imatinib attenuates inflammation and vascular leak in a ...€¦ · call for papers translational...

11
CALL FOR PAPERS Translational Research in Acute Lung Injury and Pulmonary Fibrosis Imatinib attenuates inflammation and vascular leak in a clinically relevant two-hit model of acute lung injury Alicia N. Rizzo, 1,2 Saad Sammani, 1 Adilene E. Esquinca, 1 Jeffrey R. Jacobson, 1 Joe G. N. Garcia, 3 Eleftheria Letsiou, 1 and Steven M. Dudek 1,2 1 University of Illinois Hospital and Health Sciences System, Division of Pulmonary, Critical Care, Sleep and Allergy, Chicago, Illinois; 2 University of Illinois at Chicago, Department of Pharmacology, Chicago, Illinois; 3 Arizona Health Sciences Center, University of Arizona, Tucson, Arizona Submitted 26 January 2015; accepted in final form 27 September 2015 Rizzo AN, Sammani S, Esquinca AE, Jacobson JR, Garcia JG, Letsiou E, Dudek SM. Imatinib attenuates inflammation and vascular leak in a clinically relevant two-hit model of acute lung injury. Am J Physiol Lung Cell Mol Physiol 309: L1294 –L1304, 2015. First published October 2, 2015 doi:10.1152/ajplung.00031.2015.—Acute lung injury/acute respiratory distress syndrome (ALI/ARDS), an ill- ness characterized by life-threatening vascular leak, is a significant cause of morbidity and mortality in critically ill patients. Recent preclinical studies and clinical observations have suggested a potential role for the chemotherapeutic agent imatinib in restoring vascular integrity. Our prior work demonstrates differential effects of imatinib in mouse models of ALI, namely attenuation of LPS-induced lung injury but exacerbation of ventilator-induced lung injury (VILI). Because of the critical role of mechanical ventilation in the care of patients with ARDS, in the present study we pursued an assessment of the effectiveness of imatinib in a “two-hit” model of ALI caused by combined LPS and VILI. Imatinib significantly decreased bronchoal- veolar lavage protein, total cells, neutrophils, and TNF- levels in mice exposed to LPS plus VILI, indicating that it attenuates ALI in this clinically relevant model. In subsequent experiments focusing on its protective role in LPS-induced lung injury, imatinib attenuated ALI when given 4 h after LPS, suggesting potential therapeutic effective- ness when given after the onset of injury. Mechanistic studies in mouse lung tissue and human lung endothelial cells revealed that imatinib inhibits LPS-induced NF-B expression and activation. Overall, these results further characterize the therapeutic potential of imatinib against inflammatory vascular leak. acute lung injury; acute respiratory distress syndrome; imatinib; endothelium; lipopolysaccharide; mechanical ventilation; NF-B ACUTE LUNG INJURY/ACUTE RESPIRATORY DISTRESS SYNDROME (ALI/ ARDS) is a devastating disease process characterized by severe pulmonary inflammation and vascular leak that causes hypox- emic respiratory failure in critically ill patients (51). Despite multiple clinical trials over the past few decades, targeted pharmacologic therapies are still lacking, and the mortality rate remains at 30 –35% (20). The most common causes of ARDS are sepsis and severe pneumonia; however, other inflammatory conditions can initiate this disease process, including trauma, pancreatitis, and aspiration of gastric contents (51). Multiple endogenous inflammatory mediators, as well as exogenous agents such as LPS, alter the pulmonary endothelial cell (EC) structure, leading to paracellular gap formation and vascular leak (19). Care for these patients is largely supportive, and low-tidal-volume mechanical ventilation (MV) is the only intervention proven to decrease mortality (1). However, the biophysical forces present during MV can contribute to both the inflammatory and the permeability aspects of ARDS, a process known as ventilator-induced lung injury (VILI) (44). Recent work by multiple groups indicates that the FDA- approved tyrosine kinase inhibitor, imatinib, attenuates vascu- lar permeability (5, 10, 27, 29, 45). Although imatinib was originally designed to inhibit the BCR-Abl fusion protein that causes chronic myelogenous leukemia, it also inhibits several other kinases including c-Abl, Abl-related gene (Arg), c-kit, and platelet-derived growth factor receptor (PDGFR), suggest- ing that it may have pleiotropic effects on vascular function (39). Accordingly, recent reports indicate that imatinib atten- uates the vascular leak induced by diverse stimuli including thrombin, histamine, VEGF, LPS, and reactive oxygen species (ROS) (5, 10, 27, 29, 45). These studies have indicated that multiple targets of imatinib, including c-Abl, Arg, and PDGFR, are involved in regulating the cytoskeletal rearrangements that mediate vascular permeability. Further support for a role of imatinib in the treatment of vascular leak is provided by a series of case reports in which imatinib therapy was associated with rapid resolution of pulmonary and systemic vascular leak (4, 8, 36). Although these observations suggest that imatinib may be a promising treatment for inflammatory vascular leak, the mechanisms underlying these effects remain incompletely understood. Additionally, contrary to its barrier-protective ef- fects, imatinib has well-established side effects of periorbital and subcutaneous edema, as well as pleural and pericardial effusions in patients, suggesting that it can worsen vascular leak in certain clinical contexts (17, 23, 41). Consistent with this potential for imatinib to exert both barrier-protective and edemagenic effects, we recently reported that imatinib attenuates LPS-induced lung injury but exacer- bates VILI both in vitro and in vivo (29). Because of the necessity of MV in the treatment of patients with ARDS, these observations are clinically relevant and suggest that imatinib has the potential to worsen ARDS in some situations. To Address for reprint requests and other correspondence: S. Dudek, Div. of Pulmonary, Critical Care, Sleep, and Allergy, Univ. of Illinois Hospital & Health Sciences System, CSB 915, 840 S. Wood St., Chicago, IL 60612 (e-mail: [email protected]). Am J Physiol Lung Cell Mol Physiol 309: L1294–L1304, 2015. First published October 2, 2015; doi:10.1152/ajplung.00031.2015. 1040-0605/15 Copyright © 2015 the American Physiological Society http://www.ajplung.org L1294 by 10.220.33.1 on April 28, 2017 http://ajplung.physiology.org/ Downloaded from

Upload: others

Post on 08-May-2020

6 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Imatinib attenuates inflammation and vascular leak in a ...€¦ · CALL FOR PAPERS Translational Research in Acute Lung Injury and Pulmonary Fibrosis Imatinib attenuates inflammation

CALL FOR PAPERS Translational Research in Acute Lung Injury and Pulmonary

Fibrosis

Imatinib attenuates inflammation and vascular leak in a clinically relevanttwo-hit model of acute lung injury

Alicia N. Rizzo,1,2 Saad Sammani,1 Adilene E. Esquinca,1 Jeffrey R. Jacobson,1 Joe G. N. Garcia,3

Eleftheria Letsiou,1 and Steven M. Dudek1,2

1University of Illinois Hospital and Health Sciences System, Division of Pulmonary, Critical Care, Sleep and Allergy,Chicago, Illinois; 2University of Illinois at Chicago, Department of Pharmacology, Chicago, Illinois; 3Arizona HealthSciences Center, University of Arizona, Tucson, Arizona

Submitted 26 January 2015; accepted in final form 27 September 2015

Rizzo AN, Sammani S, Esquinca AE, Jacobson JR, Garcia JG,Letsiou E, Dudek SM. Imatinib attenuates inflammation and vascularleak in a clinically relevant two-hit model of acute lung injury. Am JPhysiol Lung Cell Mol Physiol 309: L1294–L1304, 2015. Firstpublished October 2, 2015 doi:10.1152/ajplung.00031.2015.—Acutelung injury/acute respiratory distress syndrome (ALI/ARDS), an ill-ness characterized by life-threatening vascular leak, is a significantcause of morbidity and mortality in critically ill patients. Recentpreclinical studies and clinical observations have suggested a potentialrole for the chemotherapeutic agent imatinib in restoring vascularintegrity. Our prior work demonstrates differential effects of imatinibin mouse models of ALI, namely attenuation of LPS-induced lunginjury but exacerbation of ventilator-induced lung injury (VILI).Because of the critical role of mechanical ventilation in the care ofpatients with ARDS, in the present study we pursued an assessment ofthe effectiveness of imatinib in a “two-hit” model of ALI caused bycombined LPS and VILI. Imatinib significantly decreased bronchoal-veolar lavage protein, total cells, neutrophils, and TNF-� levels inmice exposed to LPS plus VILI, indicating that it attenuates ALI inthis clinically relevant model. In subsequent experiments focusing onits protective role in LPS-induced lung injury, imatinib attenuated ALIwhen given 4 h after LPS, suggesting potential therapeutic effective-ness when given after the onset of injury. Mechanistic studies inmouse lung tissue and human lung endothelial cells revealed thatimatinib inhibits LPS-induced NF-�B expression and activation.Overall, these results further characterize the therapeutic potential ofimatinib against inflammatory vascular leak.

