Skip to main content
Erschienen in: Critical Care 1/2023

Open Access 01.12.2023 | Perspective

A long-lasting porcine model of ARDS caused by pneumonia and ventilator-induced lung injury

verfasst von: Enric Barbeta, Marta Arrieta, Ana Motos, Joaquim Bobi, Hua Yang, Minlan Yang, Giacomo Tanzella, Pierluigi Di Ginnatale, Stefano Nogas, Carmen Rosa Vargas, Roberto Cabrera, Denise Battaglini, Andrea Meli, Kasra Kiarostami, Nil Vázquez, Laia Fernández-Barat, Montserrat Rigol, Ricard Mellado-Artigas, Gerard Frigola, Marta Camprubí-Rimblas, Pau Ferrer, Daniel Martinez, Antonio Artigas, Carlos Ferrando, Miquel Ferrer, Antoni Torres

Erschienen in: Critical Care | Ausgabe 1/2023

Abstract

Background

Animal models of acute respiratory distress syndrome (ARDS) do not completely resemble human ARDS, struggling translational research. We aimed to characterize a porcine model of ARDS induced by pneumonia—the most common risk factor in humans—and analyze the additional effect of ventilator-induced lung injury (VILI).

Methods

Bronchoscopy-guided instillation of a multidrug-resistant Pseudomonas aeruginosa strain was performed in ten healthy pigs. In six animals (pneumonia-with-VILI group), pulmonary damage was further increased by VILI applied 3 h before instillation and until ARDS was diagnosed by PaO2/FiO2 < 150 mmHg. Four animals (pneumonia-without-VILI group) were protectively ventilated 3 h before inoculum and thereafter. Gas exchange, respiratory mechanics, hemodynamics, microbiological studies and inflammatory markers were analyzed during the 96-h experiment. During necropsy, lobar samples were also analyzed.

Results

All animals from pneumonia-with-VILI group reached Berlin criteria for ARDS diagnosis until the end of experiment. The mean duration under ARDS diagnosis was 46.8 ± 7.7 h; the lowest PaO2/FiO2 was 83 ± 5.45 mmHg. The group of pigs that were not subjected to VILI did not meet ARDS criteria, even when presenting with bilateral pneumonia. Animals developing ARDS presented hemodynamic instability as well as severe hypercapnia despite high-minute ventilation. Unlike the pneumonia-without-VILI group, the ARDS animals presented lower static compliance (p = 0.011) and increased pulmonary permeability (p = 0.013). The highest burden of P. aeruginosa was found at pneumonia diagnosis in all animals, as well as a high inflammatory response shown by a release of interleukin (IL)-6 and IL-8. At histological examination, only animals comprising the pneumonia-with-VILI group presented signs consistent with diffuse alveolar damage.

Conclusions

In conclusion, we established an accurate pulmonary sepsis-induced ARDS model.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s13054-023-04512-8.
Enric Barbeta and Marta Arrieta contributed equally to this work.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ARDS
Acute respiratory distress syndrome
BAL
Bronchoalveolar lavage
CFU
Colony-forming unit
CVP
Central venous pressure
DP
Driving pressure
EVLW
Extravascular lung water
FiO2
Fraction of inspired oxygen
ICU
Intensive care unit
LV dPmax
Maximal left ventricular pressure rise
MAP
Mean arterial pressure
MDR
Multidrug resistance
OA
Oleic acid
PaO2
Partial pressure of oxygen in arterial blood
PAOP
Pulmonary artery occluded pressure
PaCO2
Partial pressure of carbon dioxide in arterial blood
PC
Pressure control
PEEP
Positive End-expiratory pressure
RR
Respiratory rate
VC
Volume control
VDI
Vasopressor dependency index
VILI
Ventilatory-induced lung injury
ZEEP
Zero end-expiratory pressure

Background

Acute respiratory distress syndrome (ARDS) is a syndrome characterized by capillary endothelial injury and diffuse alveolar damage secondary of cytokines release after a direct risk factor exposure (i.e., infection, aspiration, major trauma) or an indirect risk factors (i.e., sepsis, blood transfusion, major surgery) [1].
The latest definition (i.e., Berlin definition) [1] describes an acute clinical–radiological syndrome characterized by acute respiratory failure and bilateral pulmonary opacities not fully caused by heart failure, fluid overload or other pulmonary pathologies such as lung tumors or atelectasis. As a result, patients with ARDS require oxygen therapy and mechanical ventilation, which is deliverable either invasively or non-invasively.
Animal experimentation has greatly contributed to generating knowledge about ARDS. It was first done in animals with the application of mechanical ventilation and high tidal volume to promote lung damage before being shown in several randomized clinical trials [2]. The benefits of prone position were initially described in animal experimentation as well [3]. Other concepts related to ventilator-induced lung injury such as pulmonary strain, stress and mechanical power were demonstrated in animals before humans [4, 5]. Paradoxically, though, none of these insightful experiments were performed in animal models of ARDS. They were, however, done in animals without any lung injury.
Reproducing ARDS in animals is challenging. The most common cause of ARDS in humans is pneumonia [6], and its definition is based on a constellation of clinical characteristics (acute respiratory failure) and radiological signs (bilateral pulmonary edema) [1]. However, the definition of ARDS in animal models is rarely supported by a disorder of oxygenation or thoracic imaging. Indeed, most pulmonary insults used in an experimental setting differ substantially from human ARDS physiopathology (i.e., intravenous oleic acid or endotoxin administration, surfactant depletion with saline lavage, etc.) [7, 8]. Therefore, animal models usually present features inconsistent with human ARDS like short duration [7, 9] and a significant rate of reversibility [10]. All these drawbacks hamper research in animal experimentation, as it relates to therapies such as new ventilation and extracorporeal life support strategies and targeted treatments.
In this study, we aimed to characterize a double-hit porcine model of ARDS induced by the most common risk factors identified in humans [6]: pneumonia and ventilator-induced lung injury (VILI). To evaluate pneumonia as a cause of ARDS within the presence and absence of VILI, we studied two animal groups: one that comprised pigs with bilateral pneumonia with a high pulmonary strain and the other (control) with bilateral pneumonia and no significant pulmonary strain. After the Berlin criteria for ARDS were met, we monitored gas exchange, pulmonary mechanics, pulmonary and systemic hemodynamics, and pulmonary and systemic inflammatory markers. We also studied the microbiology and histology of the injured lungs.

