Background

Alveolar epithelial cell apoptosis is implicated in the onset of ventilator-induced lung injury. Death-associated protein kinase 1 (DAPK1) is associated with cell apoptosis. The hypothesis was that DAPK1 participates in ventilator-induced lung injury through promoting alveolar epithelial cell apoptosis.

Methods

Apoptosis of mouse alveolar epithelial cell was induced by cyclic stretch. DAPK1 expression was altered (knockdown or overexpressed) in vitro by using a small interfering RNA or a plasmid, respectively. C57/BL6 male mice (n = 6) received high tidal volume ventilation to establish a lung injury model. Adeno-associated virus transfection of short hairpin RNA and DAPK1 inhibitor repressed DAPK1 expression and activation in lungs, respectively. The primary outcomes were alveolar epithelial cell apoptosis and lung injury.

Results

Compared with the control group, the 24-h cyclic stretch group showed significantly higher alveolar epithelial cell apoptotic percentage (45 ± 4% fold vs. 6 ± 1% fold; P < 0.0001) and relative DAPK1 expression, and this group also demonstrated a reduced apoptotic percentage after DAPK1 knockdown (27 ± 5% fold vs. 53 ± 8% fold; P < 0.0001). A promoted apoptotic percentage in DAPK1 overexpression was observed without stretching (49 ± 6% fold vs. 14 ± 3% fold; P < 0.0001). Alterations in B-cell lymphoma 2 and B-cell lymphoma 2–associated X are associated with DAPK1 expression. The mice subjected to high tidal volume had higher DAPK1 expression and alveolar epithelial cell apoptotic percentage in lungs compared with the low tidal volume group (43 ± 6% fold vs. 4 ± 2% fold; P < 0.0001). Inhibition of DAPK1 through adeno-associated virus infection or DAPK1 inhibitor treatment appeared to be protective against lung injury with reduced lung injury score, resolved pulmonary inflammation, and repressed alveolar epithelial cell apoptotic percentage (47 ± 4% fold and 48 ± 6% fold; 35 ± 5% fold and 34 ± 4% fold; P < 0.0001, respectively).

Conclusions

DAPK1 promotes the onset of ventilator-induced lung injury by triggering alveolar epithelial cell apoptosis through intrinsic apoptosis pathway in mice.

Editor’s Perspective
What We Already Know about This Topic
  • Ventilator-related lung injury may be related to stretch-induced apoptosis

  • DAPK1 is involved in various apoptotic signal transduction pathways

  • The role of DAPK1 in ventilator-related lung injury is not well understood

What This Article Tells Us That Is New
  • In mice, high tidal volumes increased cyclic stretch, DAPK1 expression, and epithelial cell apoptosis

  • Inhibition of DAPK1 appeared to be protective against lung injury, reducing lung injury, inflammation, and apoptosis

  • DAPK1 triggers alveolar epithelial cell apoptosis and mediates ventilator-induced lung injury in mice

Mechanical ventilation provides vital life support for patients with acute respiratory distress syndrome. Unfortunately, ventilation can lead to lung stretch or even injury (ventilator-induced lung injury) and attendant adverse outcomes.1,2  Although low tidal volume can improve the outcome for patients with ventilator-induced lung injury, there is still considerable distension of some parts of the injured lung due to widespread injury, consolidation, and atelectasis.3  Thus, alveolar epithelial cell overdistension cannot be completely avoided during mechanical ventilation.

Stretch-induced apoptosis in vivo was first demonstrated in fetal rabbits after tracheal occlusion.4  An in vitro study showed that physiologic ventilation caused mechanical stretch of alveolar epithelial cells, leading to calcium signaling and the secretion of surfactant proteins.5  A moderate mechanical stretch, which is capable of increasing the cell surface area by 30% and stimulating lung inflation to ~100% total lung capacity, was sufficient to induce alveolar epithelial cell apoptosis and necrosis.6 

The ability to modulate the life or apoptosis of a cell has an immense therapeutic potential. Because the apoptosis of alveolar epithelial cells destroys the epithelial barrier of the lung and leads to subsequent ventilator-induced lung injury, the prevention of mechanical stretch–induced alveolar epithelial cell apoptosis and the restoration of normal alveolar epithelial function are crucial. However, the specific mechanism under the mechanical stretch–induced alveolar epithelial cell apoptosis is unknown.

Apoptosis is a gene-regulated and programed cell death that can be activated by various proteins. Death-associated protein kinase 1 (DAPK1) is a proapoptotic Ca2+/calmodulin–regulated serine/threonine kinase involving in the regulation of both apoptosis and necrosis.7,8  It was originally described as a positive regulator of interferon-γ–mediated cell death.9  As a proapoptotic factor, DAPK1 is involved in various apoptotic signal transduction pathways—both extrinsic and intrinsic pathways.10,11  Various death stimuli have been associated with an elevation of DAPK levels and catalytic activity.12,13  In endothelial cells, shear stress independently upregulated DAPK gene transcription and protein expression, which in turn increased apoptosis.14 

Typical markers for DAPK1 activation include a dephosphorylation at serine 308 of DAPK1 and an increased amount of DAPK1 protein.15  An activated DAPK1 phosphorylates the myosin light chain to form phosphorylated myosin light chain.16  Therefore, the phosphorylated myosin light chain level reflects the extent of DAPK1 activation.

The present study investigated the association between DAPK1 and ventilator-induced lung injury in mice. First, in vitro, it was determined whether DAPK1 activation was elevated in the presence of cyclic stretch–induced alveolar epithelial cell apoptosis. Second, in vitro, an assessment was conducted of whether cyclic stretch–induced alveolar epithelial cell apoptosis was enhanced by DAPK1. Finally, we tested whether the induction of alveolar epithelial cell apoptosis by DAPK1 was associated with the development of ventilator-induced lung injury in vivo.

In Vitro Experiments

Cell Culture.

Mouse alveolar epithelial cells (MLE-12; American Type Culture Collection, USA) were cultured and passaged in Dulbecco’s modified Eagle’s medium supplemented with 10% fetal bovine serum (Gibco, USA) and antibiotics (100 units/ml penicillin, 100 μg/ml streptomycin). The culture was maintained in a humidified atmosphere containing 5% carbon dioxide at 37°C. The cells were harvested for experiments until 70 to 80% confluent.

Cell Stretching.

Mouse alveolar epithelial cells (5 × 105/well) were inoculated on collagen 1–precoated Bioflex plates. After a 24-h culture, the medium was replaced by fresh medium containing 0.5% fetal bovine serum for another 12 h.

The cells were randomly split for cyclic stretch, static stretch, or no stretch. The cells in the cyclic stretch group were exposed to cyclic stretch by using an FX-5000T Flexercell Tension Plus system (Flexcell International, USA), at 30 cycles/min and 30% amplitude. The stretching times were 6, 12, or 24 h. For static stretching, a pattern with 30% elongation for up to 24 h was used. The cells in the control group were sown on Bioflex plates treated in the same manner as the stretched cells, but they were not subjected to any stretch. After 6, 12, or 24-h culture, the cells and the culturing media were collected for further detection.

Small Interfering RNA Intervention.

The silencing of DAPK1 expression in mouse alveolar epithelial cells was conducted via small interfering RNA. Three different DAPK1 small interfering RNAs were used (Supplemental Digital Content, table S1, https://links.lww.com/ALN/C452). The small interfering RNA and transfection reagent were purchased from RiboBio (China).

The cells (5 × 105/well) were sown on Bioflex plates. The cells were divided into three groups: control, control small interfering RNA + cyclic stretch, and DAPK1 small interfering RNA + cyclic stretch. After 24 h, the transfection mixture (small interfering RNA and transfection reagent) was prepared in a final volume of 1,000 μl to achieve a final small interfering RNA concentration of 100 nmol/l. This mixture was incubated for 7 min at room temperature to allow complex formation and then added drop-by-drop onto the cells. A control small interfering RNA was used as a negative control.