acute lung injury; acute respiratory distress syndrome; imatinib;endothelium; lipopolysaccharide; mechanical ventilation; NF-�B

ACUTE LUNG INJURY/ACUTE RESPIRATORY DISTRESS SYNDROME (ALI/ARDS) is a devastating disease process characterized by severepulmonary inflammation and vascular leak that causes hypox-emic respiratory failure in critically ill patients (51). Despitemultiple clinical trials over the past few decades, targetedpharmacologic therapies are still lacking, and the mortality rateremains at 30–35% (20). The most common causes of ARDSare sepsis and severe pneumonia; however, other inflammatoryconditions can initiate this disease process, including trauma,

pancreatitis, and aspiration of gastric contents (51). Multipleendogenous inflammatory mediators, as well as exogenousagents such as LPS, alter the pulmonary endothelial cell (EC)structure, leading to paracellular gap formation and vascularleak (19). Care for these patients is largely supportive, andlow-tidal-volume mechanical ventilation (MV) is the onlyintervention proven to decrease mortality (1). However, thebiophysical forces present during MV can contribute to boththe inflammatory and the permeability aspects of ARDS, aprocess known as ventilator-induced lung injury (VILI) (44).

Recent work by multiple groups indicates that the FDA-approved tyrosine kinase inhibitor, imatinib, attenuates vascu-lar permeability (5, 10, 27, 29, 45). Although imatinib wasoriginally designed to inhibit the BCR-Abl fusion protein thatcauses chronic myelogenous leukemia, it also inhibits severalother kinases including c-Abl, Abl-related gene (Arg), c-kit,and platelet-derived growth factor receptor (PDGFR), suggest-ing that it may have pleiotropic effects on vascular function(39). Accordingly, recent reports indicate that imatinib atten-uates the vascular leak induced by diverse stimuli includingthrombin, histamine, VEGF, LPS, and reactive oxygen species(ROS) (5, 10, 27, 29, 45). These studies have indicated thatmultiple targets of imatinib, including c-Abl, Arg, and PDGFR,are involved in regulating the cytoskeletal rearrangements thatmediate vascular permeability. Further support for a role ofimatinib in the treatment of vascular leak is provided by aseries of case reports in which imatinib therapy was associatedwith rapid resolution of pulmonary and systemic vascular leak(4, 8, 36). Although these observations suggest that imatinibmay be a promising treatment for inflammatory vascular leak,the mechanisms underlying these effects remain incompletelyunderstood. Additionally, contrary to its barrier-protective ef-fects, imatinib has well-established side effects of periorbitaland subcutaneous edema, as well as pleural and pericardialeffusions in patients, suggesting that it can worsen vascularleak in certain clinical contexts (17, 23, 41).

Consistent with this potential for imatinib to exert bothbarrier-protective and edemagenic effects, we recently reportedthat imatinib attenuates LPS-induced lung injury but exacer-bates VILI both in vitro and in vivo (29). Because of thenecessity of MV in the treatment of patients with ARDS, theseobservations are clinically relevant and suggest that imatinibhas the potential to worsen ARDS in some situations. To

Address for reprint requests and other correspondence: S. Dudek, Div. ofPulmonary, Critical Care, Sleep, and Allergy, Univ. of Illinois Hospital &Health Sciences System, CSB 915, 840 S. Wood St., Chicago, IL 60612(e-mail: [email protected]).

Am J Physiol Lung Cell Mol Physiol 309: L1294–L1304, 2015.First published October 2, 2015; doi:10.1152/ajplung.00031.2015.

1040-0605/15 Copyright © 2015 the American Physiological Society http://www.ajplung.orgL1294

by 10.220.33.1 on April 28, 2017

http://ajplung.physiology.org/D

ownloaded from

Page 2: Imatinib attenuates inflammation and vascular leak in a ...€¦ · CALL FOR PAPERS Translational Research in Acute Lung Injury and Pulmonary Fibrosis Imatinib attenuates inflammation

address this concern, in the present study, we investigated theefficacy of imatinib using a “two-hit” preclinical murine modelof ALI that combines intratracheal LPS administration fol-lowed by high-tidal-volume MV (HTVMV). Additionally, weassessed the therapeutic potential of imatinib when given afterthe onset of inflammatory lung injury, a critical step in thepreclinical testing of interventions aimed at treating existinginjury. We also characterized the effects of imatinib on LPS-induced NF-�B signaling in vitro and in vivo. Our data indicatethat imatinib has potent anti-inflammatory effects, in additionto its previously described effects on EC permeability, andfurther support its potential use as a therapeutic option forinflammatory vascular leak syndromes.

MATERIALS AND METHODS

Animal care and preclinical models of ALI. Male C57BL/6J miceaged 8–12 wk (Jackson Laboratories, Bar Harbor, ME) were used forall experiments. Mice were anesthetized with intraperitoneal ketamine(100 mg/kg) and xylazine (5 mg/kg) before LPS and VILI protocols,with additional doses given as needed during the VILI protocol toensure adequate anesthetic depth. For experiments utilizing the two-hit lung ALI model, animals were given intratracheal (it) LPS (0.5mg/kg), allowed to recover for 20 h, and were then reintubated andplaced on MV (Harvard Apparatus, Boston, MA) with room air for 4h using the following parameters: tidal volume (VT) 30 ml/kg,respiratory rate 75 breaths/min, and positive-end expiratory pressure 0cm H2O. Spontaneously breathing (SB) control animals were givenintratracheally PBS instead of LPS and were intubated but allowed tobreathe spontaneously for the 4 h. Imatinib (75 mg/kg, ip) (LCLaboratories, Woburn, MA) or vehicle (water) was given 30 minbefore LPS administration and again 30 min before the initiation ofMV. For experiments in which imatinib was administered post injury,imatinib (75 mg/kg, ip) or vehicle was given 4 h after LPS (1 mg/kg,it) or PBS. The dosage of imatinib used was chosen to be 75 mg/kgto be consistent with our previous work and within the range of whatother groups have used in mice (5, 27, 29). The dosage of imatinibused in cancer therapy ranges from 11–15 mg/kg depending on theparticular malignancy (16, 37). Although this dose is lower than whatwe used in the mouse models in this study (75 mg/kg), there aresubstantial differences in the metabolism of imatinib between miceand humans that strongly suggest that the dosage used in this study isreasonable and clinically relevant. The most striking difference be-tween the pharmacokinetics of imatinib in humans and mice is that thehalf-life of imatinib in mice is 2.4 h, compared with 18 h in humans(37, 39, 47). Thus, to attain an effective plasma concentration ofimatinib in the time frame necessary for our studies, a higher dose ofimatinib was necessary than that used clinically in humans.

At the termination of each of the animal experiments, bronchoal-veolar lavage (BAL) fluid was collected by instilling 1 ml of HBSS(Invitrogen, Grand Island, NY) through the tracheal cannula into thelungs, followed by slow recovery of the fluid. Cells were recoveredfrom the resulting BAL fluid by centrifugation (500 g, 20 min, 4°C)and counted using an automated cell counter (TC20; Bio-Rad, Her-cules, CA). Manual differential cell counts (500 cells/sample) wereperformed by a blinded investigator on cytospin samples (Shandon,Cytospin 4; Thermo Fisher Scientific, Rockford, IL) after stainingwith Kwik Diff Stain (Thermo Scientific). The BAL fluid supernatantwas centrifuged again (17,000 g, 10 min, 4°C) and stored at �80°Cfor further analysis. Lung tissues were snap-frozen in liquid nitrogenor embedded in paraformaldehyde (PFA) for histology. Select animalsin both studies were given Evans blue dye (EBD) (30 mg/kg) 1 hbefore harvest via an intrajugular injection. BAL was not performedon animals that were given EBD before harvest. All animal careprocedures and experiments were approved by the University ofIllinois at Chicago Animal Care and Use Committee.

Analysis of mouse samples. BAL fluid protein was measured usinga bicinchoninic acid assay kit (Thermo Scientific), and BAL TNF-�,IL-6, and IL-1� levels were measured using ELISA kits obtained fromeBiosciences (San Diego, CA) according to the manufacturer’s pro-tocols. Lung tissues were homogenized in radioimmunoprecipitationassay (RIPA) buffer supplemented with protease and phosphataseinhibitors using a hand-held homogenizer (Qiagen, Venlo, The Neth-erlands), subjected to three freeze-thaw cycles, and sonicated beforeprotein quantification and Western blotting. Lung tissue and BALalbumin concentrations were measured using an ELISA kit (BethylLaboratories, Montgomery, TX).