Methods

This is an experimental animal study that aimed to characterize the respiratory, hemodynamic, inflammatory, microbiological and histological features of ARDS caused by a bilateral Pseudomonas aeruginosa pneumonia and with/without the application of injurious ventilation (a high or low pulmonary strain).
The study was carried out in 10 Large White Landrace pigs (40–48 kg), subjected to 96 h of mechanical ventilation in the prone position (SERVO-I, Maquet, NJ, USA). The experiment was divided into two time frames: (1) induction and (2) model development. During induction, pigs were challenged with a multidrug-resistant (MDR) strain of P. aeruginosa and, based on the study group, mechanically ventilated with protective or injurious settings. Model development began when ARDS diagnosis was established; it concluded once the animal was put down.
As previously described, we prepared the animal and maintained deep anesthesia throughout the experiment [11]. Figure 1 displays main assessments throughout the experiment detailed in Additional file 1.

Bilateral pneumonia with VILI

This group comprised six animals. Briefly, after animal preparation, we performed 3 h of VILI with a ventilator setting that ensured harmful pulmonary dynamic strain (> 1.5) [12]. Following this, we instilled 15 mL of 107 colony-forming unit (CFU)/mL of MDR P. aeruginosa, susceptible to both meropenem and levofloxacin, into each lobe via bronchoscopy (see Additional file 1: Table S1). Injurious mechanical ventilation resumed with the same previous setting, albeit with a positive end-expiratory pressure (PEEP) of 5 cmH2O. We diagnosed ARDS when arterial partial pressure of oxygen (PaO2)/fraction of inspired oxygen (FiO2) < 150 mmHg was achieved. Once criteria for ARDS were met, ventilator mode was changed to volume control (VC) and pigs were ventilated according to a lung protective strategy. Tidal volume was set to aim for normocapnia with a DP that did not exceed 15 cmH2O [13].

Bilateral pneumonia without VILI

The control arm included four animals which presented bilateral pneumonia and received treatment with protective ventilator parameters (no significant strain). Like the previous group, 3 h of ventilation was applied before the bacterial challenge, with a setting that assured a non-harmful pulmonary strain (< 1.5) [12]. After that, we instilled 15 mL of 107 CFU/mL of MDR P. aeruginosa into each pulmonary lobe. We continued applying protective ventilation until the end of the experiment. Apart from a DP < 15 cmH2O [13], we maintained the thresholds for the most important VILI parameters under a safe range per literature [5, 12, 13] (Table 1).
Table 1
Induction and model development periods
 
Pneumonia with VILI
Pneumonia without VILI
Injurious ventilation
P. aeruginosa inoculum
Injurious ventilation
(inoculum to pneumonia diagnosis)
Injurious ventilation
(pneumonia diagnosis to ARDS diagnosis)
ARDS (to the end of the experiment)
Protective ventilation
P. aeruginosa inoculum
Protective ventilation
(inoculum to pneumonia diagnosis)
Pneumonia
(to the end of the experiment)
ARDS
Duration (h)
3.1 ± 0.1
0.9 ± 0.1
12.0 ± 0.7
16.0 ± 2.3
46.8 ± 7.7
3.1 ± 0.2
NA
7.2 ± 1.1
82.4 ± 1.2
NA
FiO2 (%)
100.0 ± 0.0
100.0 ± 0.0
100.0 ± 0.0
100.0 ± 0.0
100.0 ± 0.0
41.3 ± 0.0
NA
40.6 ± 0.6
44.4 ± 1.1
NA
TV (mL/kg)
33.3 ± 2.4
10.5 ± 0.0
27.5 ± 1.6
25.1 ± 0.9
5.7 ± 0.2
8.1 ± 0
NA
7.8 ± 0.2
7.2 ± 0.1
NA
RR (breaths/min)
12.3 ± 0.0
20.0 ± 0.0
11.3 ± 0.9
10 ± 0.0
45.8 ± 1.0
15.4 ± 0.4
NA
16.8 ± 0.1
23.7 ± 1.1
NA
PEEP (cmH2O)
0.0 ± 0.0
3.0 ± 0.0
4.9 ± 0.0
4.9 ± 0.1
9.5 ± 0.3
4.0 ± 0.0
NA
4.3 ± 0.0
4.8 ± 0.2
NA
DP (mmHg)
27.7 ± 0.2
12.5 ± 0.0
30.1 ± 0.3
29.9 ± 0.2
17.7 ± 0.5
9.5 ± 0.5
NA
12.3 ± 0.0
13.5 ± 0.4
NA
MP
NA
10.5 ± 1.6
34.7 ± 0.8
28.35 ± 1.2
26.9 ± 1.7
5.23 ± 0.7
NA
6.65 ± 1.0
10.1 ± 0.3
NA
Ventilatory mode
PC
VC
PC
PC
VC
VC
NA
VC
VC
NA
After the inoculum, a short period of protective ventilation was performed to ease bacteria engraftment. Note that this was not necessary in the other group because protective ventilation was already ongoing. Data are reported as mean ± standard error
ARDS acute respiratory distress syndrome, DP driving pressure, FiO2 fraction of inspired oxygen, MP mechanical power, P. aeruginosa, Pseudomonas aeruginosa, PC pressure control, PEEP positive end-expiratory pressure, RR respiratory rate, TV tidal volume, VC volume control

Pneumonia diagnosis

Pneumonia was clinically diagnosed in both groups based on a decline in PaO2 and one of the following signs of infection: body temperature > 38.5 °C, a white blood count > 14·109/L and purulent secretions [14]. At pneumonia diagnosis, we started antimicrobial therapy with 25 mg/kg of meropenem every 8 h and 10 mg/kg of levofloxacin every 24 h, as dual therapy recommended by international guidelines [15].

Statistical analysis

The results were reported as the mean and standard error of the mean. Categorical variables were presented as umbers and percentage. For continuous variables, we compared groups and over time using the repeated measure two-way ANOVA test. Bonferroni’s post hoc multiple comparison was used to control the experiment-wise error rate. Comparison between groups at a fixed time point was assessed using the Mann–Whitney nonparametric test. Either the Chi-square test or Fisher’s exact test was used to compare categorical variables. A two-sided p value < 0.05 was considered statistically significant. All statistical analyses were performed using the GraphPad Prism 8 (version 8.0.1 (244), San Diego, CA, USA).

Results

Ten animals (42.9 ± 1.13 kg) were divided into two groups: six animals comprised the pneumonia-with-VILI group and four animals the pneumonia-without-VILI group. In the former group, two pigs were put down before the end of the experiment due to severe respiratory failure and severe respiratory acidosis. The mortality rate reached 33.3%. All animals in the pneumonia-without-VILI group completed the study.