After a 12-h incubation, the medium was replaced by fresh medium containing 0.5% fetal bovine serum, and the cells were stretched at 30 cycles/min for 24 h to 30% elongation. The cells in the control group were not stretched. Then the cells and culture media were collected for further procedures.

DAPK1ΔCalmodulin Plasmid Transfection.

DAPK1 Δcalmodulin is an active form of the death-associated protein kinase, which lacks the calmodulin regulatory and binding domain (i.e. amino acids 266 to 312; named DAPK1Δcalmodulin). The corresponding plasmid for DAPK1Δcalmodulin was constructed as described in a previous report.17 

Mouse alveolar epithelial cells (3 × 105) were seeded in a 6-well culture plate for 24 h. A transfection mixture (i.e., plasmid, Lipofectamine 2000, and culture medium) was prepared to a final volume of 500 μl with 0.01 μg/μl DAPK1Δcalmodulin vector or control vector (empty plasmid). Lipofectamine 2000 transfection reagent (Invitrogen, USA) was used by following the manufacturer’s instructions. The transfection mixture was incubated with cells for 6 h. Then the medium was changed to Dulbecco’s modified Eagle’s medium with 10% fetal bovine serum. After another 48-h culture, the cells were collected for further detection.

Lactic Acid Dehydrogenase Measurement.

The media from the cultures of stretch cells and control cells were analyzed for lactic acid dehydrogenase activity by using a lactic acid dehydrogenase assay kit in accordance with the manufacturer’s instructions (Beyotime, China). The kit was calibrated using lactic acid dehydrogenase standards at 2 μg/ml.

Apoptosis Analysis.

Cell apoptosis was analyzed with an annexin V–fluorescein isothiocyanate apoptosis detection kit (BD Pharmingen, USA). The cells were digested by trypsin (0.05%) EDTA (0.2 g/l) for 90 s. Both attached and nonattached cells were harvested and stained with fluorescein isothiocyanate–conjugated annexin V and propidium iodide and then analyzed by flow cytometry (FACSort). Alveolar epithelial cell apoptosis was expressed as a percentage of annexin V–positive cells to total cells.

Caspase 3 Activity Assay.

Caspase 3 activity was analyzed using a caspase 3 activity assay kit (Beyotime) in accordance with the manufacturer’s instructions. Briefly, the cell lysate supernatant was mixed with buffer containing the substrate peptides for caspase attached to p-nitroanilide. The release of p-nitroanilide was quantified by measuring the absorbance at 405 nm by an enzyme-linked immunosorbent assay reader. The caspase activities were expressed as a percentage of the control.

In Vivo Experiments

Mouse Model.

The Ethics Committee of Huazhong University of Science and Technology approved and supervised this study. The animal care and use committee of Tongji Medical College of Huazhong University of Science and Technology permitted all the animal experiments.

Healthy male C57BL/6 mice (specific pathogen-free grade, aged 6 to 8 week) weighing 20 to 22 g were purchased from Wuhan University (Wuhan, China). All mice were housed in specific pathogen-free conditions for 5 days before experimental use. The mice were tested in sequential order.

For adeno-associated virus infection, DAPK1 inhibitor pretreatment, and mechanical ventilation, experiments were performed in specific pathogen-free environment with fixed temperature and humidity. The other experiments were performed in our laboratory. All the animal experiments were performed in the morning (8 to 10 am).

All the mice were anesthetized with intraperitoneal injections of ketamine (80 mg/kg) and xylazine (10 mg/kg) and intubated through the trachea. The mice were ventilated (ventilator model: Harvard, 55-7062) either at 8 ml/kg with 120 breaths/min (low tidal volume, low VT), or at 35 ml/kg with 80 breaths/min (high tidal volume, high VT) for 4 h; the positive end-expiratory pressure was set at nil for animals subjected to ventilation, whereas the control mice underwent tracheotomy but breathed spontaneously. All animals were ventilated with room air.

For some experiments, mice received DAPK1 inhibitor ((4Z)-2-[(E)-2-phenylethenyl)-4-(3-pyridinylmethylene)-5(4H)-oxazolone, 500 μg · kg−1 · day−1, intraperitoneally; Merck, Germany) or an identical dose of placebo-control solution (dimethyl sulfoxide) for 6 weeks. The mice were then ventilated at 35 ml/kg at 80 breaths/min for 4 h. All animals were euthanized by exsanguination at the end of the mechanical ventilation. The primary outcomes of the animal experiments were lung injury score and alveolar epithelial cell apoptosis.

Adeno-associated Virus Infection.

A small hairpin RNA (Supplemental Digital Content, table S1, https://links.lww.com/ALN/C452) used for gene knockdown of DAPK1 was expressed from plasmid with a U6 promoter by using an adeno-associated virus expression system (DAPK1 virus; Shanghai Genechem, China). The same adeno-associated virus containing a scramble small hairpin RNA sequence for nonmammalian gene was used as the control (referred to here as the “control-virus”).

C57BL/6 mice were divided into three groups: the control group, the control-virus group + high VT, and the DAPK1-virus group + high VT. The mice were anesthetized with intraperitoneal injections of ketamine (80 mg/kg) and xylazine (10 mg/kg) and intubated through the trachea with a 20-gauge needle. The mice were inoculated through the trachea with 106 genomic copies of DAPK1-virus in a final volume of 30 μl. The same dose of control-virus (106 genomic copies, 30 μl) was administered through the trachea to the control-virus + high VT group. Control mice received phosphate-buffered saline only. The mice in the control-virus + high VT and DAPK1-virus + high VT groups received high tidal volume ventilation (35 ml/kg, 4 h) 30 days after the adeno-associated virus instillation. Control mice received no treatment. All animals were euthanized by exsanguination at the end of mechanical ventilation.

Histologic Analysis.

Right middle lung specimens were fixed in 4% paraformaldehyde and embedded in paraffin. The embedded lung tissues were divided into 4-μm sections for staining with hematoxylin and eosin for light microscopy. Lung injury pathologic changes were blindly examined by two histologists without knowing the experimental groups. Images were captured randomly from five nonoverlapping fields per slide. Three slides were captured for each animal, and six animals were included for each group. The lung injury score calculated by following a previously published criteria (Supplemental Digital Content, table S2, https://links.lww.com/ALN/C453),18  in which each animal was examined as the unit of analysis.

Lung Wet-to-Dry Ratio.

The lung wet-to-dry ratio was used as an index of pulmonary edema formation. The wet right middle lungs were weighed and recorded as the wet weight. Then they were weighed after the incubation in an oven at 60°C for 72 h and recorded as the dry weight.

Bronchoalveolar Lavage Analysis.

Bronchoalveolar lavage was performed with phosphate-buffered saline (3×0.5 ml) and analyzed as described.19  The total number of bronchoalveolar lavage cells was determined by a hemocytometer. The protein concentration in bronchoalveolar lavage was measured by a bicinchoninic acid assay kit (Beyotime).

Cytokine Enzyme-linked Immunosorbent Assay.

Bronchoalveolar lavage and lung tissues were measured for tumor necrosis factor-α, interleukin-6, and interleukin-1β by using commercially available enzyme-linked immunosorbent assay kits (Elabscience Biotechnology Company, China).

Immunohistochemistry and Cell Death Analysis.

In animal models, alveolar epithelial cell apoptosis was visualized with terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate in situ nick-end labeling staining. Lungs were pressure-fixed and processed for paraffin embedding. Paraffin-embedded lung sections were deparaffinized and rehydrated in a graded ethanol series. After antigen retrieval and quenching of endogenous peroxidase, the sections were incubated with anti-cytokeratin 18 antibody (1:200 dilution; Abcam, China) at 4 °C overnight.