Wet:dry lung weight measurements and EBD measurements wereconducted on mice that did not undergo BAL. In these animals,mice were given an intrajugular injection of EBD (30 mg/kg) 1 hbefore harvest. Following perfusion of the vasculature, the leftlungs were immediately excised and weighed for determination ofwet lung weight. The lungs were then placed in an oven at 21°C for48 h and then weighed again for determination of dry lung weight.The right lungs of these animals were frozen in liquid nitrogen andthen homogenized in 1 ml of PBS using a hand-held homogenizer(Qiagen). The samples were then incubated in a 60°C water bathovernight, after the addition of 2 ml of formamide to each sample.The following morning, the samples were centrifuged (12,000 g,20 min, room temperature). The EBD concentration in the resultingsupernatant was determined by spectrophotometry as previouslydescribed (33).

Lung histopathology and immunohistochemistry. To assess altera-tions in the lung tissue morphology, lungs were fixed in formalin,embedded in paraffin, sectioned, mounted onto slides, and stainedwith hematoxylin-eosin (H and E). Separate slides for immunohisto-chemistry (IHC) were hydrated through a xylene and alcohol gradientbefore antigen unmasking with an EDTA-based retrieval solution.Slides were then blocked with hydrogen peroxide blocking reagent(20 min, room temperature), probed with NF-�B antibody (Abcam,Cambridge, MA) (1:100, 60 min, room temperature), and then incu-bated in anti-rabbit horseradish peroxidase (HRP) secondary antibody(Biocare, Concord, CA) (20 min, room temperature). NF-�B stainingwas detected by 3,3=-diaminobenzidine (Cell Marque, Rockland, CA)for 10 min. All slides were scanned using an Aperio ScanScope(Leica, Buffalo Grove, IL), and representative images were taken at�100 by an individual blinded to experimental condition.

Endothelial cell culture and Western blotting. Human pulmonaryartery endothelial cells (HPAEC) were purchased from Lonza (Walk-ersville, MD) and cultured in endothelial growth medium-2 supple-mented with 10% FBS (Sigma, St. Louis, MO). Cells were maintainedat 37°C in a 5% CO2 incubator and used at passages 6–8. For allexperiments, the media containing 10% FBS was replaced with mediacontaining 2% FBS 3 h before the indicated treatment. HPAEC weretreated with imatinib (40 �M) or vehicle (water) for 1 h beforechallenge with LPS (E0127:B8, Sigma no. L3880) (1 �g/ml, 0–60min). The in vitro dosage of imatinib was chosen to be consistent withthe dosage used in our previous publication (29). Although this doseis slightly higher than that used in other in vitro studies on the effectsof imatinib on vascular integrity (10 �M) (5, 10), these studiessolubilized the drug in DMSO, which increases permeability of theplasma membrane and thus increases the effective concentration ofimatinib in the cells (15). Following the indicated treatments, EC werewashed in ice-cold PBS and then harvested in RIPA buffer supple-mented with protease and phosphatase inhibitors. Protein sampleswere then prepared in Laemmli SDS sample buffer (Boston Bioprod-ucts, Ashland, MA), boiled, subjected to SDS-PAGE, and transferredto polyvinylidene difluoride membrane. The membranes were blockedin 5% BSA and incubated in the indicated antibodies (phospho-NF-�B, total NF-�B, and I�B were purchased from Cell Signaling,Danvers, MA) (overnight, 4°C). Next, HRP-conjugated secondaryantibodies (Cell signaling) were added to the membranes (60 min,room temperature), and the Pierce enhanced chemiluminescence de-

L1295IMATINIB IS PROTECTIVE IN A MURINE MODEL OF LPS AND VILI

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00031.2015 • www.ajplung.org

by 10.220.33.1 on April 28, 2017

http://ajplung.physiology.org/D

ownloaded from

Page 3: Imatinib attenuates inflammation and vascular leak in a ...€¦ · CALL FOR PAPERS Translational Research in Acute Lung Injury and Pulmonary Fibrosis Imatinib attenuates inflammation

tection system (Thermo Scientific) was used to visualize the bands.Western blots on homogenized mouse lung samples were conductedidentically. Band densities were determined using the Image J soft-ware (http://imagej.nih.gov/ij/) (National Institutes of Health).

Immunofluorescence microscopy. HPAEC grown on gelatin-coatedcoverslips were treated with imatinib and LPS as described above,fixed in 4% PFA (20 min, room temperature), permeabilized with0.1% Triton-X in PBS, and incubated with NF-�B p65 antibody(overnight, 4°C). Coverslips were then washed, incubated with fluo-rescently labeled secondary antibody, and mounted on glass slidesusing 4=,6-diamidino-2-phenylindole Prolong Gold anti-fade reagent(Invitrogen). Images were acquired by a blinded investigator usingNikon Eclipse TE2000 microscope at �63 with an oil emersion lens.

Statistical analysis. Results were expressed as mean � SE withthree to seven animals per group in each experiment. The data forBAL albumin, lung tissue albumin, EBD extravasation, and lungtissue NF-�B expression are presented as values normalized to eitherthe LPS VILI Veh (2-hit model), or to LPS Veh (postinjurystudies). Only three animals per group were used in the SB groups(2-hit model studies) and the PBS injection groups (postinjury studies)because of the very small variation between samples. All in vitro

experiments were repeated at least three independent times. Student’st-test was used to compare between two groups, and one-wayANOVA with Tukey’s post hoc test was used to compare betweenmultiple groups. All statistical analyses were performed using Graph-Pad Prism 6 software with the significance level set to P 0.05 forall experiments.

RESULTS

Imatinib attenuates vascular leak and inflammation in aclinically relevant two-hit model of ALI. We recently reportedthat imatinib attenuates LPS-induced ALI but exacerbates VILIin both cell culture and murine models (29). However, becauseof the necessity of MV in the treatment of ARDS, inflamma-tion- and mechanical force-induced cellular injuries commonlycoexist in patients. In this study, we utilized a clinicallyrelevant two-hit murine model of LPS combined with VILI(22, 31) to further assess the efficacy of imatinib in ALI. Inthese studies, mice were challenged with LPS (0.5 mg/kg, it)(t � 0 h), followed by HTVMV (VT � 30 ml/kg) (t � 20–24

A

B C

D E

F G

0

200

400

600

800

1000

BA

L Pr

otei

n (μ

g/m

L)

Veh IM Veh IM

BAL Protein

0.0

0.5

1.0

1.5

Tiss

ue A

lbum

in(n

orm

aliz

ed)

* * #

Lung Tissue Albumin

4

5

6

7

Wet

:Dry

Wet:Dry Lung Weight

0.0

0.5

1.0

1.5

EBD

(nor

mal

ized

)

EBD Extravasation

0.0

0.5

1.0

1.5

2.0

BA

L A

lbum

in(n

orm

aliz

ed)

**

BAL Albumin

Veh Veh IM

LPS + VILISB

LPS + VILISBVeh IM Veh IM

LPS + VILISB

Veh IM Veh IMLPS + VILISB

Veh IM Veh IMLPS + VILISB

Veh IM Veh IMLPS + VILISB

** #

** # *

* #

Time (hrs)

InterventionImatinib IP75 mg/kg

Imatinib IP75 mg/kg

LPS IT0.5 mg/kg

MV 75 breaths/min30 ml/kg, 4 hrs

-0.5 0 19.5 20 24

Fig. 1. Imatinib attenuates vascular leak inmice challenged with a 2-hit model of LPSand mechanical ventilation (MV). Mice werechallenged with LPS [0.5 mg/kg, intratrache-ally (it)] (t � 0 h) and MV (respiratory rate75, tidal volume 30 ml/kg, positive end-expi-ratory pressure 0 cm H2O) (t � 20–24 h) asoutlined in A. Spontaneously breathing (SB)control mice received PBS (vs. LPS) andwere intubated for 4 h without MV. Bron-choalveolar lavage (BAL) fluid, plasma, andlungs were harvested from the animals imme-diately after MV. Imatinib (IM) (75 mg/kg,ip) or vehicle was administered 0.5 h beforeLPS administration and before the initiationof MV. Lung permeability was assessed byBAL protein content (B), BAL fluid albumin(C), and lung tissue albumin (D). Addition-ally, in separate animals, Evans Blue dye(EBD) was injected (30 mg/kg, iv) 1 h beforeharvest, and representative extravasation intoharvested lung tissue is shown (E) and quan-tified in multiple samples (F). The left lung ofeach of these animals was used for calculationof lung wet:dry ratio (G). SB (n � 3), SB imatinib (n � 3), LPS ventilator-inducedlung injury (VILI) (n � 3–11) and LPS VILI imatinib (n � 3–6). *P 0.05 compared withSB controls and #P 0.05 compared withuntreated animals.