Induction period in pigs with pneumonia according to the presence or absence of VILI

Table 1 and Fig. 2 display parameters related to mechanical ventilation, gas exchange and respiratory mechanics observed during the induction period. During this period, we performed a 15-mL P. aeruginosa challenge of 7.77 ± 0.11 log CFU/mL in each pulmonary lobe in both groups.
After the bacterial challenge, we continued injurious ventilation in the VILI group for 28.04 ± 3.03 h until the Berlin criteria were met, with the mean PaO2/FiO2 being 129.62 ± 6.1 mmHg at ARDS diagnosis. Bilateral opacities were identified by X-ray in all six pigs. In contrast, the pneumonia-without-VILI group never met the Berlin criteria for ARDS. PaO2/FiO2 did not decrease below 300 mmHg at any time point (mean PaO2/FiO2 was 389.90 ± 3.97 mmHg) (p < 0.0002) (Fig. 2).
No significant differences were found in the hemodynamic parameters between groups during the induction period, as shown in Fig. 3.
Pneumonia was clinically and microbiology established at 11.99 ± 0.67 and 7.25 ± 1.12 h post-inoculum in the pneumonia-with-and-without-VILI groups, respectively (p = 0.01). Additional file 1: Table S2 shows the achieved criteria of pneumonia diagnosis. No differences in P. aeruginosa burden were observed between study groups at pneumonia diagnosis, neither in tracheal secretions (p = 0.46) nor in BAL samples (p = 0.57) (Fig. 4).

Model development: respiratory function

Animals comprising the pneumonia-with-VILI group maintained a PaO2/FiO2 below 300 mmHg until the end of experiment (model development period). The mean duration of an ARDS diagnosis in animals that survived was 58.7 ± 1.3 h. For those who were killed due to clinical instability, it was 29.9 h and 16.3 h each. During this period (model development), the mean PaO2/FiO2 was 187.80 ± 28.46 mmHg, with the lowest measure recorded at 83.00 ± 5.45 mmHg. Figure 2 displays the evolution of respiratory system mechanics during model development. Respiratory system compliance was lower in animals with VILI compared with those without VILI (14.54 ± 1.30 and 21.77 ± 0.32 cmH2O/L) (p = 0.011). Furthermore, DP and mechanical power showed significant differences between groups (p = 0.011 and p = 0.001, respectively). Mean values were 17.65 ± 0.50 cmH2O and 26.87 ± 1.72 J/min in the group of pneumonia with VILI and 13.46 ± 0.34 cmH2O and 10.09 ± 0.27 J/min in the group of pneumonia without VILI. After the ARDS diagnosis time point, PaCO2 in animals with VILI was significantly higher than in those without VILI (69.28 ± 10.95 and 38.85 ± 1.076 mmHg, p = 0.027). This also was observed in exhaled minute volume (11.71 ± 0.32 and 7.02 ± 0.10 L/min, p < 0.001). Furthermore, mean EVLW was 840.2 ± 61.73 mL/kg in the pneumonia-with-VILI group and 385.6 ± 6.90 mL/kg in the pneumonia-without-VILI group (p = 0.013), as shown in Fig. 2. Ventilator setting adjustments are displayed in Additional file 1: Fig. S1.

Model development: systemic and pulmonary hemodynamics

Mean pulmonary pressure increased in pigs with ARDS: Mean values were 26.23 ± 0.73 mmHg compared with 22.79 ± 0.28 mmHg in in the pneumonia-without-VILI group (p = 0.056). Mean arterial pressure in pigs with ARDS remained at 72.48 ± 1.23 mmHg. In the other group, it was 84.79 ± 5.21 mmHg (p = 0.003). Pigs from the pneumonia-with-VILI group presented a higher vasopressor dependency index (VDI); however, it was not significant (0.63 ± 0.16 vs 0.03 ± 0.02, p = 0.158). After an ARDS diagnosis, systemic vascular resistances were lower in the pneumonia-with-VILI group than in the pneumonia-without-VILI group (907.3 ± 44.76 dynes/s/cm−5 1432.71 ± 124.41 dynes/s/cm−5; p = 0.02). Figure 3 shows other hemodynamic parameters. Laboratory findings and fluid balance are reported in Additional file 1: Table S3 and Fig. S2.

Imaging, necropsy and histology

We performed X-ray images of two different projections at baseline and the end of the experiment. While normal lung appearance was found at baseline, bilateral opacities were observed at the end of the experiment in both groups. X-ray images were also performed at ARDS diagnosis in animals with VILI, revealing bilateral opacities too. At necropsy, macroscopic findings were more severe in those with VILI than in animals presenting pneumonia without VILI, as shown in Additional file 1: Fig. S3.
Figure 5 displays the main lung findings. Macroscopically, all pigs presented with findings consistent with bilateral pneumonia. However, animals that developed ARDS also had widespread hepatization, severe edematous appearance, pleural effusion and adherence. Importantly, in the pneumonia-with-VILI group, we found higher vascular permeability, as revealed by both a higher lung/body weight ratio (2.71 ± 0.26 in the pneumonia-with-VILI group vs. 1.61 ± 0.13% in the pneumonia-without-VILI group, p = 0.009) and lung wet-to-dry ratio (7.56 ± 0.73 in the pneumonia-with-VILI group vs. 4.03 ± 1.03% in the pneumonia-without-VILI group, p = 0.029).
In the pneumonia-with-VILI group, all except one (83%) presented hyaline membrane formation consistent with diffuse alveolar damage (DAD). However, DAD and hyaline membranes were absent in all the animals in which pneumonia was induced without VILI. Furthermore, when compared with the pneumonia-without-VILI group, animals that developed ARDS also presented a rise in intra-alveolar (p = 0.033) and interstitial neutrophil infiltration (p = 0.005), as well as increased septal edema (p = 0.038). Alveolar edema, intra-alveolar fibrin and hemorrhage, proliferation of alveolar type II cells and epithelial denudation were reported in both groups without differences. In Additional file 1: Fig. S4, the same features are displayed by dependent and non-dependent areas.

Systemic and local inflammatory markers

As shown in Additional file 1: Fig. S5, systemic IL-6 and IL-8 had significant time-dependent changes (p < 0.0001 and p = 0.024). At pneumonia diagnosis, plasma IL-6 was higher in the pneumonia-with-VILI group (579.94 ± 138.79 vs. 356.79 ± 233.66 pg/mL, p = 0.038). However, after the ARDS diagnosis (model development period), these differences disappeared. IL-6 and IL-8 also presented an increase in BAL, albeit without significant differences between groups.

Microbiology assessments

Figure 5 displays the main microbiological results. Throughout the model development period, animals from the pneumonia-with-VILI group presented a lower burden of P. aeruginosa in tracheal aspirates (4.24 ± 0.68 log CFU/mL in the pneumonia-with-VILI group and 6.01 ± 0.32 log CFU/mL in the pneumonia-without-VILI group, p = 0.027). However, it was not different for lung tissue or BAL. At the end of the experiment, tracheal aspirate cultures from both groups tested > 3 log CFU/mL. In lung tissue, mean values were below 3 log CFU/mL in both groups.