On the second day, after phosphate-buffered saline wash, The sections were incubated with cell apoptosis reaction mixture (Roche, Canada) for 1 h. The sections were washed and incubated with secondary antibodies at room temperature for 1 h. 4,6-Diamino-2-phenylindole was added to stain the sections for 5 min. Images of the stained lung sections were analyzed under a confocal microscope. Images were captured as described for histologic analysis. Alveolar epithelial cell apoptosis was examined with each animal as the unit of analysis. The data were presented by a percentage of double-positive (terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate in situ nick-end labeling staining and anti-cytokeratin 18 staining) cells to total cells observed in the field.

Western Blot.

At the end of the experiments, total protein extracts were subjected to sodium dodecyl sulfate–polyacrylamide gel electrophoresis and then transferred to polyvinylidene difluoride membranes as previously reported.20  The transformed membranes were probed with the following primary antibodies: DAPK1 (1:1,000; Sigma–Aldrich, USA), phosphorylated DAPK1 (1:100; Cell Signaling Technology, USA), phosphorylated myosin light chain (1:500; Cell Signaling Technology), myosin light chain (1:1,000; Abcam, USA), B-cell lymphoma 2 (1:1,000; Santa Cruz, USA), and B-cell lymphoma 2–associated X (1:1,000; Santa Cruz).

Statistical Analysis

No statistical power calculation was performed before the study; sample sizes were the estimates based on experience and preliminary data. Except for the evaluation of the lung injury score, investigators were not blinded during data collection or analysis. Outliers were evaluated with no necessary action. No data was excluded or missing. The data are presented in scatter plots (n ≤ 4) or bar graphs (n ≥ 5) with means ± SD. The distribution of all data was tested for normality with Shapiro–Wilk tests. ANOVA was two-way repeated, and then the Bonferroni post hoc test was used for analyzing changes in lactic acid dehydrogenase concentration, DAPK1 levels and activation at different time points. Two independent sample t tests were performed for analysis of the changes in two-group comparisons. One-way ANOVA and the Bonferroni post hoc test were used for comparing more than two groups. All the statistical analyses were performed using Prism 5 (GraphPad Software, USA). A two-tailed P value less than 0.05 was considered statistically significant.

Alveolar Epithelial Cell Apoptosis and DAPK1 Activation in Response to Cyclic Stretch

In this study, cyclic stretch was utilized as a stress model to simulate ventilator-induced lung injury. To characterize alveolar epithelial cell under this stress model, several cellular and molecular responses toward cyclic stretch of different durations were compared (fig. 1). Although 6-h stretch had little impact to the cell morphology throughout the culture, most of the cells are round and clustered together after a 12-h stretch, and nearly all cells were detached and suspended in the culture medium after a 24-h stretch (fig. 1A).

Fig. 1.

Cell apoptosis profiles and death-associated protein kinase 1 (DAPK1) activation in mouse alveolar epithelial cells subjected to cyclic stretch at 30% elongation for 6, 12, or 24 h. (A) Cell morphology was observed by an optical microscope. (B) The percentage of cell apoptosis was examined by flow cytometry. (C) Lactic acid dehydrogenase concentration was measured in the media of the corresponding cultures. (D, E) Protein levels of DAPK1 (D), phosphorylated DAPK1 (E), myosin light chain (myosin light chain), and phosphorylated myosin light chain (MLC) was measured by Western blot. (F) The phosphorylated MLC-to-MLC ratio was calculated. The scatter graphs are from four experiments. #P < 0.05 compared with the control group at the same time points.

Fig. 1.

Cell apoptosis profiles and death-associated protein kinase 1 (DAPK1) activation in mouse alveolar epithelial cells subjected to cyclic stretch at 30% elongation for 6, 12, or 24 h. (A) Cell morphology was observed by an optical microscope. (B) The percentage of cell apoptosis was examined by flow cytometry. (C) Lactic acid dehydrogenase concentration was measured in the media of the corresponding cultures. (D, E) Protein levels of DAPK1 (D), phosphorylated DAPK1 (E), myosin light chain (myosin light chain), and phosphorylated myosin light chain (MLC) was measured by Western blot. (F) The phosphorylated MLC-to-MLC ratio was calculated. The scatter graphs are from four experiments. #P < 0.05 compared with the control group at the same time points.

Close modal

Consistent with the morphology changes, the differential apoptotic percentage started to be significant in the 12-h stretch group by flow cytometry analysis compared with the nonstretched control, which was even more severe after a 24-h stretch (45 ± 4% fold vs. 6 ± 1% fold; P < 0.0001; n = 4/group; fig. 1B). Cell necrosis was not significantly increased in any of these three groups (data not shown). The lactic acid dehydrogenase concentration in the stretched cells was elevated along with the increase in the stretch duration (fig. 1C).

At the molecular level, only the 24-h group showed a significant increase in DAPK1 expression in the stretched cells over the nonstretched control (1.8 ± 0.2 fold vs. 0.5 ± 0.1 fold; P < 0.0001; n = 4/group; fig. 1D), whereas the phosphorylated DAPK1 (the inactive form of DAPK1) was significantly reduced (0.5 ± 0.2 fold vs. 1.3 ± 0.2 fold; P < 0.0001; n = 4/group; fig. 1E). The ratio of phosphorylated myosin light chain (a catalytic substrate of DAPK1)–to–myosin light chain was higher after 24 h of stretching (2.0 ± 0.3 fold vs. 0.6 ± 0.1 fold; P < 0.0001; n = 4/group; fig. 1F).

To confirm whether the above observed changes were specific to the cyclic stretch model, the same evaluation was done for static stretch model (n = 4/group). A morphologic change was also observed after static stretch of cells (Supplemental Digital Content, fig. S1A, https://links.lww.com/ALN/C444). However, compared with control group, there was no significant change observed in lactic acid dehydrogenase concentration and the apoptotic percentage of cells subjected to static stretch (Supplemental Digital Content, fig. S1, B–D, https://links.lww.com/ALN/C444). The levels of DAPK1 and phosphorylated DAPK1 and the phosphorylated myosin light chain–to–myosin light chain ratio were also similar between the stretched and control cells (Supplemental Digital Content, fig. S1, E–G, https://links.lww.com/ALN/C444).

These results indicated that it was not likely that static stretching could induce alveolar epithelial cell apoptosis or DAPK1 activation. Therefore, static stretching was not chosen in the experiments following.

Effect of DAPK1 Knockdown on Cyclic Stretch–induced Alveolar Epithelial Cell Apoptosis

To determine whether the expression of DAPK1 affects cyclic stretch–induced alveolar epithelial cell apoptosis, we knocked down DAPK1 by using small interfering RNA in vitro. DAPK1 small interfering RNA transfection alone had no effect on cell morphology, apoptosis, or myosin light chain phosphorylation but verified the transfection efficiency (Supplemental Digital Content, fig. S2, https://links.lww.com/ALN/C445). Meanwhile, the cyclic stretch–induced DAPK1 activation was also inhibited after DAPK1 small interfering RNA transfection (Supplemental Digital Content, fig. S3, https://links.lww.com/ALN/C446). The changes in morphology after cyclic stretch in control small interfering RNA transfected cells were not observed in the DAPK1 small interfering RNA transfected cells. The lactic acid dehydrogenase release in DAPK1 small interfering RNA transfected cells was less than that in the control small interfering RNA group (829 ± 142 U/l vs. 1,056 ± 152 U/l, P < 0.0001; n = 4/group; fig. 2, A and B). More importantly, both cyclic stretch–induced alveolar epithelial cell apoptotic percentage and caspase 3 activity were significantly reduced in the transfected cells compared with the control small interfering RNA transfected cells (27 ± 5% fold vs. 53 ± 8% fold; 2.3 ± 0.2 fold of control vs. 4.6 ± 0.7 fold of control; P = 0.005, P < 0.0001; n = 4/group; fig. 2, C–E).