L1296 IMATINIB IS PROTECTIVE IN A MURINE MODEL OF LPS AND VILI

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00031.2015 • www.ajplung.org

by 10.220.33.1 on April 28, 2017

http://ajplung.physiology.org/D

ownloaded from

Page 4: Imatinib attenuates inflammation and vascular leak in a ...€¦ · CALL FOR PAPERS Translational Research in Acute Lung Injury and Pulmonary Fibrosis Imatinib attenuates inflammation

h). Imatinib (or vehicle) was given 30 min before LPS injection(t � �0.5 h) and again 30 min before MV (t � 19.5 h) (Fig.1A). In this study, we utilized identical imatinib treatments andMV parameters as in our previous work (29) and thereforehave not included redundant data here on the effects of pre-treatment with imatinib in LPS-induced lung injury or VILIalone. As expected, mice exposed to the two-hit model dis-played increased BAL protein (�4-fold), indicative of in-creased vascular leak. Imatinib was protective in this modeland reduced BAL protein levels by 33% (P 0.01) (Fig. 1B).To further quantify the vascular leak, albumin levels in BALfluid and lung tissue were quantified. Imatinib attenuated theincrease in lung tissue albumin induced by LPS plus VILI by22% (P 0.05) (Fig. 1C). We also observed a trend towarddecreased BAL albumin in imatinib-treated animals (22%decline, P � 0.2) (Fig. 1D). Additionally, EBD extravasationinto the lungs, another well-characterized measurement ofvascular leak (21), was decreased by 33% (P 0.05) inanimals receiving imatinib (Fig. 1, E and F). Furthermore, LPSplus VILI significantly increased lung wet:dry weight ratio, ameasure of pulmonary edema, and imatinib attenuated thisincrease by 62% (P 0.05) (Fig. 1G). Together, these dataindicate that imatinib attenuates vascular leak in this two-hitmodel of ALI.

We next assessed pulmonary inflammation in these animalsby measuring BAL cell counts and lung histology. Comparedwith control mice, the two-hit injury model caused a dramaticincrease in total BAL cells and BAL neutrophils, which weredecreased by 56 and 52%, respectively, in imatinib-treated

animals (P 0.001 for both) (Fig. 2, A and B). Histologicanalyses of H and E-stained lung tissue revealed that micechallenged with LPS plus MV displayed architectural distor-tion and neutrophil invasion, which were substantially de-creased in the imatinib-treated animals as evidenced by repre-sentative images from three independent mice treated withimatinib, along with LPS plus VILI, and three vehicle-treatedcontrol animals that were also subjected to LPS plus VILI (Fig.2C). Together these data demonstrate that imatinib attenuatesinflammation in this clinically relevant two-hit model of ALI.

Imatinib is protective after the onset of inflammation in LPSinduced-lung injury. Given that the efficacy of imatinib in thetwo-hit model is similar to that observed in LPS-induced ALIalone, we next employed this more easily interpreted LPS-onlymodel to evaluate the potential for imatinib to attenuate ALIafter the onset of injury. For these studies, mice were chal-lenged with LPS (1.0 mg/kg, it) or PBS and then receivedimatinib (75 mg/kg, ip) or vehicle 4 h later. The dosage of LPSwas increased in these experiments compared with the two-hitinjury model to cause a more robust injury in the setting of asingle-hit model. Animals were harvested 18 h after LPSadministration, and BAL and lung tissue were collected andanalyzed identically to the two-hit model studies. As expected,BAL protein, BAL albumin, and lung tissue albumin levelswere all increased in the LPS-challenged mice compared withPBS-treated control animals. Additionally, despite being given4 h after the onset of injury, imatinib retained its protectiveeffects and attenuated vascular leak as measured by BAL totalprotein content (40% decline, P 0.05) and BAL albumin

Veh IM Veh IM0

1

2

3

4

5

BA

L C

ell C

ount

(x10

6 )

SB LPS + VILI

*

* #

BAL Cell Count

Veh IM Veh IM0

1

2

3

4

BA

L N

eutr

ophi

l Cou

nt (x

106 )

SB LPS + VILI

*BAL Neutrophil CountA B

C LPS + MVSB

Veh

IM

* #

Fig. 2. Imatinib decreases lung inflammationin mice challenged with LPS and MV. Micewere subjected to the 2-hit lung injury model(LPS VILI), and lung inflammation wasquantified by BAL total cell counts (A) andBAL neutrophil counts (B). Representativehematoxylin and eosin (H and E)-stained lungsections are shown (C). Each H and E imagewas obtained from a different animal. SB (n �3), SB imatinib (n � 3), LPS VILI (n �3–11), and LPS VILI imatinib (n � 3–6).*P 0.05 compared with SB controls and #P 0.05 compared with untreated animals.

L1297IMATINIB IS PROTECTIVE IN A MURINE MODEL OF LPS AND VILI

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00031.2015 • www.ajplung.org

by 10.220.33.1 on April 28, 2017

http://ajplung.physiology.org/D

ownloaded from

Page 5: Imatinib attenuates inflammation and vascular leak in a ...€¦ · CALL FOR PAPERS Translational Research in Acute Lung Injury and Pulmonary Fibrosis Imatinib attenuates inflammation

(51% decline, P 0.05) (Fig. 3, A and B). We also observeda trend toward decreased lung tissue albumin (24% decline,P � 0.15) compared with LPS alone (Fig. 3C). These datawere supported by measurements of EBD extravasation andwet:dry lung weight. The leakage of EBD into the lung paren-chyma was decreased by 25% (P 0.05) in the animalsreceiving imatinib after LPS (Fig. 3, D and E), and the wet:drylung weight ratio was decreased by 50% (P 0.05) in theimatinib-treated animals (Fig. 3F).

We then measured the BAL cell counts and carried outhistological analyses of the lung tissue to measure the inflam-matory damage present in these animals. As expected, theLPS-challenged mice displayed drastically higher BAL cellcounts (Fig. 4A) and neutrophil numbers (Fig. 4B) than thePBS-treated control animals. Similar to previous experimentsin which the imatinib was administered 30 min before the timeof injury, imatinib given 4 h after LPS still attenuated LPS-induced BAL cell counts by 41% (P 0.05) and neutrophilcounts by 43% (P 0.05). In agreement with these data,histological assessment of H and E-stained lung sections frommultiple independent animals demonstrated decreased edemaformation, neutrophil invasion, and disruption of lung tissue

morphology in the mice receiving imatinib (Fig. 4C). Together,these studies suggest that imatinib retains barrier-protectiveand anti-inflammatory effects in LPS-induced ALI when givenafter the onset of injury.

Imatinib attenuates LPS-induced TNF-� production in vivo.To further characterize the anti-inflammatory effects of ima-tinib, we next utilized ELISAs to measure its effects onproduction of the inflammatory cytokines TNF-�, IL-6, andIL-1�. Imatinib reduced BAL levels of TNF-� in the two-hitinjury model (88%, P � 0.001) (Fig. 5A). We also observed atrend toward decreased TNF-� in the LPS-only postinjurymodel (21%, P � 0.33) (Fig. 5C). In both injury models therealso was a trend that did not reach statistical significancetoward decreased BAL IL-6 in imatinib-treated mice (Fig. 5, Band D), consistent with our previous observations (29). NeitherIL-6 nor TNF-� was detected in the BAL of the controlanimals in either model (data not shown). IL-1� was increasedin both ALI models, but imatinib treatment did not alter itslevels (data not shown).