Discussion

We developed a model of pulmonary sepsis-induced ARDS in pigs that had long-lasting features consistent with human ARDS. Our model was characterized by the following aspects: (I) development of severe oxygenation disorders; (II) development of hypercapnia despite the application of high-minute ventilation (reflecting an increase in dead-space ventilation); (III) decrease in respiratory system static compliance; (IV) increase in pulmonary permeability; (V) presence of cardiovascular dysfunction (sepsis-induced vasoplegia); (VI) pulmonary and systemic inflammation; and (VII) signs of diffuse alveolar damage.
The model presented fulfills recent recommendations for animal experimentation in the field of acute lung injury [16]. That considered, we demonstrated histological evidence of tissue injury; alteration of the alveolar–capillary barrier; presence of systemic and pulmonary inflammatory response; and severe respiratory dysfunction. Furthermore, other relevant characteristics specific to the model should be underscored: First and foremost, the pulmonary insult used in the induction period is the main cause of ARDS in humans (bacterial pneumonia). Moreover, the second hit used in the model (VILI) might be unavoidable in human ARDS, as ventilation with high strain and stress is needed to maintain gas exchange in the most severe patients [6]. Second, the large duration of the experiment allows for tracking changes in respiratory physiology. Third, large animals allow for advanced monitoring (i.e., electrical impedance tomography, and analysis of chest wall and pulmonary respiratory mechanics, pulmonary and systemic hemodynamics, etc.) as well as evaluation of complex therapies such as extracorporeal membrane oxygenation and mechanical ventilation.

Comparison of animal models and their characteristics

Given the clinically relevant insults used (pneumonia and VILI) as well as the physiological characteristics exhibited during the 96-h experiment period, we believe that the model presented has higher accuracy in reproducing human ARDS compared with others [7, 8, 16, 17].
In line with our study, the repeated lavage model presents remarkable gas exchange disorders and diminished compliance due to surfactant depletion. Specifically, our ARDS model shows a mean PaO2/FiO2 which, throughout the model development, ranges in moderate severity with periods of severe instability. Moreover, notably diminished and progressively deteriorating compliance is found throughout the model development period as well. Surfactant depletion is not the main mechanism of respiratory failure in adult ARDS; hence, this model fails to reproduce other clinical features of ARDS. For example, in contrast from our model, only modest epithelial injury signs are present; permeability changes, scarce; and clinical features described, reversible, lasting less than 24 h [9, 10, 18]. Our model, however, presents a histology that demonstrates that severe epithelial damage and disorders in oxygenation are maintained up to approximately 60 h. In the surfactant depletion model, some authors reported an increase in BAL cytokines, but the role of inflammation is unclear [9]. We demonstrated, nonetheless, an increase in both systemic and pulmonary inflammation and signs of sepsis-associated cardiovascular dysfunction.
Similar flaws are identified in the oleic acid (OA) model. This one mimic ARDS caused by fat embolism, an exceptional etiology of syndrome [10]. In the OA model, the pathophysiology of pulmonary injury is unknown and unlikely similar to the most common causes of ARDS such as pneumonia, the risk factor used in our model [6]. OA administration raises pulmonary arterial pressure and intrapulmonary shunt [1921] and produces a marked increase in EVLW [20]. We also showed a rise in the mean pulmonary arterial pressure and EVLW. We tracked changes in PaO2/FiO2 as surrogate of pulmonary shunt, identifying periods with a value below 100 mmHg (i.e., severe ARDS). In the OA model, an inflammatory response is shown with an increase in some inflammatory markers in lung such as IL-6 and IL-1 beta [2224]. At histological examination, however, only mild signs of lung injury are found with no hyaline membrane formation [20, 23, 25]. In our model, though, we demonstrated DAD in 83% of histological specimens of pigs with ARDS. In the OA model, the severity of lung injury is somewhat unpredictable because animals present different responses per OA doses and sudden, intense hemodynamic instability during infusion [23, 25]. Like the repeated lavage model, the oleic acid-induced lung injury is also reversible within several hours, contrasting with our 60-h ARDS period [21, 23].
Systemic administration of bacterial lipopolysaccharides (LPS) was one of the earliest approaches used to study sepsis-induced ARDS. LPS administration markedly decreases PaO2 and systemic arterial pressure. It also provides important information about host inflammatory responses in BAL and serum [7, 26]. For that reason, it is a good choice for pathophysiological studies of infection and ARDS [7, 26]. Our model follows this approach; however, we instill viable bacteria—rather than bacterial toxins—into the lungs, thus fulfilling the complete clinical picture of severe pneumonia. During the model development, a high burden of P. aeruginosa was found in BAL and tracheal aspirates. While we did not find differences in BAL samples, we did identify a lower bacterial burden in tracheal aspirates from animals with ARDS in comparison with those who did not develop it. The interpretation of this finding is not clear. We speculate that the plasma leak into the lungs of pigs with ARDS prompted tracheal aspirations more frequently and promoted bacterial clearance. Histological alterations observed in the LPS model are mainly neutrophil parenchymal infiltration and mild edema, features that resemble those seen in pneumonia. However, in this model, hyaline membranes are lacking [7, 27], suggesting milder epithelial damage and lower pulmonary permeability compared with our model. Other limitations of this model are short duration, significant differences in endotoxin response between species and variability in biological activity of endotoxin preparations [8]. In contrast, the duration of our model is enough to perform multiple analyses, given its reproducible nature as well.
The smoke/burn is a reproducible model that resembles the clinical time course of ARDS during the first 24 h. It can be used in studies up to 96 h [28]. However, smoke inhalation as etiology of ARDS is not frequent, and the pathophysiology of lung injury is different from sepsis [6]. Although some studies achieve severe respiratory failure [28], this does not last longer 24 h [29, 30]. Inflammatory markers as IL-6 are found elevated in BAL and plasma [28, 31]. Histology observations demonstrate a presence of inflammatory infiltrate, mild hemorrhage and edema, albeit no hyaline membranes [28, 31, 32].
Injurious ventilation has been used as a unique or double-hit model to reproduce ARDS in animals [8, 32]. In rodent animal models, VILI causes an impairment in respiratory mechanics [33, 34], tissue injury [33, 35] and pulmonary permeability [34], but oxygenation disorders are mild and do not usually meet ARDS criteria [3639]. These findings in small animals are difficult to translate in terms of humans. Contrarily, in larger animal models, VILI provokes severe respiratory failure. Unlike our model, though, it does not last longer than 24 h [3842]. In these models, inflammation caused by VILI was also evaluated by several methods including cytokine analysis in BAL fluid, plasma and lung tissue, and either immunohistochemistry or immunofluorescence [8, 33-44]. While some studies confirmed that VILI induces a pulmonary inflammatory response [3339], others did not [44]. These differences might be explained by either the intensity of VILI observed between studies or the primary pulmonary insult performed before aggressive ventilation was started [45]. Interpretations of our findings should consider that we used a DP of up to 30 cmH2O to ensure a clinically significant pulmonary strain and that such ventilation was performed in lungs affected by a primary inflammatory insult (pneumonia). Interestingly, as observed in our model, other authors reported the presence of hyaline membranes and diffuse alveolar damage in animals ventilated in an injurious fashion [40].