Fig. 2.

Cyclic stretch–induced apoptosis attenuated after knockdown of death-associated protein kinase 1 (DAPK1) in mouse alveolar epithelial cells. DAPK1 expression in mouse alveolar epithelial cells was silenced by small interfering RNA. After 12 h, cells received cyclic stretch at 30% elongation for 24 h. (A) A change in cell morphology was observed under an optical microscope. (i) Control group. (ii) Control small interfering RNA + cyclic stretch group. (iii) DAPK1 small interfering RNA + cyclic stretch group. (B) Lactic acid dehydrogenase concentration was measured in the media of the corresponding cultures. (C, D) The percentage of cell apoptosis was examined by flow cytometry. (E) Caspase 3 activity was detected. (F, G) Western blot was used to analysis levels of B-cell lymphoma 2 (Bcl-2) and Bcl-2–associated X (Bax). The scatter graphs are from four independent experiments. #P < 0.05 compared with control group; *P < 0.05 compared with control small interfering RNA + cyclic stretch group.

Fig. 2.

Cyclic stretch–induced apoptosis attenuated after knockdown of death-associated protein kinase 1 (DAPK1) in mouse alveolar epithelial cells. DAPK1 expression in mouse alveolar epithelial cells was silenced by small interfering RNA. After 12 h, cells received cyclic stretch at 30% elongation for 24 h. (A) A change in cell morphology was observed under an optical microscope. (i) Control group. (ii) Control small interfering RNA + cyclic stretch group. (iii) DAPK1 small interfering RNA + cyclic stretch group. (B) Lactic acid dehydrogenase concentration was measured in the media of the corresponding cultures. (C, D) The percentage of cell apoptosis was examined by flow cytometry. (E) Caspase 3 activity was detected. (F, G) Western blot was used to analysis levels of B-cell lymphoma 2 (Bcl-2) and Bcl-2–associated X (Bax). The scatter graphs are from four independent experiments. #P < 0.05 compared with control group; *P < 0.05 compared with control small interfering RNA + cyclic stretch group.

Close modal

To explore the downstream pathways of DAPK1, we also examined the intrinsic mitochondrial pathway molecules. The results suggested associations between DAPK1 knockdown and enhanced B-cell lymphoma 2 expression (1.6 ± 0.2 fold vs. 1.1 ± 0.1 fold; P = 0.004; n = 4/group; fig. 2F) and between DAPK1 knockdown and repressed B-cell lymphoma 2–associated X expression (5.4 ± 0.3 fold vs. 6.1 ± 0.4 fold; P = 0.003; n = 4/group; fig. 2G). Thus, the underlying mechanism of cyclic stretch–induced alveolar epithelial cell apoptosis may involve intrinsic mitochondrial pathways.

Effect of DAPK1 Overexpression on Alveolar Epithelial Cell Apoptosis without Mechanical Stretch

To confirm that DAPK1 participated in cyclic stretch–induced alveolar epithelial cell apoptosis, the influence of DAPK1 overexpression on alveolar epithelial cell apoptosis was examined. It was noted that DAPK1Δcalmodulin plasmid transfection was associated with significantly higher DAPK1 expression and activation compared with the control plasmid group (Supplemental Digital Content, fig. S4, https://links.lww.com/ALN/C447), as well as cell floating and shrinkage (fig. 3A) and higher lactic acid dehydrogenase concentration (852 ± 99 U/l vs. 510 ± 71 U/l; P = 0.003; n = 4/group; fig. 3B). DAPK1 overexpression was also associated with a higher percentage of apoptotic cells (49 ± 6% fold vs. 14 ± 3% fold; P < 0.0001; n = 4/group; fig. 3, C and D), increased caspase 3 activity (4.8 ± 0.3 fold of control vs. 1.3 ± 0.2 fold of control; P < 0.0001; n = 4/group; fig. 3E), lower B-cell lymphoma 2 expression (1.1 ± 0.2 fold vs. 2.4 ± 0.5 fold; P = 0.004; n = 4/group; fig. 3F), and higher B-cell lymphoma 2–associated X expression (4.4 ± 0.1 fold vs. 2.8 ± 0.3 fold; P < 0.0001; n = 4/group; fig. 3G), relative to the control plasmid group. These data suggest that DAPK1 overexpression is capable of inducing alveolar epithelial cell apoptosis and activating intrinsic mitochondrial pathways.

Fig. 3.

Overexpression of death-associated protein kinase 1 (DAPK1) was associated with an increase in mouse alveolar epithelial cells apoptosis. Mouse alveolar epithelial cells were transfected with DAPK1Δcalmodulin (DAPK1ΔCaM) plasmid to increase active DAPK1. (A) A change in cell morphology was observed under an optical microscope. (B) Lactic acid dehydrogenase concentration was measured in the media of the corresponding cultures. (C, D) The percentage of cell apoptosis was examined by flow cytometry. (E) Caspase 3 activity was detected. (F, G) Western blot was used to analysis levels of B-cell lymphoma 2 (Bcl-2) and Bcl-2–associated X (Bax). The scatter graphs are from four independent experiments. #P < 0.05 compared with control group; *P < 0.05 compared with control plasmid group.

Fig. 3.

Overexpression of death-associated protein kinase 1 (DAPK1) was associated with an increase in mouse alveolar epithelial cells apoptosis. Mouse alveolar epithelial cells were transfected with DAPK1Δcalmodulin (DAPK1ΔCaM) plasmid to increase active DAPK1. (A) A change in cell morphology was observed under an optical microscope. (B) Lactic acid dehydrogenase concentration was measured in the media of the corresponding cultures. (C, D) The percentage of cell apoptosis was examined by flow cytometry. (E) Caspase 3 activity was detected. (F, G) Western blot was used to analysis levels of B-cell lymphoma 2 (Bcl-2) and Bcl-2–associated X (Bax). The scatter graphs are from four independent experiments. #P < 0.05 compared with control group; *P < 0.05 compared with control plasmid group.

Close modal

Effect of High VT Ventilation on Signs of Ventilator-induced Lung Injury, Alveolar Epithelial Cell Apoptosis, and DAPK1 Activation

We have confirmed the proapoptotic effect of DAPK1 in alveolar epithelial cells that received cyclic stretch in vitro. To determine the related effect of DAPK1 in ventilator-induced lung injury, the effect of different tidal ventilation on ventilator-induced lung injury, alveolar epithelial cell apoptosis, and DAPK1 activation were investigated in vivo (fig. 4). Compared with the lung tissues of the low VT group, those in the high VT group showed greater histologic changes, including damage of the alveolar structure with septal thickening and hemorrhage and significantly higher lung injury score (0.50 ± 0.05 score/field vs. 0.27 ± 0.13 score/field; P = 0.005; n = 6/group; fig. 4, A and B). The high VT also had significant increases in the total number of cells ([8.3 ± 0.7] × 104 cells vs. [4.6 ± 0.7] × 104 cells; P = 0.002; n = 6/group; fig. 4C), protein concentration in bronchoalveolar lavage (657 ± 58 µg/ml vs. 516 ± 38 µg/ml; P = 0.006; n = 6/group; fig. 4D), wet-to-dry lung ratio (5.7 ± 0.9 fold vs. 4.1 ± 0.2 fold; P = 0.038; n = 6/group; fig. 4E), and levels of the proinflammatory cytokines tumor necrosis factor-α, interleukin-6, and interleukin-1β in bronchoalveolar lavage and lung tissues (fig. 4, F–K). Concomitantly, the terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate in situ nick-end labeling staining indicated a significantly higher percentage of apoptotic events in the lungs of the high VT group compared with the low VT group (43 ± 6% fold vs. 4 ± 2% fold; P < 0.0001; n = 6/group; fig. 4, L and M). The above evidence suggested that, in vivo, high VT ventilation induced alveolar epithelial cell apoptosis.