Imatinib decreases LPS-induced NF-�B activation inHPAEC and expression in mouse lung tissue. Imatinib inhibitsLPS-induced VCAM-1 expression and cytokine production in

* #

A B

C D

E F

0

500

1000

1500

BA

L Pr

otei

n (μ

g/m

L)

Veh IM Veh IM

PBS LPS

*

BAL Protein

0.0

0.5

1.0

1.5

Tiss

ue A

lbum

in(n

orm

aliz

ed)

Veh IM Veh IM

PBS LPS

**

Lung Tissue Albumin

4

5

6

7

Wet

:Dry

Veh IM Veh IMPBS LPS

Wet:Dry Lung Weight

*

0.0

0.5

1.0

1.5

EBD

(nor

mal

ized

)

Veh IM Veh IM

EBD Extravasation

*

0.0

0.5

1.0

1.5

BA

L A

lbum

in(n

orm

aliz

ed)

Veh IM Veh IM

PBS LPS

BAL Albumin

*

Veh Veh IM

PBS LPS

* #

* #* #

Fig. 3. Imatinib decreases LPS-induced vascu-lar leak when given after injury onset. Micewere challenged with LPS (1.0 mg/kg, it) (vs.PBS) and then received imatinib (75 mg/kg, ip)(vs. vehicle) 4 h later. Samples were harvested18 h after LPS administration. Pulmonary vas-cular permeability was quantified in these ani-mals by measuring BAL protein (A), BAL al-bumin (B), and lung tissue albumin (C). Inseparate animals, EBD was injected (30 mg/kg,iv) 1 h before harvest, and representative ex-travasation into harvested lung tissue is shown(D) and quantified in multiple samples (E). Theleft lung of each of these animals was used forcalculation of lung wet:dry ratio (F). SB (n � 3),SB imatinib (n � 3), LPS (n � 3–6), andLPS imatinib (n � 3–6). *P 0.05 comparedwith SB controls and #P 0.05 compared withuntreated animals.

L1298 IMATINIB IS PROTECTIVE IN A MURINE MODEL OF LPS AND VILI

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00031.2015 • www.ajplung.org

by 10.220.33.1 on April 28, 2017

http://ajplung.physiology.org/D

ownloaded from

Page 6: Imatinib attenuates inflammation and vascular leak in a ...€¦ · CALL FOR PAPERS Translational Research in Acute Lung Injury and Pulmonary Fibrosis Imatinib attenuates inflammation

cultured human lung EC (29). To determine whether theseeffects are mediated by decreased NF-�B activation, HPAECwere pretreated with imatinib (40 �M, 60 min) and thenchallenged with LPS (1 �g/ml, 0–60 min). Cell lysates werethen assessed by Western blotting for NF-�B phosphorylated atSer536, indicative of activated NF-�B (40). LPS increasedNF-�B phosphorylation threefold at 30 min and fourfold at 60min, whereas imatinib attenuated this LPS-induced NF-�Bactivation at both time points (P 0.05) (Figs. 6, A and B).These changes were found to be independent of levels of I�B(Fig. 6A). In confirmatory immunofluorescence experiments,imatinib inhibited LPS-induced translocation of NF-�B p65 tothe nuclei in HPAEC (Fig. 6C), which is indicative of de-creased transcriptional activation (49).

To determine the effects of imatinib on LPS-induced NF-�Bexpression in vivo, we performed Western blot analysis ofwhole lung homogenates from LPS-challenged mice that re-ceived imatinib (or vehicle control) 4 h after the onset ofinjury. NF-�B expression that was 39% lower was observed inthe whole lung homogenates of mice receiving imatinib com-pared with the vehicle-treated controls (P 0.05) (Fig. 7, Aand B). In confirmatory experiments, we further evaluatedNF-�B expression in vivo using IHC of formalin-fixed, paraf-fin-embedded sections from LPS-challenged mice receivingimatinib or vehicle. LPS substantially increased NF-�B expres-sion in lung tissue, whereas imatinib attenuated this increase(Fig. 7C). Together, these studies demonstrate that imatinibinhibits LPS-induced NF-�B activation in human pulmonaryEC as well as total NF-�B expression in mouse lungs.

DISCUSSION

Inflammatory vascular leak underlies the pathophysiology ofseveral disorders that afflict critically ill patients, includingsepsis and ARDS (25, 51). In these disease processes, multipleinflammatory stimuli combine to alter the EC cytoskeleton,which results in intercellular gap formation, increased perme-ability, and ultimately edema formation (19). Although thereare no effective therapies to protect EC barrier function, recentwork from several groups has demonstrated that imatinib, anFDA-approved tyrosine kinase inhibitor, attenuates vascularleak induced by multiple inflammatory stimuli (5, 10, 27, 45).Imatinib-mediated barrier protection has also been reported invivo using the murine cecal ligation and puncture sepsis modeland LPS-induced ALI in both immunocompetent and immu-nocompromised mice (5, 27, 29). Additionally, a recent seriesof case reports suggests that imatinib may reduce vascularpermeability in certain conditions in humans (4, 8, 36), sug-gesting that imatinib may have therapeutic potential in patientswith inflammatory vascular leak syndromes.

However, contrary to these barrier-protective effects, ima-tinib commonly causes side effects indicative of increasedvascular leak, including periorbital and subcutaneous edema,as well as pleural and pericardial effusions (17, 23, 41).Additionally, we recently reported that imatinib exacerbatesVILI in both animal and cell culture models of this condition(cyclic stretch and HTVMV, respectively) (29). Because of thecrucial role of MV in ARDS treatment, this observation raisessignificant concerns regarding the potential efficacy of imatinib

Veh IM Veh IM0

1

2

3

4

5

BA

L C

ell C

ount

(x10

6 )

*

* #

BAL Cell Count

Veh IM Veh IM0

1

2

3

4

BA

L N

eutr

ophi

l Cou

nt (x

106 )

*BAL Neutrophil CountA B

C LPSPBS

Veh

IM

* #

LPSPBS LPSPBS

Fig. 4. Imatinib decreases LPS-induced inflam-mation when given after injury onset. Micewere challenged with LPS (1.0 mg/kg, it) (vs.PBS) and then received imatinib (75 mg/kg, ip)(vs. vehicle) 4 h later. Lung inflammation wasthen quantified by BAL total cell counts (A)and BAL neutrophil counts (B). RepresentativeH and E-stained lung sections are shown (C).Each H and E image was obtained from a dif-ferent animal. SB (n � 3), SB imatinib (n � 3),LPS (n � 3–6), and LPS imatinib (n � 3–6).*P 0.05 compared with SB controls and #P 0.05 compared with untreated animals.

L1299IMATINIB IS PROTECTIVE IN A MURINE MODEL OF LPS AND VILI

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00031.2015 • www.ajplung.org

by 10.220.33.1 on April 28, 2017

http://ajplung.physiology.org/D

ownloaded from

Page 7: Imatinib attenuates inflammation and vascular leak in a ...€¦ · CALL FOR PAPERS Translational Research in Acute Lung Injury and Pulmonary Fibrosis Imatinib attenuates inflammation

in patients with ARDS. The present study advances our knowl-edge concerning the therapeutic potential of imatinib in ALI/ARDS by demonstrating its efficacy in a clinically relevant,two-hit mouse model of ALI induced by a combination of LPSand VILI (Figs. 1 and 2). In these experiments, the ventilatorparameters and imatinib treatments were identical to those inour previous work (29), and for this reason the data from VILIalone are not repeated in this study. Although the currentARDS Network lung-protective strategy recommends startingwith 6 ml/kg VT (1), we elected to utilize a relatively high VT

(30 ml/kg) because of the differences in human vs. mouse lungarchitecture as well as to maximize the deleterious effects ofVILI in this model, thus biasing our study toward accepting thenull hypothesis that imatinib is not efficacious in murine ALI.Additionally, the dosage of LPS was lowered from 1.0 mg/kgto 0.5 mg/kg to better titrate the level of lung injury to test thisnull hypothesis. Given that our study was biased towardidentifying the potential deleterious effects of imatinib, itsprotective effects in this two-hit model are particularly encour-aging. These data strongly suggest that the protective effects ofimatinib on LPS-induced ALI outweigh its deleterious effectsin VILI. Given that our data demonstrate a protective effect ofimatinib in the two-hit model despite maximizing its potentialdeleterious effects (by using a VT that is substantially higherthan the ARDS Network recommendation), we believe thatfurther preclinical testing of this therapeutic agent for ARDS iswarranted.

Additional experiments demonstrate the potential of ima-tinib to attenuate ALI when given after the onset of injury(Figs. 3 and 4), an essential characteristic for any interven-tion being considered as therapy for established disease.Interestingly, the BAL TNF-� and IL-6 levels were muchhigher in the LPS-only model compared with the two-hitmodel, which we hypothesize is due to the higher dose ofLPS used in the LPS-only experiments. Additionally, weobserved that the effect of imatinib on the levels of thesecytokines was diminished in the LPS-only model, whichsuggests that the upregulation of these inflammatory cyto-kines occurs early in LPS-induced ALI. Although this studyfocused on the effects of imatinib on LPS-induced NF-�Bsignaling, it is important to note that VILI also inducesNF-�B signaling (28, 32). Thus additional research is nec-essary to determine the effects of imatinib on NF-�B sig-naling in the context of MV alone.

