Trauma
2002
;
52
:
611
7; discussion 617
7.
Schröder
O
,
Schulte
KM
,
Ostermann
P
,
Röher
HD
,
Ekkernkamp
A
,
Laun
RA
:
Heat shock protein 70 genotypes HSPA1B and HSPA1L influence cytokine concentrations and interfere with outcome after major injury.
Crit Care Med
2003
;
31
:
73
9
8.
Koh
AY
,
Priebe
GP
,
Ray
C
,
Van Rooijen
N
,
Pier
GB
:
Inescapable need for neutrophils as mediators of cellular innate immunity to acute Pseudomonas aeruginosa pneumonia.
Infect Immun
2009
;
77
:
5300
10
9.
Zhuo
H
,
Yang
K
,
Lynch
SV
,
Dotson
RH
,
Glidden
DV
,
Singh
G
,
Webb
WR
,
Elicker
BM
,
Garcia
O
,
Brown
R
,
Sawa
Y
,
Misset
B
,
Wiener-Kronish
JP
:
Increased mortality of ventilated patients with endotracheal Pseudomonas aeruginosa without clinical signs of infection.
Crit Care Med
2008
;
36
:
2495
503
10.
Pittet
JF
,
Lee
H
,
Pespeni
M
,
O’Mahony
A
,
Roux
J
,
Welch
WJ
:
Stress-induced inhibition of the NF-kappaB signaling pathway results from the insolubilization of the IkappaB kinase complex following its dissociation from heat shock protein 90.
J Immunol
2005
;
174
:
384
94
11.
Skerrett
SJ
,
Wilson
CB
,
Liggitt
HD
,
Hajjar
AM
:
Redundant Toll-like receptor signaling in the pulmonary host response to Pseudomonas aeruginosa.
Am J Physiol Lung Cell Mol Physiol
2007
;
292
:
L312
22
12.
Ganter
MT
,
Ware
LB
,
Howard
M
,
Roux
J
,
Gartland
B
,
Matthay
MA
,
Fleshner
M
,
Pittet
JF
:
Extracellular heat shock protein 72 is a marker of the stress protein response in acute lung injury.
Am J Physiol Lung Cell Mol Physiol
2006
;
291
:
L354
61
13.
Sun
L
,
Guo
RF
,
Newstead
MW
,
Standiford
TJ
,
Macariola
DR
,
Shanley
TP
:
Effect of IL-10 on neutrophil recruitment and survival after Pseudomonas aeruginosa challenge.
Am J Respir Cell Mol Biol
2009
;
41
:
76
84
14.
Chmiel
JF
,
Konstan
MW
,
Saadane
A
,
Krenicky
JE
,
Lester Kirchner
H
,
Berger
M
:
Prolonged inflammatory response to acute Pseudomonas challenge in interleukin-10 knockout mice.
Am J Respir Crit Care Med
2002
;
165
:
1176
81
15.
Steinhauser
ML
,
Hogaboam
CM
,
Kunkel
SL
,
Lukacs
NW
,
Strieter
RM
,
Standiford
TJ
:
IL-10 is a major mediator of sepsis-induced impairment in lung antibacterial host defense.
J Immunol
1999
;
162
:
392
9
16.
Penttilä
T
,
Haveri
A
,
Tammiruusu
A
,
Vuola
JM
,
Lahesmaa
R
,
Puolakkainen
M
:
Chlamydia pneumoniae infection in IL-10 knock out mice: Accelerated clearance but severe pulmonary inflammatory response.
Microb Pathog
2008
;
45
:
25
9
17.
Greenberger
MJ
,
Strieter
RM
,
Kunkel
SL
,
Danforth
JM
,
Goodman
RE
,
Standiford
TJ
:
Neutralization of IL-10 increases survival in a murine model of Klebsiella pneumonia.
J Immunol
1995
;
155
:
722
9
18.
van der Sluijs
KF
,
van Elden
LJ
,
Nijhuis
M
,
Schuurman
R
,
Pater
JM
,
Florquin
S
,
Goldman
M
,
Jansen
HM
,
Lutter
R
,
van der Poll
T
:
IL-10 is an important mediator of the enhanced susceptibility to pneumococcal pneumonia after influenza infection.