Fig. 4.

Ventilator-induced lung injury, death-associated protein kinase 1 (DAPK1) activation, and cell apoptosis were observed after application of different tidal volume in vivo. C57BL/6 mice were anesthetized, intubated, and either ventilated with low tidal volume (Low VT; 8 ml/kg with 120 breaths/min) or high tidal volume (High VT; 35 ml/kg, 80 breaths/min) for 4 h or not ventilated (control). (A, B) A lung injury score was based on lung histology with hematoxylin and eosin. (C–E) Lung injury was further evaluated based on cell number in bronchoalveolar lavage (C), protein concentration (D), and wet-to-dry ratio (E). (F–K) Proinflammatory cytokines in bronchoalveolar lavage and lung tissues were evaluated by enzyme-linked immunosorbent assay kits: tumor necrosis factor-α (TNF-α; F, G), interleukin-1β (IL-β; H, I), and interleukin-6 (IL-6; J, K). (L, M) Terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate in situ nick-end labeling staining was used to evaluate the percentage of alveolar epithelial cell apoptosis. DAPK1, phosphorylated DAPK1, myosin light chain (MLC), and phosphorylated MLC expression was detected by Western blot. (N) Ratio of DAPK1 to phosphorylated DAPK1. (O) Ratio of phosphorylated MLC to MLC. n = 6/group. #P < 0.05 compared with control group; *P < 0.05 compared with low tidal volume group.

Fig. 4.

Ventilator-induced lung injury, death-associated protein kinase 1 (DAPK1) activation, and cell apoptosis were observed after application of different tidal volume in vivo. C57BL/6 mice were anesthetized, intubated, and either ventilated with low tidal volume (Low VT; 8 ml/kg with 120 breaths/min) or high tidal volume (High VT; 35 ml/kg, 80 breaths/min) for 4 h or not ventilated (control). (A, B) A lung injury score was based on lung histology with hematoxylin and eosin. (C–E) Lung injury was further evaluated based on cell number in bronchoalveolar lavage (C), protein concentration (D), and wet-to-dry ratio (E). (F–K) Proinflammatory cytokines in bronchoalveolar lavage and lung tissues were evaluated by enzyme-linked immunosorbent assay kits: tumor necrosis factor-α (TNF-α; F, G), interleukin-1β (IL-β; H, I), and interleukin-6 (IL-6; J, K). (L, M) Terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate in situ nick-end labeling staining was used to evaluate the percentage of alveolar epithelial cell apoptosis. DAPK1, phosphorylated DAPK1, myosin light chain (MLC), and phosphorylated MLC expression was detected by Western blot. (N) Ratio of DAPK1 to phosphorylated DAPK1. (O) Ratio of phosphorylated MLC to MLC. n = 6/group. #P < 0.05 compared with control group; *P < 0.05 compared with low tidal volume group.

Close modal

Moreover, the ratios of DAPK1/phosphorylated DAPK1 (1.8 ± 0.3 fold vs. 0.5 ± 0.2 fold; P = 0.005; n = 6/group; fig. 4N) and phosphorylated myosin light chain/myosin light chain (1.7 ± 0.4 fold vs. 0.6 ± 0.1 fold; P = 0.002; n = 6/group; fig. 4O) were significantly higher in the lung tissues of the high VT group compared with the low VT group. All of the above indicated that ventilator-induced lung injury was induced by high VT ventilation and associated with alveolar epithelial cell apoptosis and DAPK1 activation.

Effect of DAPK1 Knockdown on Alveolar Epithelial Cell Apoptosis and Development of Ventilator-induced Lung Injury

To investigate whether DAPK1 knockdown in vivo had the same effect as that in vitro, we knocked down DAPK1 in lungs and evaluated alveolar epithelial cell apoptosis and ventilator-induced lung injury. The lung injury score and the alveolar epithelial cell apoptotic percentage of the DAPK1–adeno-associated virus–infected mice were comparable with that of the control-virus group (Supplemental Digital Content, fig. S5, https://links.lww.com/ALN/C448), but the high VT–induced DAPK1 level and activation were lower (Supplemental Digital Content, fig. S6, https://links.lww.com/ALN/C449). More importantly, compared with the control-virus group, DAPK1–adeno-associated virus infection was associated with attenuation in high VT–induced destruction of lung tissue (lung injury score, 0.40 ± 0.06 score/field vs. 0.54 ± 0.05 score/field; P < 0.0001; n = 6/group; fig. 5, A and B), cell infiltration ([5.3 ± 0.7] × 104 cells vs. [7.5 ± 0.8] × 104 cells; P < 0.0001; n = 6/group; fig. 5C), protein exudation in bronchoalveolar lavage (582 ± 50 µg/ml vs. 713 ± 35 µg/ml; P < 0.001; n = 6/group; fig. 5D), and lung edema (wet to dry ratio, 4.4 ± 0.4 fold vs. 5.1 ± 0.2 fold; P = 0.001; n = 6/group; fig. 5E). At the same time, DAPK1–adeno-associated virus–pretreated mice showed lower levels of the proinflammatory cytokines tumor necrosis factor-α, interleukin-6, and interleukin-1β in bronchoalveolar lavage and lung tissues, compared with the mice in the control-virus group (fig. 5, F–K), whereas terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate in situ nick-end labeling staining indicated smaller alveolar epithelial cell apoptotic percentage induced by high VT (35 ± 5% fold vs. 47 ± 4% fold; P < 0.0001; n = 6/group; fig. 5, L and M).

Fig. 5.

Ventilator-induced lung injury and alveolar epithelial cell apoptosis in lungs after death-associated protein kinase 1 (DAPK1) knockdown. C57BL/6 mice were inoculated through trachea with 106 genomic copies of DAPK1 small hairpin RNA vector (DAPK1-virus) or control vector (control-virus) in a final volume of 30 μl. Control mice received phosphate-buffered saline through trachea only. After 30 days, high tidal volume (VT) ventilation (35 ml/kg, 80 breaths/min, 4 h) was applied to mice in the control-virus + High VT group and DAPK1-virus + High VT group. Control mice received no treatment. (A, B) Lung injury score was based on lung histology with hematoxylin and eosin. (C–E) Lung injury was further evaluated based on cell number in bronchoalveolar lavage (C), protein concentration (D), and wet-to-dry ratio (E). (F–K) Proinflammatory cytokines in bronchoalveolar lavage and lung tissues were evaluated by enzyme-linked immunosorbent assay kits: tumor necrosis factor-α (TNF-α; F, G), interleukin-1β (IL-β; H, I), and interleukin-6 (IL-6; J, K). (L, M) terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate in situ nick-end labeling staining was used to evaluate the percentage of alveolar epithelial cell apoptosis. n = 6/group. #P < 0.05 compared with control group; *P < 0.05 compared with control-virus + High VT group.

Fig. 5.

Ventilator-induced lung injury and alveolar epithelial cell apoptosis in lungs after death-associated protein kinase 1 (DAPK1) knockdown. C57BL/6 mice were inoculated through trachea with 106 genomic copies of DAPK1 small hairpin RNA vector (DAPK1-virus) or control vector (control-virus) in a final volume of 30 μl. Control mice received phosphate-buffered saline through trachea only. After 30 days, high tidal volume (VT) ventilation (35 ml/kg, 80 breaths/min, 4 h) was applied to mice in the control-virus + High VT group and DAPK1-virus + High VT group. Control mice received no treatment. (A, B) Lung injury score was based on lung histology with hematoxylin and eosin. (C–E) Lung injury was further evaluated based on cell number in bronchoalveolar lavage (C), protein concentration (D), and wet-to-dry ratio (E). (F–K) Proinflammatory cytokines in bronchoalveolar lavage and lung tissues were evaluated by enzyme-linked immunosorbent assay kits: tumor necrosis factor-α (TNF-α; F, G), interleukin-1β (IL-β; H, I), and interleukin-6 (IL-6; J, K). (L, M) terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate in situ nick-end labeling staining was used to evaluate the percentage of alveolar epithelial cell apoptosis. n = 6/group. #P < 0.05 compared with control group; *P < 0.05 compared with control-virus + High VT group.