The effect of ventilator-induced lung injury in pneumonia

In our model, the group with low pulmonary strain—no VILI—presented oxygen disorders that were mild and did not meet the Berlin criteria for ARDS. On the other hand, pigs comprising the pneumonia-with-VILI group had sustained life-threatening gas exchange disorders, as well as other remarkable disturbances. In the current model, however, we did not find clinically relevant extrapulmonary organ dysfunctions other than hemodynamic failure (sepsis-induced vasoplegia). The latter was more prominent in the pneumonia-with-VILI group. Of note, while pigs with ARDS exhibited a high inflammatory response, this was not clearly influenced by the presence of VILI during model development. Importantly, high mortality (33.3%) found only in the group of pigs ventilated with the high pulmonary strain was driven by respiratory acidosis and refractory respiratory failure.
The findings described between groups merit clinical interpretation. Refractory respiratory failure—either manifested as severe hypoxemia or respiratory acidosis—represents a non-negligible cause of death and morbidity in ARDS [46, 47]. Given the extreme oxygenation and ventilation disorders found in the group of pigs with VILI, maintaining protective ventilation was challenging after the ARDS diagnosis (model development period). This is revealed by an increase in mechanical power throughout all the experiment which, consequently, might have dramatically diminished the chances of pulmonary recovery in the group with VILI. Our results, therefore, suggest that a high pulmonary strain might ease progression to ARDS in pneumonia and perhaps raise respiratory-attributable death.

Limitations

Some limitations are present in this study. First, as a result of severe respiratory failure, mortality reported in this model was high (33.3%). Second, we only studied ten non-randomized pigs and just those treated with high pulmonary strain fulfilled the diagnostic criteria for ARDS. Nevertheless, the dispersion of data was low, and our findings were consistent. Due to logistical constraints, animals were not randomized. Third, critically ill patients usually present with significant comorbidities that determine prognosis. However, animals used in the model were female, young and healthy. Moreover, the sequence of events in the clinical setting differs, as patients initially experience an insult that results in respiratory insufficiency, followed by the need for mechanical ventilation. In our model, healthy animals underwent injurious ventilation, were then subjected to bacterial challenge and finally continued to receive injurious ventilation. Fourth, P. aeruginosa is a cause of pneumonia in patients with specific risk factors such as airway diseases (i.e., chronic obstructive pulmonary disease) and immunosuppression. Fifth, there was not a control group of pigs without pneumonia that received treatment with aggressive ventilation.

Conclusion

In conclusion, we established an accurate pulmonary sepsis-induced ARDS model. To carry out this model, it is necessary to induce bilateral pneumonia and increase pulmonary damage with VILI. The clinically relevant pulmonary insults and features exhibited, long duration and capacity for invasive procedures and continuous monitoring to be performed constitute the most important elements of this model. For those reasons stated, we believe that this model is appropriate for studying novel lung protective strategies and respiratory rescue therapies for very severe ARDS.

Acknowledgements

We would like to thank Cristina Miralles who included and processed all of the histological samples. We would also like to thank Anthony Armenta for reviewing the English language and syntax used throughout the manuscript.

Declarations

All procedures were done following the European Directive 2010/63/UE and Spanish RD 53/2013 regulations related to the Guide for the Care and Use of Laboratory Animals. The study protocol was approved by the Animal Experimentation Ethics Committee of the University of Barcelona (approval reference number: DTES 10543).