J Immunol
2004
;
172
:
7603
9
19.
Muenzer
JT
,
Davis
CG
,
Chang
K
,
Schmidt
RE
,
Dunne
WM
,
Coopersmith
CM
,
Hotchkiss
RS
:
Characterization and modulation of the immunosuppressive phase of sepsis.
Infect Immun
2010
;
78
:
1582
92
20.
Reddy
RC
,
Chen
GH
,
Newstead
MW
,
Moore
T
,
Zeng
X
,
Tateda
K
,
Standiford
TJ
:
Alveolar macrophage deactivation in murine septic peritonitis: Role of interleukin 10.
Infect Immun
2001
;
69
:
1394
401
21.
Kalechman
Y
,
Gafter
U
,
Gal
R
,
Rushkin
G
,
Yan
D
,
Albeck
M
,
Sredni
B
:
Anti-IL-10 therapeutic strategy using the immunomodulator AS101 in protecting mice from sepsis-induced death: Dependence on timing of immunomodulating intervention.
J Immunol
2002
;
169
:
384
92
22.
Zhang
H
,
Zhang
L
,
Yu
F
,
Liu
Y
,
Liang
Q
,
Deng
G
,
Chen
G
,
Liu
M
,
Xiao
X
:
HSF1 is a transcriptional activator of IL-10 gene expression in RAW264.7 macrophages.
Inflammation
2012
;
35
:
1558
66
23.
Luo
X
,
Zuo
X
,
Zhang
B
,
Song
L
,
Wei
X
,
Zhou
Y
,
Xiao
X
:
Release of heat shock protein 70 and the effects of extracellular heat shock protein 70 on the production of IL-10 in fibroblast-like synoviocytes.
Cell Stress Chaperones
2008
;
13
:
365
73
24.
Howard
M
,
Roux
J
,
Lee
H
,
Miyazawa
B
,
Lee
JW
,
Gartland
B
,
Howard
AJ
,
Matthay
MA
,
Carles
M
,
Pittet
JF
:
Activation of the stress protein response inhibits the STAT1 signalling pathway and iNOS function in alveolar macrophages: Role of Hsp90 and Hsp70.
Thorax
2010
;
65
:
346
53
25.
Tadie
JM
,
Bae
HB
,
Jiang
S
,
Park
DW
,
Bell
CP
,
Yang
H
,
Pittet
JF
,
Tracey
K
,
Thannickal
VJ
,
Abraham
E
,
Zmijewski
JW
:
HMGB1 promotes neutrophil extracellular trap formation through interactions with Toll-like receptor 4.
Am J Physiol Lung Cell Mol Physiol
2013
;
304
:
L342
9
26.
Lafargue
M
,
Xu
L
,
Carlès
M
,
Serve
E
,
Anjum
N
,
Iles
KE
,
Xiong
X
,
Giffard
R
,
Pittet
JF
:
Stroke-induced activation of the α7 nicotinic receptor increases Pseudomonas aeruginosa lung injury.
FASEB J
2012
;
26
:
2919
29
27.
Bae
HB
,
Zmijewski
JW
,
Deshane
JS
,
Tadie
JM
,
Chaplin
DD
,
Takashima
S
,
Abraham
E
:
AMP-activated protein kinase enhances the phagocytic ability of macrophages and neutrophils.
FASEB J
2011
;
25
:
4358
68
28.
Pittet
JF
,
Koh
H
,
Fang
X
,
Iles
K
,
Christiaans
S
,
Anjun
N
,
Wagener
BM
,
Park
DW
,
Zmijewski
JW
,
Matthay
MA
,
Roux
J
:
HMGB1 accelerates alveolar epithelial repair via an IL-1β- and αvβ6 integrin-dependent activation of TGF-β1.
PLoS One
2013
;
8
:
e63907
29.
Gorska
M
,
Popowska
U
,
Sielicka-Dudzin
A
,
Kuban-Jankowska
A
,
Sawczuk
W
,
Knap
N
,
Cicero
G
,
Wozniak
F
:
Geldanamycin and its derivatives as Hsp90 inhibitors.