Close modal

Effect of DAPK1 Inhibitor on Alveolar Epithelial Cell Apoptosis and Development of Ventilator-induced Lung Injury

Because the activation of DAPK1 included an increase of DAPK1 expression and an enhanced activity, we used DAPK1 inhibitor to suppress DAPK1 activity in vivo to verify the detrimental effects of DAPK1 in ventilator-induced lung injury. The influence of DAPK1 inhibition on high VT–induced ventilator-induced lung injury was investigated (fig. 6). In the DAPK1 inhibitor + high VT group, the DAPK1 level and activation were less than that of the control group given dimethyl sulfoxide (Supplemental Digital Content, fig. S7, https://links.lww.com/ALN/C440). In DAPK1 inhibitor–pretreated mice with no or low VT ventilation, the lung injury score, DAPK1, and alveolar epithelial cell apoptotic percentage were comparable (Supplemental Digital Content, fig. S8, https://links.lww.com/ALN/C451).

Fig. 6.

Ventilator-induced lung injury and alveolar epithelial cell apoptosis in lungs after the application of death-associated protein kinase 1 (DAPK1) inhibitor. C57BL/6 mice received DAPK1 inhibitor (500 μg · kg−1 · day−1, intraperitoneally) or identical dosage of placebo-control solution (dimethyl sulfoxide) for 6 weeks. The mice in the control group were injected with phosphate-buffered saline only. After 6 weeks, the mice were ventilated at 35 ml/kg with 80 breaths/min for 4 h. (A, B) Control mice received no ventilation. Lung injury score was based on lung histology with hematoxylin and eosin. (C–E) Lung injury was further evaluated based on cell number in bronchoalveolar lavage (C), protein concentration (D), and wet-to-dry ratio (E). (F–K) Proinflammatory cytokines in bronchoalveolar lavage and lung tissues were evaluated by enzyme-linked immunosorbent assay kits: tumor necrosis factor-α (TNF-α; F, G), interleukin-1β (IL-β; H, I), and interleukin-6 (IL-6; J, K). (L, M) Terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate in situ nick-end labeling staining was used to evaluate the percentage of alveolar epithelial cell apoptosis. n = 6/group. #P < 0.05 compared with control group; *P < 0.05 compared with dimethyl sulfoxide + high tidal volume (VT) group.

Fig. 6.

Ventilator-induced lung injury and alveolar epithelial cell apoptosis in lungs after the application of death-associated protein kinase 1 (DAPK1) inhibitor. C57BL/6 mice received DAPK1 inhibitor (500 μg · kg−1 · day−1, intraperitoneally) or identical dosage of placebo-control solution (dimethyl sulfoxide) for 6 weeks. The mice in the control group were injected with phosphate-buffered saline only. After 6 weeks, the mice were ventilated at 35 ml/kg with 80 breaths/min for 4 h. (A, B) Control mice received no ventilation. Lung injury score was based on lung histology with hematoxylin and eosin. (C–E) Lung injury was further evaluated based on cell number in bronchoalveolar lavage (C), protein concentration (D), and wet-to-dry ratio (E). (F–K) Proinflammatory cytokines in bronchoalveolar lavage and lung tissues were evaluated by enzyme-linked immunosorbent assay kits: tumor necrosis factor-α (TNF-α; F, G), interleukin-1β (IL-β; H, I), and interleukin-6 (IL-6; J, K). (L, M) Terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate in situ nick-end labeling staining was used to evaluate the percentage of alveolar epithelial cell apoptosis. n = 6/group. #P < 0.05 compared with control group; *P < 0.05 compared with dimethyl sulfoxide + high tidal volume (VT) group.

Close modal

After high VT ventilation, DAPK1 inhibitor–pretreated mice showed less lung damage (lung injury score, 0.46 ± 0.03 score/field vs. 0.60 ± 0.04 score/field; P < 0.0001; n = 6/group; fig. 6, A and B), less cell infiltration ((5.3 ± 0.7) × 104 cells vs. (7.1 ± 1.5) × 104 cells; P = 0.003; n = 6/group; fig. 6C), and less protein exudation in bronchoalveolar lavage (579 ± 48 μg/ml vs. 706 ± 35 μg/ml; P = 0.002; n = 6/group; fig. 6D) and alleviated lung edema (wet to dry ratio, 5.0 ± 0.3 fold vs. 5.9 ± 0.4 fold; P = 0.004; n = 6/group; fig. 6E), compared with that of the control group given dimethyl sulfoxide. Meanwhile, DAPK1 inhibitor pretreatment was also associated with lower levels of the proinflammatory cytokines tumor necrosis factor-α, interleukin-6, and interleukin-1β in bronchoalveolar lavage and lung tissues (fig. 6, F–K), and terminal deoxynucleotidyl transferase–mediated deoxyuridine triphosphate in situ nick-end labeling staining showed a lower high VT–induced apoptotic percentage (48 ± 6% fold vs. 34 ± 4% fold; P < 0.0001; n = 6/group; fig. 6, L and M).

The present study investigated the following three hypotheses: (1) DAPK1 activation is elevated in response to cyclic stretch–induced alveolar epithelial cell apoptosis in vitro, (2) cyclic stretch–induced alveolar epithelial cell apoptosis is enhanced by DAPK1, and (3) DAPK1-induced alveolar epithelial cell apoptosis correlates with the development of ventilator-induced lung injury in vivo.

Alveolar epithelial cell apoptosis is associated with epithelial dysfunction during acute respiratory distress syndrome and ventilator-induced lung injury. A previous study reported that a stretching amplitude increasing surface area by 30% would deform the alveolar basement membrane as much as total lung inflation could, which was previously reported to be associated with ventilator-induced lung injury.21  Therefore, this amplitude was chosen for the current study. The actual stretch experienced during mechanical ventilation of the injured and inhomogeneous lungs is very likely identical to or even more severe than the chosen stretching amplitude, even when ventilation is conducted under a protective protocol.22  Considering apoptosis secondary to overdistension cannot be completely avoided even with minimal standard ventilation; reducing stretch-induced apoptosis can be a potential strategy to prevent ventilator-induced lung injury.

Among the numerous cell death molecules, DAPK1 is crucial for the induction of apoptosis via several cell death signals, including those generated by death receptors, cytokines, matrix detachment, etc. In the current study, gene knockdown and overexpression technology were used to evaluate the relationship between alveolar epithelial cell apoptosis and DAPK1 activation. The findings suggested DAPK1 as a positive controller of cyclic stretch–induced alveolar epithelial cell apoptosis and a transducer delivering mechanical stress.