Competing interests

The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Ranieri VM, Rubenfeld GD, Thompson BT, Ferguson ND, Caldwell E, Fan E, Camporota L, Slutsky AS. Acute respiratory distress syndrome: the Berlin definition. JAMA. 2012;307(23):2526–33.PubMed Ranieri VM, Rubenfeld GD, Thompson BT, Ferguson ND, Caldwell E, Fan E, Camporota L, Slutsky AS. Acute respiratory distress syndrome: the Berlin definition. JAMA. 2012;307(23):2526–33.PubMed
2.
Zurück zum Zitat Kolobow T, Moretti MP, Fumagalli R, Mascheroni D, Prato P, Chen V, Joris M. Severe impairment in lung function induced by high peak airway pressure during mechanical ventilation. An experimental study. Am Rev Respir Dis. 1987;135(2):312–5.PubMed Kolobow T, Moretti MP, Fumagalli R, Mascheroni D, Prato P, Chen V, Joris M. Severe impairment in lung function induced by high peak airway pressure during mechanical ventilation. An experimental study. Am Rev Respir Dis. 1987;135(2):312–5.PubMed
3.
Zurück zum Zitat Broccard A, Shapiro RS, Schmitz LL, Adams AB, Nahum A, Marini JJ. Prone positioning attenuates and redistributes ventilator-induced lung injury in dogs. Crit Care Med. 2000;28(2):295–303.PubMedCrossRef Broccard A, Shapiro RS, Schmitz LL, Adams AB, Nahum A, Marini JJ. Prone positioning attenuates and redistributes ventilator-induced lung injury in dogs. Crit Care Med. 2000;28(2):295–303.PubMedCrossRef
4.
Zurück zum Zitat Protti A, Andreis DT, Monti M, Santini A, Sparacino CC, Langer T, Votta E, Gatti S, Lombardi L, Leopardi O, Masson S, Cressoni M, Gattinoni L. Lung stress and strain during mechanical ventilation: any difference between statics and dynamics? Crit Care Med. 2013;41(4):1046–55.PubMedCrossRef Protti A, Andreis DT, Monti M, Santini A, Sparacino CC, Langer T, Votta E, Gatti S, Lombardi L, Leopardi O, Masson S, Cressoni M, Gattinoni L. Lung stress and strain during mechanical ventilation: any difference between statics and dynamics? Crit Care Med. 2013;41(4):1046–55.PubMedCrossRef
5.
Zurück zum Zitat Cressoni M, Gotti M, Chiurazzi C, Massari D, Algieri I, Amini M, Cammaroto A, Brioni M, Montaruli C, Nikolla K, Guanziroli M, Dondossola D, Gatti S, Valerio V, Vergani GL, Pugni P, Cadringher P, Gagliano N, Gattinoni L. Mechanical power and development of ventilator-induced lung injury. Anesthesiology. 2016;124(5):1100–8.PubMedCrossRef Cressoni M, Gotti M, Chiurazzi C, Massari D, Algieri I, Amini M, Cammaroto A, Brioni M, Montaruli C, Nikolla K, Guanziroli M, Dondossola D, Gatti S, Valerio V, Vergani GL, Pugni P, Cadringher P, Gagliano N, Gattinoni L. Mechanical power and development of ventilator-induced lung injury. Anesthesiology. 2016;124(5):1100–8.PubMedCrossRef
6.
Zurück zum Zitat Bellani G, Laffey JG, Pham T, Fan E, Brochard L, Esteban A, Gattinoni L, van Haren F, Larsson A, McAuley DF, Ranieri M, Rubenfeld G, Thompson BT, Wrigge H, Slutsky AS, Pesenti A; LUNG SAFE Investigators; ESICM Trials Group. Epidemiology, patterns of care, and mortality for patients with acute respiratory distress syndrome in intensive care units in 50 countries. JAMA. 2016;315(8): 788–800. Bellani G, Laffey JG, Pham T, Fan E, Brochard L, Esteban A, Gattinoni L, van Haren F, Larsson A, McAuley DF, Ranieri M, Rubenfeld G, Thompson BT, Wrigge H, Slutsky AS, Pesenti A; LUNG SAFE Investigators; ESICM Trials Group. Epidemiology, patterns of care, and mortality for patients with acute respiratory distress syndrome in intensive care units in 50 countries. JAMA. 2016;315(8): 788–800.
7.
Zurück zum Zitat Engel M, Nowacki RME, Jonker EM, Ophelders D, Nikiforou M, Kloosterboer N, Zimmermann LJI, van Waardenburg DA, Kramer BW. A comparison of four different models of acute respiratory distress syndrome in sheep. Respir Res. 2020;21(1):1–11.CrossRef Engel M, Nowacki RME, Jonker EM, Ophelders D, Nikiforou M, Kloosterboer N, Zimmermann LJI, van Waardenburg DA, Kramer BW. A comparison of four different models of acute respiratory distress syndrome in sheep. Respir Res. 2020;21(1):1–11.CrossRef
9.
Zurück zum Zitat Muellenbach RM, Kredel M, Bernd Z, Johannes A, Kuestermann J, Schuster F, Schwemmer U, Wurmb T, Wunder C, Roewer N, Brederlau J. Acute respiratory distress induced by repeated saline lavage provides stable experimental conditions for 24 hours in pigs. Exp Lung Res. 2009;35(3):222–33.PubMedCrossRef Muellenbach RM, Kredel M, Bernd Z, Johannes A, Kuestermann J, Schuster F, Schwemmer U, Wurmb T, Wunder C, Roewer N, Brederlau J. Acute respiratory distress induced by repeated saline lavage provides stable experimental conditions for 24 hours in pigs. Exp Lung Res. 2009;35(3):222–33.PubMedCrossRef
10.
Zurück zum Zitat van der Kloot TE, Blanch L, Youngblood AM, Weinert C, Adams AB, Marini JJ, Shapiro RS, Nahum A. Recruitment maneuvers in three experimental models of acute lung injury: effect on lung volume and gas exchange. Am J Respir Crit Care Med. 2000;161(5):1485–94.PubMedCrossRef van der Kloot TE, Blanch L, Youngblood AM, Weinert C, Adams AB, Marini JJ, Shapiro RS, Nahum A. Recruitment maneuvers in three experimental models of acute lung injury: effect on lung volume and gas exchange. Am J Respir Crit Care Med. 2000;161(5):1485–94.PubMedCrossRef
11.
Zurück zum Zitat Li Bassi G, Rigol M, Marti JD, Saucedo L, Ranzani OT, Roca I, Cabanas M, Muñoz L, Giunta V, Luque N, Rinaudo M, Esperatti M, Fernandez-Barat L, Ferrer M, Vila J, Ramirez J, Torres A. A novel porcine model of ventilator-associated pneumonia caused by oropharyngeal challenge with pseudomonas aeruginosa. Anesthesiology. 2014;120(5):1205–15.PubMedCrossRef Li Bassi G, Rigol M, Marti JD, Saucedo L, Ranzani OT, Roca I, Cabanas M, Muñoz L, Giunta V, Luque N, Rinaudo M, Esperatti M, Fernandez-Barat L, Ferrer M, Vila J, Ramirez J, Torres A. A novel porcine model of ventilator-associated pneumonia caused by oropharyngeal challenge with pseudomonas aeruginosa. Anesthesiology. 2014;120(5):1205–15.PubMedCrossRef
12.
Zurück zum Zitat Protti A, Andreis DT, Milesi M, Iapichino GE, Monti M, Comini B, Pugni P, Melis V, Santini A, Dondossola D, Gatti S, Lombardi L, Votta E, Carlesso E, Gattinoni L. Lung anatomy, energy load, and ventilator-induced lung injury. Intensive Care Med Exp. 2015;3(1):1–18.CrossRef Protti A, Andreis DT, Milesi M, Iapichino GE, Monti M, Comini B, Pugni P, Melis V, Santini A, Dondossola D, Gatti S, Lombardi L, Votta E, Carlesso E, Gattinoni L. Lung anatomy, energy load, and ventilator-induced lung injury. Intensive Care Med Exp. 2015;3(1):1–18.CrossRef