Front Biosci (Landmark Ed)
2012
;
17
:
2269
77
30.
Selva
E
,
Brest
P
,
Loubat
A
,
Lassalle
S
,
Auberger
P
,
Hofman
P
:
Inhibition of apoptosis induced by heat shock preconditioning is associated with decreased phagocytosis in human polymorphonuclear leukocytes through inhibition of Rac and Cdc42.
Immunol Cell Biol
2007
;
85
:
257
64
31.
Singh
IS
,
Gupta
A
,
Nagarsekar
A
,
Cooper
Z
,
Manka
C
,
Hester
L
,
Benjamin
IJ
,
He
JR
,
Hasday
JD
:
Heat shock co-activates interleukin-8 transcription.
Am J Respir Cell Mol Biol
2008
;
39
:
235
42
32.
Maity
TK
,
Henry
MM
,
Tulapurkar
ME
,
Shah
NG
,
Hasday
JD
,
Singh
IS
:
Distinct, gene-specific effect of heat shock on heat shock factor-1 recruitment and gene expression of CXC chemokine genes.
Cytokine
2011
;
54
:
61
7
33.
Weiss
YG
,
Bromberg
Z
,
Raj
N
,
Raphael
J
,
Goloubinoff
P
,
Ben-Neriah
Y
,
Deutschman
CS
:
Enhanced heat shock protein 70 expression alters proteasomal degradation of IkappaB kinase in experimental acute respiratory distress syndrome.
Crit Care Med
2007
;
35
:
2128
38
34.
Shi
Y
,
Tu
Z
,
Tang
D
,
Zhang
H
,
Liu
M
,
Wang
K
,
Calderwood
SK
,
Xiao
X
:
The inhibition of LPS-induced production of inflammatory cytokines by HSP70 involves inactivation of the NF-kappaB pathway but not the MAPK pathways.
Shock
2006
;
26
:
277
84
35.
Dokladny
K
,
Lobb
R
,
Wharton
W
,
Ma
TY
,
Moseley
PL
:
LPS-induced cytokine levels are repressed by elevated expression of HSP70 in rats: Possible role of NF-kappaB.
Cell Stress Chaperones
2010
;
15
:
153
63
36.
Seeley
EJ
,
Matthay
MA
,
Wolters
PJ
:
Inflection points in sepsis biology: From local defense to systemic organ injury.
Am J Physiol Lung Cell Mol Physiol
2012
;
303
:
L355
63
37.
Chatterjee
A
,
Dimitropoulou
C
,
Drakopanayiotakis
F
,
Antonova
G
,
Snead
C
,
Cannon
J
,
Venema
RC
,
Catravas
JD
:
Heat shock protein 90 inhibitors prolong survival, attenuate inflammation, and reduce lung injury in murine sepsis.
Am J Respir Crit Care Med
2007
;
176
:
667
75
38.
Schroeder
S
,
Lindemann
C
,
Hoeft
A
,
Putensen
C
,
Decker
D
,
von Ruecker
AA
,
Stüber
F
:
Impaired inducibility of heat shock protein 70 in peripheral blood lymphocytes of patients with severe sepsis.
Crit Care Med
1999
;
27
:
1080
4
39.
Kee
C
,
Cheong
KY
,
Pham
K
,
Waterer
GW
,
Temple
SE
:
Genetic variation in heat shock protein 70 is associated with septic shock: Narrowing the association to a specific haplotype.
Int J Immunogenet
2008
;
35
:
465
73
40.
Wachstein
J
,
Tischer
S
,
Figueiredo
C
,
Limbourg
A
,
Falk
C
,
Immenschuh
S
,
Blasczyk
R
,
Eiz-Vesper
B
:
HSP70 enhances immunosuppressive function of CD4(+)CD25(+)FoxP3(+) T regulatory cells and cytotoxicity in CD4(+)CD25(-) T cells.
PLoS One
2012
;
7
:
e51747
41.