The effects of imatinib in several different vascular leakmodels suggest that multiple imatinib-sensitive kinases maycontribute to these effects. Notably, the nonreceptor tyrosinekinase Arg is activated downstream of thrombin, histamine,and VEGF and contributes to vascular leak by inhibitingRac activation and impairing focal adhesion remodeling (5).Subsequent work demonstrated that the closely related ki-nase c-Abl also contributes to the cytoskeletal rearrange-ments that occur downstream of VEGF, thrombin, andhistamine via effects on the small GTPases Rac1 and Rap1,

0

100

200

300

400

TNFα

(pg/

mL)

Veh IM

BAL TNFα

#

LPS + VILI

2000

4000

6000

8000

10000

IL-6

(pg/

mL)

0Veh IM

BAL IL-6

LPS + VILI

0

200

400

600

TNFα

(pg/

mL)

Veh IM

BAL TNFα

LPS

2000

4000

6000

8000

10000

IL-6

(pg/

mL)

0Veh IM

BAL IL-6

LPS

A B

C D

Fig. 5. Imatinib decreases BAL TNF-� levels in aclinically relevant mouse model of acute lung injury(ALI). Inflammatory cytokines implicated in ALI(TNF-�, IL-6) were measured in the BAL fluid ofmice challenged with the 2-hit model (LPS VILI)(A and B) or the LPS-only postinjury model (C andD). 2-hit injury data represent SB (n � 3), SB imatinib(n � 3), LPS MV (n � 6), and LPS MV imatinib(n � 7). Post-LPS data represent SB (n � 3), SB imatinib (n � 3), LPS (n � 3–6), and LPS imatinib(n � 3–6). #P 0.05.

L1300 IMATINIB IS PROTECTIVE IN A MURINE MODEL OF LPS AND VILI

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00031.2015 • www.ajplung.org

by 10.220.33.1 on April 28, 2017

http://ajplung.physiology.org/D

ownloaded from

Page 8: Imatinib attenuates inflammation and vascular leak in a ...€¦ · CALL FOR PAPERS Translational Research in Acute Lung Injury and Pulmonary Fibrosis Imatinib attenuates inflammation

as well as by inhibition of actin-myosin contractility in EC(10). Additionally, both c-Abl and Arg are activated inresponse to oxidative stress (6, 7), and inhibition of c-Ablwith imatinib in murine microvascular EC has antioxidanteffects, including increasing protein expression of glutathi-one peroxidase 1 and catalase (45). However, despite thesebeneficial effects of Abl family kinase inhibition on ECpermeability, c-Abl is critically involved in the barrier-enhancing responses to both sphingosine 1-phosphate andFTY720 (18, 50). Additionally, loss of EC c-Abl causesincreased apoptosis and vascular dysfunction in EC-specificc-Abl knockout mice (11). Together, these observationssupport a model in which Abl family kinases are criticalmediators of vascular function with both protective anddisruptive effects in a stimulus-specific manner.

We recently reported that imatinib attenuates LPS-inducedVCAM-1 expression and inflammatory cytokine production(IL-6, IL-8) in cultured HPAEC (29). In EC, the process oftranscriptional activation of NF-�B mediates the expression ofmultiple cellular adhesion molecules (CAMs), includingICAM, VCAM-1, and E-selectin, that are involved in neutro-phil extravasation (13). Thus, in the current study, we exam-ined the effects of imatinib on NF-�B expression and activa-tion. Imatinib inhibited LPS-induced NF-�B phosphorylationand nuclear translocation in cultured human lung EC (Fig. 6)and NF-�B expression in mouse lung tissue (Fig. 7). Interest-ingly, imatinib did not alter VCAM-1 expression in the lungtissue of LPS-challenged mice (data not shown). Possibleexplanations for this observation include the multitude of celltypes present in the whole lung tissue as well as the time point

at which the animals were harvested relative to the expectedincrease in VCAM-1 expression.

In support of these data, recent work indicates that ima-tinib decreases NF-�B activation in macrophages and inhuman myeloid cells (12, 52), and anti-inflammatory effectsof imatinib have been reported in several inflammatory lungdiseases including idiopathic pulmonary fibrosis, pulmonaryarterial hypertension, and asthma (9, 14, 42), as well assystemic conditions, such as rheumatoid arthritis and mul-tiple sclerosis (2, 3). However, interestingly, the effects ofimatinib on inflammation and NF-�B activation appear tovary with cell type and length of exposure. In pancreatic�-cells, imatinib initially increases NF-�B activation, butafter a longer exposure it causes a dampened cytokineresponse (26, 34). Further exploration of the effects ofimatinib on inflammatory signaling in pulmonary EC mayprovide additional insights into its therapeutic potential inALI/ARDS and other inflammatory lung diseases (Fig. 8).Although in the present study we did not explore the effectsof imatinib on NF-�B signaling specifically in the VILI-alone model or the two-hit injury model, NF-�B activity isknown to contribute to the pathogenesis of VILI (24, 35,43), so it will be interesting in future experiments to char-acterize how imatinib alters VILI-induced NF-�B signaling.However, VILI is an extremely complex process in whichmultiple pathways are involved, including MAP kinasesignaling, actin-cytoskeletal rearrangements, and activationof cyclic adenosine monophosphate regulatory element-binding protein (30, 35, 38, 48), suggesting that the delete-rious effects of imatinib reported in the VILI model (29) are

Veh IM Veh IM Veh IM0

1

2

3

4

5NFκB Phosphorylation

Con LPS (30') LPS (60')

pNFκ

B/to

tal N

FκB

(nor

m to

Veh

/Con

)

**

A B

C

LegendRed: total NFκBBlue: DAPI

PBS LPS 30 min LPS 60 min

Veh

IM

pNFκB

NFκB

IκB

Actin

LPS (min)

Imatinib

-

-

-

+

30

+

30

-

60

+

60

-

Fig. 6. Imatinib inhibits LPS-induced NF-�Bphosphorylation and nuclear translocation invitro. Human pulmonary artery endothelialcells (HPAEC) were treated with imatinib(40 �M, 60 min) and then challenged withLPS (1 �g/ml, 0–60 min). Lysates wereharvested for Western blots for phosphory-lated NF-�B p65 (S536), total NF-�B p65protein, I�B, and actin and quantified bydensitometry (A and B). HPAEC plated onglass coverslips were subjected to identicalconditions, and immunofluorescence micros-copy was conducted to determine the local-ization of total NF-�B p65 protein (red).4=,6-diamidino-2-phenylindole (DAPI)(blue) was used to stain the nuclei (whitearrows) (C). Data are representative of 3independent experiments. *P 0.05 com-pared with nonchallenged samples.

L1301IMATINIB IS PROTECTIVE IN A MURINE MODEL OF LPS AND VILI

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00031.2015 • www.ajplung.org

by 10.220.33.1 on April 28, 2017

http://ajplung.physiology.org/D

ownloaded from

Page 9: Imatinib attenuates inflammation and vascular leak in a ...€¦ · CALL FOR PAPERS Translational Research in Acute Lung Injury and Pulmonary Fibrosis Imatinib attenuates inflammation

likely due to its effects on pathways other than NF-�Bsignaling.

An important limitation of our work is that imatinib inhibitsmultiple kinases, and therefore specific conclusions about themechanism responsible for its protective effects cannot bemade. For example, imatinib decreased PDGFR expression ina model of LPS-induced ALI in neutropenic mice (27), andinhibition of PDGFR with imatinib has been implicated in theresolution of vascular leak across the blood-brain barrier inmice (46). In our previous work, we observed that the effect ofimatinib on LPS-induced VCAM-1 upregulation was mediatedby c-Abl, but not Arg (29). Given that VCAM-1 upregulationoccurs downstream of NF-�B activation, we anticipate that theeffects of imatinib on NF-�B activation are mediated by c-Abl.

However, additional work is necessary to determine the mech-anisms underlying these protective effects in ALI, includingthe pathway through which imatinib inhibits LPS-inducedNF-�B activation.

In conclusion, we have demonstrated that imatinib isefficacious in two clinically relevant murine models of ALI.These results suggest that imatinib attenuates the inflamma-tion and vascular leak induced by LPS when combined withVILI and that these protective effects outweigh the delete-rious effects of this intervention previously reported in VILIalone. Multiple imatinib targets may be implicated in theseeffects, and additional studies are required to characterizetheir roles in the regulation of endothelial barrier integrityand inflammatory signaling pathways. Given that imatiniband several additional Abl family kinase inhibitors arecurrently in clinical use for multiple medical conditions(39), this work has potential for rapid translation intoclinical trials in patients.