13.
Zurück zum Zitat Amato MBP, Meade MO, Slutsky AS, Brochard L, Costa ELV, Schoenfeld DA, Stewart TE, Briel M, Talmor D, Mercat A, Richard JC, Carvalho CR, Brower RG. Driving pressure and survival in the acute respiratory distress syndrome. N Engl J Med. 2015;372(8):747–55.PubMedCrossRef Amato MBP, Meade MO, Slutsky AS, Brochard L, Costa ELV, Schoenfeld DA, Stewart TE, Briel M, Talmor D, Mercat A, Richard JC, Carvalho CR, Brower RG. Driving pressure and survival in the acute respiratory distress syndrome. N Engl J Med. 2015;372(8):747–55.PubMedCrossRef
14.
Zurück zum Zitat American Thoracic Society and Infectious Diseases Society of America. Guidelines for the management of adults with hospital-acquired, ventilator-associated, and healthcare-associated pneumonia. Am J Respir Crit Care Med. 2005; 171(4): 388. American Thoracic Society and Infectious Diseases Society of America. Guidelines for the management of adults with hospital-acquired, ventilator-associated, and healthcare-associated pneumonia. Am J Respir Crit Care Med. 2005; 171(4): 388.
15.
Zurück zum Zitat Torres A, Niederman MS, Chastre J, Ewig S, Fernandez-Vandellos P, Hanberger H, Kollef M, Li Bassi G, Luna CM, Martin-Loeches I, Paiva JA, Read RC, Rigau D, Timsit JF, Welte T, Wunderink R. International ERS/ESICM/ESCMID/ALAT guidelines for the management of hospital-acquired pneumonia and ventilator-associated pneumonia: Guidelines for the management of hospital-acquired pneumonia (HAP)/ventilator-associated pneumonia (VAP) of the European Respiratory Society (ERS), European Society of Intensive Care Medicine (ESICM), European Society of Clinical Microbiology and Infectious Diseases (ESCMID) and Asociación Latinoamericana del Tórax (ALAT). Eur Respir J. 2017;50(3):1700582.PubMedCrossRef Torres A, Niederman MS, Chastre J, Ewig S, Fernandez-Vandellos P, Hanberger H, Kollef M, Li Bassi G, Luna CM, Martin-Loeches I, Paiva JA, Read RC, Rigau D, Timsit JF, Welte T, Wunderink R. International ERS/ESICM/ESCMID/ALAT guidelines for the management of hospital-acquired pneumonia and ventilator-associated pneumonia: Guidelines for the management of hospital-acquired pneumonia (HAP)/ventilator-associated pneumonia (VAP) of the European Respiratory Society (ERS), European Society of Intensive Care Medicine (ESICM), European Society of Clinical Microbiology and Infectious Diseases (ESCMID) and Asociación Latinoamericana del Tórax (ALAT). Eur Respir J. 2017;50(3):1700582.PubMedCrossRef
16.
Zurück zum Zitat Kulkami HS, Lee JS, Bastarache JA, Kuebler WM, Downey GP, Albaiceta GM, Altemeier WA, Artigas A, Bates JHT, Calfee CS, Dela Cruz CS, Dickson RP, Englert JA, Everitt JI, Fessler MB, Gelman AE, Gowdy KM, Groshong SD, Herold S, Homer RJ, Horowitz JC, Hsia CCW, Kurahashi K, Laubach VE, Looney MR, Lucas R, Mangalmurti NS, Manicone AM, Martin TR, Matalon S, Matthay MA, McAuley DF, McGrath-Morrow SA, Mizgerd JP, Montgomery SA, Moore BB, Noël A, Perlman CE, Reilly JP, Schmidt EP, Skerrett SJ, Suber TL, Summers C, Suratt BT, Takata M, Tuder R, Uhlig S, Witzenrath M, Zemans RL, Matute-Bello G. Update on the features and measurements of experimental acute lung injury in animals an official American Thoracic Society Workshop report. Am J Respir Cell Mol Biol. 2022;66(2):e1–14.CrossRef Kulkami HS, Lee JS, Bastarache JA, Kuebler WM, Downey GP, Albaiceta GM, Altemeier WA, Artigas A, Bates JHT, Calfee CS, Dela Cruz CS, Dickson RP, Englert JA, Everitt JI, Fessler MB, Gelman AE, Gowdy KM, Groshong SD, Herold S, Homer RJ, Horowitz JC, Hsia CCW, Kurahashi K, Laubach VE, Looney MR, Lucas R, Mangalmurti NS, Manicone AM, Martin TR, Matalon S, Matthay MA, McAuley DF, McGrath-Morrow SA, Mizgerd JP, Montgomery SA, Moore BB, Noël A, Perlman CE, Reilly JP, Schmidt EP, Skerrett SJ, Suber TL, Summers C, Suratt BT, Takata M, Tuder R, Uhlig S, Witzenrath M, Zemans RL, Matute-Bello G. Update on the features and measurements of experimental acute lung injury in animals an official American Thoracic Society Workshop report. Am J Respir Cell Mol Biol. 2022;66(2):e1–14.CrossRef
17.
Zurück zum Zitat Zhou Z, Kozlowski J, Schuster DP. Physiologic, biochemical, and imaging characterization of acute lung injury in mice. Am J Respir Crit Care Med. 2005;172(3):344–51.PubMedPubMedCentralCrossRef Zhou Z, Kozlowski J, Schuster DP. Physiologic, biochemical, and imaging characterization of acute lung injury in mice. Am J Respir Crit Care Med. 2005;172(3):344–51.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Ballard-Croft C, Wang D, Sumpter LR, Zhou X, Zwischenberger JB. Large-animal models of acute respiratory distress syndrome. Ann Thorac Surg. 2012;93(4):1331–9.PubMedCrossRef Ballard-Croft C, Wang D, Sumpter LR, Zhou X, Zwischenberger JB. Large-animal models of acute respiratory distress syndrome. Ann Thorac Surg. 2012;93(4):1331–9.PubMedCrossRef
19.
Zurück zum Zitat Pagnamenta A, Bouckaert Y, Wauthy P, Brimioulle S, Naeije R. Continuous versus pulsatile pulmonary hemodynamics in canine oleic acid lung injury. Am J Respir Crit Care Med. 2000;162(3):936–40.PubMedCrossRef Pagnamenta A, Bouckaert Y, Wauthy P, Brimioulle S, Naeije R. Continuous versus pulsatile pulmonary hemodynamics in canine oleic acid lung injury. Am J Respir Crit Care Med. 2000;162(3):936–40.PubMedCrossRef
20.
Zurück zum Zitat Shah NS, Nakayama DK, Jacob TD, Nishio I, Imai T, Billiar TR, Exler R, Yousem SA, Motoyama EK, Peitzman AB, et al. Efficacy of inhaled nitric oxide in oleic acid-induced acute lung injury. Crit Care Med. 1997;25(1):153–8.PubMedCrossRef Shah NS, Nakayama DK, Jacob TD, Nishio I, Imai T, Billiar TR, Exler R, Yousem SA, Motoyama EK, Peitzman AB, et al. Efficacy of inhaled nitric oxide in oleic acid-induced acute lung injury. Crit Care Med. 1997;25(1):153–8.PubMedCrossRef
21.