Vega
VL
,
De Maio
A
:
Increase in phagocytosis after geldanamycin treatment or heat shock: Role of heat shock proteins.
J Immunol
2005
;
175
:
5280
7
42.
Wang
R
,
Town
T
,
Gokarn
V
,
Flavell
RA
,
Chandawarkar
RY
:
HSP70 enhances macrophage phagocytosis by interaction with lipid raft-associated TLR-7 and upregulating p38 MAPK and PI3K pathways.
J Surg Res
2006
;
136
:
58
69
43.
Kovalchin
JT
,
Wang
R
,
Wagh
MS
,
Azoulay
J
,
Sanders
M
,
Chandawarkar
RY
:
In vivo delivery of heat shock protein 70 accelerates wound healing by up-regulating macrophage-mediated phagocytosis.
Wound Repair Regen
2006
;
14
:
129
37
44.
Hashimoto
S
,
Pittet
JF
,
Hong
K
,
Folkesson
H
,
Bagby
G
,
Kobzik
L
,
Frevert
C
,
Watanabe
K
,
Tsurufuji
S
,
Wiener-Kronish
J
:
Depletion of alveolar macrophages decreases neutrophil chemotaxis to Pseudomonas airspace infections.
Am J Physiol
1996
;
270
(
5 Pt 1
):
L819
28
45.
Kooguchi
K
,
Hashimoto
S
,
Kobayashi
A
,
Kitamura
Y
,
Kudoh
I
,
Wiener-Kronish
J
,
Sawa
T
:
Role of alveolar macrophages in initiation and regulation of inflammation in Pseudomonas aeruginosa pneumonia.
Infect Immun
1998
;
66
:
3164
9
46.
Drusano
GL
,
Vanscoy
B
,
Liu
W
,
Fikes
S
,
Brown
D
,
Louie
A
:
Saturability of granulocyte kill of Pseudomonas aeruginosa in a murine model of pneumonia.
Antimicrob Agents Chemother
2011
;
55
:
2693
5
47.
Laichalk
LL
,
Danforth
JM
,
Standiford
TJ
:
Interleukin-10 inhibits neutrophil phagocytic and bactericidal activity.
FEMS Immunol Med Microbiol
1996
;
15
:
181
7
48.
Roilides
E
,
Katsifa
H
,
Tsaparidou
S
,
Stergiopoulou
T
,
Panteliadis
C
,
Walsh
TJ
:
Interleukin 10 suppresses phagocytic and antihyphal activities of human neutrophils.
Cytokine
2000
;
12
:
379
87
49.
McConnell
KW
,
McDunn
JE
,
Clark
AT
,
Dunne
WM
,
Dixon
DJ
,
Turnbull
IR
,
Dipasco
PJ
,
Osberghaus
WF
,
Sherman
B
,
Martin
JR
,
Walter
MJ
,
Cobb
JP
,
Buchman
TG
,
Hotchkiss
RS
,
Coopersmith
CM
:
Streptococcus pneumoniae and Pseudomonas aeruginosa pneumonia induce distinct host responses.
Crit Care Med
2010
;
38
:
223
41
50.
Pène
F
,
Zuber
B
,
Courtine
E
,
Rousseau
C
,
Ouaaz
F
,
Toubiana
J
,
Tazi
A
,
Mira
JP
,
Chiche
JD
:
Dendritic cells modulate lung response to Pseudomonas aeruginosa in a murine model of sepsis-induced immune dysfunction.
J Immunol
2008
;
181
:
8513
20
51.
Nieuwenhuis
EE
,
Matsumoto
T
,
Exley
M
,
Schleipman
RA
,
Glickman
J
,
Bailey
DT
,
Corazza
N
,
Colgan
SP
,
Onderdonk
AB
,
Blumberg
RS
:
CD1d-dependent macrophage-mediated clearance of Pseudomonas aeruginosa from lung.
Nat Med
2002
;
8
:
588
93
52.
Cyktor
JC
,
Turner
J
:
Interleukin-10 and immunity against prokaryotic and eukaryotic intracellular pathogens.
Infect Immun
2011
;
79
:
2964
73