GRANTS

This work was supported by NHLBI/NIH HL058064 (J. Garcia),F30HL121982 (A. Rizzo), AHA 14PRE18860021 (A. Rizzo), and Universityof Illinois institutional funds.

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

Author contributions: A.N.R., J.G.G., and S.M.D. conception and design ofresearch; A.N.R., S.S., A.E.E., and E.L. performed experiments; A.N.R.,J.G.G., and E.L. analyzed data; A.N.R., J.R.J., and S.M.D. interpreted resultsof experiments; A.N.R. prepared figures; A.N.R. drafted manuscript; A.N.R.and S.M.D. approved final version of manuscript; J.R.J., J.G.G., E.L., andS.M.D. edited and revised manuscript.

Veh IM0.0

0.5

1.0

1.5N

FκB

(rel

ativ

e to

act

in)

LPS

*

A

B C Veh LPS

Veh

IM

NFκB

Actin

LPS

Imatinib

+

-

+

-

+

-

+

-

+

-

+

-

+

+

+

+

+

+

+

+

+

+Fig. 7. Imatinib decreases NF-�B expressionin mouse lungs after LPS. Mice were chal-lenged with LPS (1.0 mg/kg, it) (vs. PBS) andthen received imatinib (75 mg/kg, ip) (vs.vehicle) 4 h later. Lung tissue homogenateswere collected, subjected to Western blottingfor total NF-�B p65 protein, and quantified bydensitometry (A and B). Lanes in A representlung homogenates from individual animals.Representative immunohistochemistry imagesare shown for NF-�B p65 taken by an indi-vidual blinded to experimental condition (C).*P 0.05.

Fig. 8. Potential protective effects of imatinib in lung endothelial inflammationinduced by LPS plus VILI. In this proposed schema, stimulation of lungendothelial cells (EC) by LPS VILI results in activation of the Abl kinasesand subsequent downstream signaling that includes increased NF-�B activity,upregulation of VCAM-1, release of cytokines (TNF-�, IL-6), and increasedpulmonary neutrophil (PMN) recruitment. Imatinib inhibits Abl family kinasesto attenuate these effects in vitro and in vivo. Nuc, nucleus.

L1302 IMATINIB IS PROTECTIVE IN A MURINE MODEL OF LPS AND VILI

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00031.2015 • www.ajplung.org

by 10.220.33.1 on April 28, 2017

http://ajplung.physiology.org/D

ownloaded from

Page 10: Imatinib attenuates inflammation and vascular leak in a ...€¦ · CALL FOR PAPERS Translational Research in Acute Lung Injury and Pulmonary Fibrosis Imatinib attenuates inflammation

REFERENCES

1. Ventilation with lower tidal volumes compared with traditional tidalvolumes for acute lung injury, and the acute respiratory distress syndrome.The Acute Respiratory Distress Syndrome Network. N Engl J Med 342:1301–1308, 2000.

2. Adzemovic MZ, Zeitelhofer M, Eriksson U, Olsson T, Nilsson I.Imatinib ameliorates neuroinflammation in a rat model of multiple scle-rosis by enhancing blood-brain barrier integrity and by modulating theperipheral immune response. PLoS One 8: e56586, 2013.

3. Akashi N, Matsumoto I, Tanaka Y, Inoue A, Yamamoto K, Umeda N,Hayashi T, Goto D, Ito S, Sekiguchi K, Sumida T. Comparativesuppressive effects of tyrosine kinase inhibitors imatinib and nilotinib inmodels of autoimmune arthritis. Mod Rheumatol 21: 267–275, 2011.

4. Aman J, Peters MJ, Weenink C, van Nieuw Amerongen GP, VonkNoordegraaf A. Reversal of vascular leak with imatinib. Am J Respir CritCare Med 188: 1171–1173, 2013.

5. Aman J, van Bezu J, Damanafshan A, Huveneers S, Eringa EC, VogelSM, Groeneveld AB, Vonk Noordegraaf A, van Hinsbergh VW, vanNieuw Amerongen GP. Effective treatment of edema and endothelialbarrier dysfunction with imatinib. Circulation 126: 2728–2738, 2012.

6. Cao C, Leng Y, Huang W, Liu X, Kufe D. Glutathione peroxidase 1 isregulated by the c-Abl and Arg tyrosine kinases. J Biol Chem 278:39609–39614, 2003.

7. Cao C, Leng Y, Kufe D. Catalase activity is regulated by c-Abl and Argin the oxidative stress response. J Biol Chem 278: 29667–29675, 2003.

8. Carnevale-Schianca F, Gallo S, Rota-Scalabrini D, Sangiolo D, Fiz-zotti M, Caravelli D, Capaldi A, Anselmetti G, Palesandro E,D’Ambrosio L, Coha V, Obert R, Aglietta M, Grignani G. Completeresolution of life-threatening bleomycin-induced pneumonitis after treat-ment with imatinib mesylate in a patient with Hodgkin’s lymphoma: hopefor severe chemotherapy-induced toxicity? J Clin Oncol 29: e691–693,2011.

9. Chen A, Lee SM, Gao B, Shannon S, Zhu Z, Fang D. c-Abl-mediatedtyrosine phosphorylation of the T-bet DNA-binding domain regulatesCD4 T-cell differentiation and allergic lung inflammation. Mol Cell Biol31: 3445–3456, 2011.

10. Chislock EM, Pendergast AM. Abl family kinases regulate endothelialbarrier function in vitro and in mice. PLoS One 8: e85231, 2013.

11. Chislock EM, Ring C, Pendergast AM. Abl kinases are required forvascular function, Tie2 expression, and angiopoietin-1-mediated survival.Proc Natl Acad Sci USA 110: 12432–12437, 2013.

12. Ciarcia R, Vitiello MT, Galdiero M, Pacilio C, Iovane V, d’Angelo D,Pagnini D, Caparrotti G, Conti D, Tomei V, Florio S, Giordano A.Imatinib treatment inhibit IL-6, IL-8, NF-KB and AP-1 production andmodulate intracellular calcium in CML patients. J Cell Physiol 227:2798–2803, 2012.

13. Collins T, Read MA, Neish AS, Whitley MZ, Thanos D, Maniatis T.Transcriptional regulation of endothelial cell adhesion molecules: NF-�Band cytokine-inducible enhancers. FASEB J 9: 899–909, 1995.

14. Daniels CE, Wilkes MC, Edens M, Kottom TJ, Murphy SJ, LimperAH, Leof EB. Imatinib mesylate inhibits the profibrogenic activity ofTGF-beta and prevents bleomycin-mediated lung fibrosis. J Clin Invest114: 1308–1316, 2004.

15. de Ménorval MA, Mir LM, Fernández ML, Reigada R. Effects ofdimethyl sulfoxide in cholesterol-containing lipid membranes: a compar-ative study of experiments in silico and with cells. PLoS One 7: e41733,2012.

16. Druker BJ, Guilhot F, O’Brien SG, Gathmann I, Kantarjian H,Gattermann N, Deininger MW, Silver RT, Goldman JM, Stone RM,Cervantes F, Hochhaus A, Powell BL, Gabrilove JL, Rousselot P,Reiffers J, Cornelissen JJ, Hughes T, Agis H, Fischer T, Verhoef G,Shepherd J, Saglio G, Gratwohl A, Nielsen JL, Radich JP, SimonssonB, Taylor K, Baccarani M, So C, Letvak L, Larson RA, InvestigatorsI. Five-year follow-up of patients receiving imatinib for chronic myeloidleukemia. N Engl J Med 355: 2408–2417, 2006.

17. Druker BJ, Talpaz M, Resta DJ, Peng B, Buchdunger E, Ford JM,Lydon NB, Kantarjian H, Capdeville R, Ohno-Jones S, Sawyers CL.Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinasein chronic myeloid leukemia. N Engl J Med 344: 1031–1037, 2001.

18. Dudek SM, Chiang ET, Camp SM, Guo Y, Zhao J, Brown ME,Singleton PA, Wang L, Desai A, Arce FT, Lal R, Van Eyk JE, ImamSZ, Garcia JG. Abl tyrosine kinase phosphorylates nonmuscle Myosin

light chain kinase to regulate endothelial barrier function. Mol Biol Cell21: 4042–4056, 2010.

19. Dudek SM, Garcia JG. Cytoskeletal regulation of pulmonary vascularpermeability. J Appl Physiol 91: 1487–1500, 2001.

20. Erickson SE, Martin GS, Davis JL, Matthay MA, Eisner MD, NIHNHLBI ARDS Network. Recent trends in acute lung injury mortality:1996–2005. Crit Care Med 37: 1574–1579, 2009.