It has been reported that mechanical stress leads to lower cellular levels of two anti-apoptotic proteins, B-cell lymphoma 2 and B-cell lymphoma–associated X,4  which stabilize mitochondria. Cyclic stretch–induced alveolar epithelial cell apoptosis models have revealed alterations in the intrinsic apoptosis pathway, specifically in the following components: caspase 9, cytochrome c, mitochondrial membrane potential, and reactive oxygen species.23  In stretch-induced apoptosis models, it was reported that the extrinsic death pathway is activated via the factor-associated suicide ligand/ factor-associated suicide system.24  In the present study, intrinsic mitochondrial pathway components responded to both DAPK1 suppression and activation, especially the B-cell lymphoma 2 family proteins: B-cell lymphoma 2 and B-cell lymphoma 2–associated X. The results suggested that DAPK1 promoted cyclic stretch–induced cell apoptosis via the intrinsic apoptosis pathway and eventually activated caspase 3 leading to cell death. Moreover, a prominent feature of DAPK-induced cell death is the effect on the cytoskeleton, including a loss of matrix attachment.25  Activation of DAPK1 promotes apoptosis by uncoupling stress fibers and focal adhesions and by breaking the balance between contractile and adhesion forces.26  This may also explain the correlation between cyclic stretch–mediated alveolar epithelial cell apoptosis and DAPK1 observed in the present study. Additional research is needed to further clarify this.

In vivo, adverse pulmonary permeability usually results from the damage to alveolar epithelium, in which alveoli encounter abnormal edema and high concentrations of extravasated macromolecules.27  Mechanical stretch is one of factors damaging the alveolar–epithelial barrier. It has been well accepted that dysfunction of the alveolar epithelial barrier caused by mechanical stretch can induce alveolar flooding and the infiltration of inflammatory cells in ventilator-induced lung injury.28  Although it is widely accepted that inflammation is a direct cause of lung injury during mechanical ventilation, alveolar epithelial cell apoptosis also adversely affects the epithelial barrier. In addition, excessive apoptosis will exacerbate the inflammatory response and worsen lung injury.29 

In the present study, mice were subjected to severely injurious ventilation by applying a high VT of 35 ml/kg for 4 h, whereas the in vitro stretching time did not correspond with that. This is because mechanical ventilation in vivo may adversely affect many physiologic processes,30  such as pulmonary immune responses, or induce inflammation.31  In addition, lung injury to an animal must be investigated within a feasible time, and therefore the duration of mechanical ventilation in vivo differs from the stretching time needed in vitro.

In the current study, the experiments with different tidal volume ventilation showed that injurious mechanical ventilation could damage alveolar epithelial barrier, and trigger inflammation and apoptosis in the lungs. A significant lung stress or strain in vivo is required for DAPK1 activation.

This study suggests that DAPK1 promoted ventilator-induced lung injury in vivo by inducing alveolar epithelial cell apoptosis. First, after the DAPK1 knockdown in the lungs, the integrity of histologic structures and the reduction in lung injury score indicated a protective effect during the DAPK1 deficiency. In addition, there was lower permeability, less high VT–induced pulmonary inflammation, and reduced high VT–induced alveolar epithelial cell apoptotic percentage in lungs deprived of DAPK1. Because DAPK1 activation was measured by two indicators: DAPK1 level and phosphorylated myosin light chain,12  DAPK1 inhibitor was applied to inhibit DAPK1 activation to strengthen the results obtained by the adeno-associated virus infection. To summarize, this study suggests that DAPK1 contributes to the in vivo development of ventilator-induced lung injury through its proapoptotic effect, whereas inhibition of DAPK1 maintains epithelial permeability and attenuates inflammation through the reduction of alveolar epithelial cell apoptotic percentage. However, we cannot attribute all the harmful effects of DAPK1 in ventilator-induced lung injury to the promotion of alveolar epithelial cell apoptosis in vivo. DAPK influences many pathways, including inflammation, cell skeleton remodeling, autophagy, etc.25,32,33  More studies are still needed to further characterize DAPK1 in ventilator-induced lung injury.

Study Limitations

This study has some limitations. First, the study focus was apoptosis, whereas the role of inflammation was not investigated, especially in vivo. Inflammation is a vital feature in the development of ventilator-induced lung injury, and DAPK1 also has an important function in inflammation. Second, although the in vivo studies indicated a significant increase in alveolar epithelial cell apoptosis percentage after high VT, alveolar epithelial cell necrosis was not studied. Necrosis constitutes irreversible changes that occur after cell death and contribute to tissue damage.34  More studies remain to be done to confirm whether apoptosis or necrosis is predominant in ventilator-induced lung injury. Third, the mechanism of ventilator-induced lung injury is difficult to investigate in a clinical study. More approaches are needed to testify our idea in a clinical setting. Finally, only male mice were used in the in vivo study, because the female sex hormones may affect the immune response.35  Therefore, there may be some bias due to missing data for female mice.

Conclusions

This study suggests that DAPK1 participates in cyclic stretch–induced alveolar epithelial cell apoptosis through intrinsic apoptotic pathways and promotes ventilator-induced lung injury by inducing alveolar epithelial cell apoptosis in the lungs. Inhibition of DAPK1 expression mitigated cyclic stretch–induced alveolar epithelial cell apoptosis and ventilator-induced lung injury.

Acknowledgments

The authors thank Lu Youming, Ph.D., Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei province, China, for the gift of death-associated protein kinase 1 Δcalmoduli plasmid, and Yuwen Li, M.D., Ph.D., Hayward Genetics Center, Tulane University School of Medicine, New Orleans, Louisiana, for the modification of the manuscript.

Research Support

Supported by grant Nos. 81372036 and 81601669 from the National Natural Science Foundation of China and grant No. WJ2017Q020 from the Young Scientists Funds of the Health Department in Hubei Province.

Competing Interests

The authors declare no competing interests.