21. Garcia AN, Vogel SM, Komarova YA, Malik AB. Permeability ofendothelial barrier: cell culture and in vivo models. Methods Mol Biol 763:333–354, 2011.

22. Goldman JL, Sammani S, Kempf C, Saadat L, Letsiou E, Wang T,Moreno-Vinasco L, Rizzo AN, Fortman JD, Garcia JG. Pleiotropiceffects of interleukin-6 in a “two-hit” murine model of acute respiratorydistress syndrome. Pulm Circ 4: 280–288, 2014.

23. Goldsby R, Pulsipher M, Adams R, Coffin C, Albritton K, Wagner L.Unexpected pleural effusions in 3 pediatric patients treated with STI-571.J Pediatr Hematol Oncol 24: 694–695, 2002.

24. Held HD, Boettcher S, Hamann L, Uhlig S. Ventilation-induced chemo-kine and cytokine release is associated with activation of nuclear factor-kappaB and is blocked by steroids. Am J Respir Crit Care Med 163:711–716, 2001.

25. Hotchkiss RS, Karl IE. The pathophysiology and treatment of sepsis. NEngl J Med 348: 138–150, 2003.

26. Hägerkvist R, Sandler S, Mokhtari D, Welsh N. Amelioration ofdiabetes by imatinib mesylate (Gleevec): role of �-cell NF-�B activationand anti-apoptotic preconditioning. FASEB J 21: 618–628, 2007.

27. Kim IK, Rhee CK, Yeo CD, Kang HH, Lee DG, Lee SH, Kim JW.Effect of tyrosine kinase inhibitors, imatinib and nilotinib, in murinelipopolysaccharide-induced acute lung injury during neutropenia recovery.Crit Care 17: R114, 2013.

28. Ko YA, Yang MC, Huang HT, Hsu CM, Chen LW. NF-�B activationin myeloid cells mediates ventilator-induced lung injury. Respir Res 14:69, 2013.

29. Letsiou E, Rizzo AN, Sammani S, Naureckas P, Jacobson JR, GarciaJG, Dudek SM. Differential and opposing effects of imatinib on LPS- andventilator-induced lung injury. Am J Physiol Lung Cell Mol Physiol 308:L259–L269, 2015.

30. Li LF, Yu L, Quinn DA. Ventilation-induced neutrophil infiltrationdepends on c-Jun N-terminal kinase. Am J Respir Crit Care Med 169:518–524, 2004.

31. Lin SM, Lin HC, Lee KY, Huang CD, Liu CY, Wang CH, Kuo HP.Ventilator-induced injury augments interleukin-1beta production and neu-trophil sequestration in lipopolysaccharide-treated lungs. Shock 28: 453–460, 2007.

32. Liu YY, Liao SK, Huang CC, Tsai YH, Quinn DA, Li LF. Role fornuclear factor-kappaB in augmented lung injury because of interactionbetween hyperoxia and high stretch ventilation. Transl Res 154: 228–240,2009.

33. Moitra J, Sammani S, Garcia JG. Re-evaluation of Evans Blue dye asa marker of albumin clearance in murine models of acute lung injury.Transl Res 150: 253–265, 2007.

34. Mokhtari D, Li T, Lu T, Welsh N. Effects of Imatinib Mesylate(Gleevec) on human islet NF-kappaB activation and chemokine produc-tion in vitro. PLoS One 6: e24831, 2011.

35. Ning QM, Wang XR. Activations of mitogen-activated protein kinase andnuclear factor-kappaB by mechanical stretch result in ventilation-inducedlung injury. Med Hypotheses 68: 356–360, 2007.

36. Overbeek MJ, van Nieuw Amerongen GP, Boonstra A, Smit EF,Vonk-Noordegraaf A. Possible role of imatinib in clinical pulmonaryveno-occlusive disease. Eur Respir J 32: 232–235, 2008.

37. Peng B, Lloyd P, Schran H. Clinical pharmacokinetics of imatinib. ClinPharmacokinet 44: 879–894, 2005.

38. Pugin J. Molecular mechanisms of lung cell activation induced by cyclicstretch. Crit Care Med 31: S200–S206, 2003.

39. Rix U, Hantschel O, Dürnberger G, Remsing Rix LL, Planyavsky M,Fernbach NV, Kaupe I, Bennett KL, Valent P, Colinge J, Köcher T,Superti-Furga G. Chemical proteomic profiles of the BCR-ABL inhibi-tors imatinib, nilotinib, and dasatinib reveal novel kinase and nonkinasetargets. Blood 110: 4055–4063, 2007.

40. Sakurai H, Chiba H, Miyoshi H, Sugita T, Toriumi W. IkappaB kinasesphosphorylate NF-kappaB p65 subunit on serine 536 in the transactivationdomain. J Biol Chem 274: 30353–30356, 1999.

L1303IMATINIB IS PROTECTIVE IN A MURINE MODEL OF LPS AND VILI

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00031.2015 • www.ajplung.org

by 10.220.33.1 on April 28, 2017

http://ajplung.physiology.org/D

ownloaded from

Page 11: Imatinib attenuates inflammation and vascular leak in a ...€¦ · CALL FOR PAPERS Translational Research in Acute Lung Injury and Pulmonary Fibrosis Imatinib attenuates inflammation

41. Scheinfeld N. Imatinib mesylate and dermatology part 2: a review of thecutaneous side effects of imatinib mesylate. J Drugs Dermatol 5: 228–231, 2006.

42. Schermuly RT, Dony E, Ghofrani HA, Pullamsetti S, Savai R, RothM, Sydykov A, Lai YJ, Weissmann N, Seeger W, Grimminger F.Reversal of experimental pulmonary hypertension by PDGF inhibition. JClin Invest 115: 2811–2821, 2005.

43. Shu YS, Tao W, Miao QB, Zhu YB, Yang YF. Improvement ofventilation-induced lung injury in a rodent model by inhibition of inhib-itory �B kinase. J Trauma Acute Care Surg 76: 1417–1424, 2014.

44. Slutsky AS, Ranieri VM. Ventilator-induced lung injury. N Engl J Med369: 2126–2136, 2013.

45. Stephens RS, Servinsky LE, Rentsendorj O, Kolb TM, Pfeifer A,Pearse DB. Protein kinase G increases antioxidant function in lungmicrovascular endothelial cells by inhibiting the c-Abl tyrosine kinase. AmJ Physiol Cell Physiol 306: C559–C569, 2014.

46. Su EJ, Fredriksson L, Geyer M, Folestad E, Cale J, Andrae J, Gao Y, PietrasK, Mann K, Yepes M, Strickland DK, Betsholtz C, Eriksson U, LawrenceDA. Activation of PDGF-CC by tissue plasminogen activator impairs blood-brainbarrier integrity during ischemic stroke. Nat Med 14: 731–737, 2008.

47. Tan SY, Kan E, Lim WY, Chay G, Law JH, Soo GW, Bukhari NI,Segarra I. Metronidazole leads to enhanced uptake of imatinib in brain,liver and kidney without affecting its plasma pharmacokinetics in mice. JPharm Pharmacol 63: 918–925, 2011.

48. Uhlig U, Haitsma JJ, Goldmann T, Poelma DL, Lachmann B, Uhlig S.Ventilation-induced activation of the mitogen-activated protein kinasepathway. Eur Respir J 20: 946–956, 2002.

49. Wadgaonkar R, Linz-McGillem L, Zaiman AL, Garcia JG. Endothe-lial cell myosin light chain kinase (MLCK) regulates TNFalpha-inducedNFkappaB activity. J Cell Biochem 94: 351–364, 2005.

50. Wang L, Chiang ET, Simmons JT, Garcia JG, Dudek SM. FTY720-induced human pulmonary endothelial barrier enhancement is mediated byc-Abl. Eur Respir J 38: 78–88, 2011.

51. Ware LB, Matthay MA. The acute respiratory distress syndrome. N EnglJ Med 342: 1334–1349, 2000.

52. Wolf AM, Wolf D, Rumpold H, Ludwiczek S, Enrich B, Gastl G,Weiss G, Tilg H. The kinase inhibitor imatinib mesylate inhibits TNF-alpha production in vitro and prevents TNF-dependent acute hepaticinflammation. Proc Natl Acad Sci USA 102: 13622–13627, 2005.

L1304 IMATINIB IS PROTECTIVE IN A MURINE MODEL OF LPS AND VILI

AJP-Lung Cell Mol Physiol • doi:10.1152/ajplung.00031.2015 • www.ajplung.org

by 10.220.33.1 on April 28, 2017

http://ajplung.physiology.org/D

ownloaded from