1.
Henderson
WR
,
Chen
L
,
Amato
MBP
,
Brochard
LJ
.
Fifty years of research in ARDS: Respiratory mechanics in acute respiratory distress syndrome.
Am J Respir Crit Care Med
.
2017
;
196
:
822
33
2.
Beitler
JR
,
Malhotra
A
,
Thompson
BT
.
Ventilator-induced lung injury.
Clin Chest Med
.
2016
;
37
:
633
46
3.
Gattinoni
L
,
Marini
JJ
,
Pesenti
A
,
Quintel
M
,
Mancebo
J
,
Brochard
L
.
The “baby lung” became an adult.
Intensive Care Med
.
2016
;
42
:
663
73
4.
De Paepe
ME
,
Johnson
BD
,
Papadakis
K
,
Luks
FI
.
Lung growth response after tracheal occlusion in fetal rabbits is gestational age-dependent.
Am J Respir Cell Mol Biol
.
1999
;
21
:
65
76
5.
Hobi
N
,
Ravasio
A
,
Haller
T
.
Interfacial stress affects rat alveolar type II cell signaling and gene expression.
Am J Physiol Lung Cell Mol Physiol
.
2012
;
303
:
L117
29
6.
Hammerschmidt
S
,
Kuhn
H
,
Grasenack
T
,
Gessner
C
,
Wirtz
H
.
Apoptosis and necrosis induced by cyclic mechanical stretching in alveolar type II cells.
Am J Respir Cell Mol Biol
.
2004
;
30
:
396
402
7.
Tu
W
,
Xu
X
,
Peng
L
,
Zhong
X
,
Zhang
W
,
Soundarapandian
MM
,
Balel
C
,
Wang
M
,
Jia
N
,
Zhang
W
,
Lew
F
,
Chan
SL
,
Chen
Y
,
Lu
Y
.
DAPK1 interaction with NMDA receptor NR2B subunits mediates brain damage in stroke.
Cell
.
2010
;
140
:
222
34
8.
Zhao
J
,
Zhao
D
,
Poage
GM
,
Mazumdar
A
,
Zhang
Y
,
Hill
JL
,
Hartman
ZC
,
Savage
MI
,
Mills
GB
,
Brown
PH
.
Death-associated protein kinase 1 promotes growth of p53-mutant cancers.
J Clin Invest
.
2015
;
125
:
2707
20
9.
Deiss
LP
,
Feinstein
E
,
Berissi
H
,
Cohen
O
,
Kimchi
A
.
Identification of a novel serine/threonine kinase and a novel 15-kD protein as potential mediators of the gamma interferon-induced cell death.
Genes Dev
.
1995
;
9
:
15
30
10.
Aberg
M
,
Johnell
M
,
Wickström
M
,
Siegbahn
A
.
Tissue factor FVIIa prevents the extrinsic pathway of apoptosis by regulation of the tumor suppressor death-associated protein kinase 1 (DAPK1).
Thromb Res
.
2011
;
127
:
141
8
11.
Tian
X
,
Shi
Y
,
Liu
N
,
Yan
Y
,
Li
T
,
Hua
P
,
Liu
B
.
Upregulation of DAPK contributes to homocysteine-induced endothelial apoptosis via the modulation of Bcl2/Bax and activation of caspase 3.
Mol Med Rep
.
2016
;
14
:
4173
9
12.
Yamamoto
M
,
Hioki
T
,
Ishii
T
,
Nakajima-Iijima
S
,
Uchino
S
.
DAP kinase activity is critical for C(2)-ceramide-induced apoptosis in PC12 cells.
Eur J Biochem
.
2002
;
269
:
139
47
13.
Shang
T
,
Joseph
J
,
Hillard
CJ
,
Kalyanaraman
B
.
Death-associated protein kinase as a sensor of mitochondrial membrane potential: Role of lysosome in mitochondrial toxin-induced cell death.
J Biol Chem
.
2005
;
280
:
34644
53
14.
Rennier
K
,
Ji
JY
.
Shear stress regulates expression of death-associated protein kinase in suppressing TNFα-induced endothelial apoptosis.
J Cell Physiol
.
2012
;
227
:
2398
411
15.
Bialik
S
,
Kimchi
A
.
The death-associated protein kinases: Structure, function, and beyond.
Annu Rev Biochem
.
2006
;
75
:
189
210
16.
Temmerman
K
,
Simon
B
,
Wilmanns
M
.
Structural and functional diversity in the activity and regulation of DAPK-related protein kinases.
FEBS J
.
2013
;
280
:
5533
50
17.
Cohen
O
,
Feinstein
E
,
Kimchi
A
.
DAP-kinase is a Ca2+/calmodulin-dependent, cytoskeletal-associated protein kinase, with cell death-inducing functions that depend on its catalytic activity.
EMBO J
.
1997
;
16
:
998
1008
18.
Matute-Bello
G
,
Downey
G
,
Moore
BB
,
Groshong
SD
,
Matthay
MA
,
Slutsky
AS
,
Kuebler
WM
;
Acute Lung Injury in Animals Study Group
.
An official American Thoracic Society workshop report: Features and measurements of experimental acute lung injury in animals.
Am J Respir Cell Mol Biol
.
2011
;
44
:
725
38
19.
Yaxin
W
,
Shanglong
Y
,
Huaqing
S
,
Hong
L
,
Shiying
Y
,
Xiangdong
C
,
Ruidong
L
,
Xiaoying
W
,
Lina
G
,
Yan
W
.
Resolvin D1 attenuates lipopolysaccharide induced acute lung injury through CXCL-12/CXCR4 pathway.
J Surg Res
.
2014
;
188
:
213
21
20.
Li
H
,
Wu
Z
,
Feng
D
,
Gong
J
,
Yao
C
,
Wang
Y
,
Yuan
S
,
Yao
S
,
Shang
Y
.
BML-111, a lipoxin receptor agonist, attenuates ventilator-induced lung injury in rats.
Shock
.
2014
;
41
:
311
6
21.
Chandel
NS
,
Sznajder
JI
.
Stretching the lung and programmed cell death.
Am J Physiol Lung Cell Mol Physiol
.
2000
;
279
:
L1003
4
22.
Hammerschmidt
S
,
Kuhn
H
,
Gessner
C
,
Seyfarth
HJ
,
Wirtz
H
.
Stretch-induced alveolar type II cell apoptosis: Role of endogenous bradykinin and PI3K-Akt signaling.
Am J Respir Cell Mol Biol
.
2007
;
37
:
699
705
23.
Kuhn
H
,
Nieuwenhuijsen
H
,
Karthe
B
,
Wirtz
H
.
Stretch-induced apoptosis in rat alveolar epithelial cells is mediated by the intrinsic mitochondrial pathway.
Exp Lung Res
.
2017
;
43
:
49
56
24.
Kroon
AA
,
Delriccio
V
,
Tseu
I
,
Kavanagh
BP
,
Post
M
.
Mechanical ventilation-induced apoptosis in newborn rat lung is mediated via FasL/Fas pathway.
Am J Physiol Lung Cell Mol Physiol
.
2013
;
305
:
L795
804
25.
Ivanovska
J
,
Mahadevan
V
,
Schneider-Stock
R
.
DAPK and cytoskeleton-associated functions.
Apoptosis
.
2014
;
19
:
329
38
26.
Kuo
JC
,
Lin
JR
,
Staddon
JM
,
Hosoya
H
,
Chen
RH
.
Uncoordinated regulation of stress fibers and focal adhesions by DAP kinase.
J Cell Sci
.
2003
;
116
:
4777
90
27.
Herrero
R
,
Sanchez
G
,
Lorente
JA
.
New insights into the mechanisms of pulmonary edema in acute lung injury.
Ann Transl Med
.
2018
;
6
:
32
28.
Le
A
,
Damico
R
,
Damarla
M
,
Boueiz
A
,
Pae
HH
,
Skirball
J
,
Hasan
E
,
Peng
X
,
Chesley
A
,
Crow
MT
,
Reddy
SP
,
Tuder
RM
,
Hassoun
PM
.
Alveolar cell apoptosis is dependent on p38 MAP kinase-mediated activation of xanthine oxidoreductase in ventilator-induced lung injury.
J Appl Physiol (1985)
.
2008
;
105
:
1282
90
29.
Schmidt
EP
,
Tuder
RM
.
Role of apoptosis in amplifying inflammatory responses in lung diseases.
J Cell Death
.
2010
;
3
:
41
53
30.
Ngiam
N
,
Kavanagh
BP
.
Ventilator-induced lung injury: The role of gene activation.
Curr Opin Crit Care
.
2012
;
18
:
16
22
31.
Kuipers
MT
,
Aslami
H
,
Janczy
JR
,
van der Sluijs
KF
,
Vlaar
AP
,
Wolthuis
EK
,
Choi
G
,
Roelofs
JJ
,
Flavell
RA
,
Sutterwala
FS
,
Bresser
P
,
Leemans
JC
,
van der Poll
T
,
Schultz
MJ
,
Wieland
CW
.
Ventilator-induced lung injury is mediated by the NLRP3 inflammasome.
Anesthesiology
.
2012
;
116
:
1104
15
32.
Lopes
F
,
Keita
ÅV
,
Saxena
A
,
Reyes
JL
,
Mancini
NL
,
Al Rajabi
A
,
Wang
A
,
Baggio
CH
,
Dicay
M
,
van Dalen
R
,
Ahn
Y
,
Carneiro
MBH
,
Peters
NC
,
Rho
JM
,
MacNaughton
WK
,
Girardin
SE
,
Jijon
H
,
Philpott
DJ
,
Söderholm
JD
,
McKay
DM
.
ER-stress mobilization of death-associated protein kinase-1–dependent xenophagy counteracts mitochondria stress–induced epithelial barrier dysfunction.
J Biol Chem
.
2018
;
293
:
3073
87
33.
Lai
MZ
,
Chen
RH
.
Regulation of inflammation by DAPK.
Apoptosis
.
2014
;
19
:
357
63
34.
Tang
PS
,
Mura
M
,
Seth
R
,
Liu
M
.
Acute lung injury and cell death: How many ways can cells die?
Am J Physiol Lung Cell Mol Physiol
.
2008
;
294
:
L632
41
35.
Klein
SL
,
Flanagan
KL
.
Sex differences in immune responses.
Nat Rev Immunol
.
2016
;
16
:
626
38