Zurück zum Zitat Grotjohan HP, van der Heijde RMJL, Jansen JRC, Wagenvoort CA, Versprille A. A stable model of respiratory distress by small injections of oleic acid in pigs. Intensive Care Med. 1996;22(4):336–44.PubMedCrossRef Grotjohan HP, van der Heijde RMJL, Jansen JRC, Wagenvoort CA, Versprille A. A stable model of respiratory distress by small injections of oleic acid in pigs. Intensive Care Med. 1996;22(4):336–44.PubMedCrossRef
22.
Zurück zum Zitat Gonçalves-De-Albuquerque CF, Silva AR, Burth P, de Moraes IMM, Oliveira FMDJ, Younes-Ibrahim M, dos Santos MC, D’Ávila H, Bozza PT, Faria Neto HC, Faria MV. Oleic acid induces lung injury in mice through activation of the ERK pathway. Mediators Inflamm. 2012;2012:956509.PubMedPubMedCentralCrossRef Gonçalves-De-Albuquerque CF, Silva AR, Burth P, de Moraes IMM, Oliveira FMDJ, Younes-Ibrahim M, dos Santos MC, D’Ávila H, Bozza PT, Faria Neto HC, Faria MV. Oleic acid induces lung injury in mice through activation of the ERK pathway. Mediators Inflamm. 2012;2012:956509.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Borges AM, Ferrari RS, Thomaz LDGR, Ulbrich JM, Félix EA, Silvello D, Andrade CF. Challenges and perspectives in porcine model of acute lung injury using oleic acid. Pulm Pharmacol Ther. 2019;59:101837.PubMedCrossRef Borges AM, Ferrari RS, Thomaz LDGR, Ulbrich JM, Félix EA, Silvello D, Andrade CF. Challenges and perspectives in porcine model of acute lung injury using oleic acid. Pulm Pharmacol Ther. 2019;59:101837.PubMedCrossRef
24.
Zurück zum Zitat Richard JC, Bregeon F, Leray V, le Bars D, Costes N, Tourvieille C, Lavenne F, Devouassoux-Shisheboran M, Gimenez G, Guerin C. Effect of activated protein C on pulmonary blood flow and cytokine production in experimental acute lung injury. Intensive Care Med. 2007;33(12):2199–206. PubMedCrossRef Richard JC, Bregeon F, Leray V, le Bars D, Costes N, Tourvieille C, Lavenne F, Devouassoux-Shisheboran M, Gimenez G, Guerin C. Effect of activated protein C on pulmonary blood flow and cytokine production in experimental acute lung injury. Intensive Care Med. 2007;33(12):2199–206. PubMedCrossRef
25.
Zurück zum Zitat Kamuf J, Garcia-Bardon A, Ziebart A, Thomas R, Rümmler R, Möllmann C, Hartmann EK. Oleic acid-injection in pigs as a model for acute respiratory distress syndrome. J Vis Exp. 2018;140:e57783. Kamuf J, Garcia-Bardon A, Ziebart A, Thomas R, Rümmler R, Möllmann C, Hartmann EK. Oleic acid-injection in pigs as a model for acute respiratory distress syndrome. J Vis Exp. 2018;140:e57783.
26.
Zurück zum Zitat Treml B, Neu N, Kleinsasser A, Gritsch C, Finsterwalder T, Geiger R, Schuster M, Janzek E, Loibner H, Penninger J, Loeckinger A. Recombinant angiotensin-converting enzyme 2 improves pulmonary blood flow and oxygenation in lipopolysaccharide-induced lung injury in piglets. Crit Care Med. 2010;38(2):596–601.PubMedCrossRef Treml B, Neu N, Kleinsasser A, Gritsch C, Finsterwalder T, Geiger R, Schuster M, Janzek E, Loibner H, Penninger J, Loeckinger A. Recombinant angiotensin-converting enzyme 2 improves pulmonary blood flow and oxygenation in lipopolysaccharide-induced lung injury in piglets. Crit Care Med. 2010;38(2):596–601.PubMedCrossRef
27.
Zurück zum Zitat Mohammed RUR, Zollinger NT, McCain AR, Romaguera-Matas R, Harris SP, Buesing KL, Borden MA, Terry BS. Testing oxygenated microbubbles via intraperitoneal and intrathoracic routes on a large pig model of LPS-induced acute respiratory distress syndrome. Physiol Rep. 2022;10(17):e15451.PubMedPubMedCentralCrossRef Mohammed RUR, Zollinger NT, McCain AR, Romaguera-Matas R, Harris SP, Buesing KL, Borden MA, Terry BS. Testing oxygenated microbubbles via intraperitoneal and intrathoracic routes on a large pig model of LPS-induced acute respiratory distress syndrome. Physiol Rep. 2022;10(17):e15451.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Zhou X, Wang D, Ballard-Croft CK, Simon SR, Lee HM, Zwischenberger JB. A tetracycline analog improves acute respiratory distress syndrome survival in an ovine model. Ann Thorac Surg. 2010;90(2):419–26.PubMedCrossRef Zhou X, Wang D, Ballard-Croft CK, Simon SR, Lee HM, Zwischenberger JB. A tetracycline analog improves acute respiratory distress syndrome survival in an ovine model. Ann Thorac Surg. 2010;90(2):419–26.PubMedCrossRef
29.
Zurück zum Zitat Yamamoto Y, Enkhbaatar P, Sousse LE, Sakurai H, Rehberg SW, Asmussen S, Kraft ER, Wright CL, Bartha E, Cox RA, Hawkins HK, Traber LD, Traber MG, Szabo C, Herndon DN, Traber DL. Nebulization with γ-tocopherol ameliorates acute lung injury after burn and smoke inhalation in the ovine model. Shock. 2012;37(4):408.PubMedPubMedCentralCrossRef Yamamoto Y, Enkhbaatar P, Sousse LE, Sakurai H, Rehberg SW, Asmussen S, Kraft ER, Wright CL, Bartha E, Cox RA, Hawkins HK, Traber LD, Traber MG, Szabo C, Herndon DN, Traber DL. Nebulization with γ-tocopherol ameliorates acute lung injury after burn and smoke inhalation in the ovine model. Shock. 2012;37(4):408.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Palmieri TL, Enkhbaatar P, Bayliss R, Traber LD, Cox RA, Hawkins HK, Herndon DN, Greenhalgh DG, Traber DL. Continuous nebulized albuterol attenuates acute lung injury in an ovine model of combined burn and smoke inhalation. Crit Care Med. 2006;34(6):1719–24.PubMedCrossRef Palmieri TL, Enkhbaatar P, Bayliss R, Traber LD, Cox RA, Hawkins HK, Herndon DN, Greenhalgh DG, Traber DL. Continuous nebulized albuterol attenuates acute lung injury in an ovine model of combined burn and smoke inhalation. Crit Care Med. 2006;34(6):1719–24.PubMedCrossRef
31.
Zurück zum Zitat Leiphrakpam PD, Weber HR, McCain A, Matas RR, Duarte EM, Buesing KL. A novel large animal model of smoke inhalation-induced acute respiratory distress syndrome. Respir Res. 2021;22(1):1–15.CrossRef Leiphrakpam PD, Weber HR, McCain A, Matas RR, Duarte EM, Buesing KL. A novel large animal model of smoke inhalation-induced acute respiratory distress syndrome. Respir Res. 2021;22(1):1–15.CrossRef
32.
Zurück zum Zitat Mountford PA, Leiphrakpam PD, Weber HR, McCain A, Scribner RT, Scribner RM, Martínez E, Chen J, Borden MA, Buesing K. Colonic Oxygen Microbubbles Augment Systemic Oxygenation and CO2 Removal in a Porcine Smoke Inhalation Model of Severe Hypoxia. bioRxiv. 2021. Mountford PA, Leiphrakpam PD, Weber HR, McCain A, Scribner RT, Scribner RM, Martínez E, Chen J, Borden MA, Buesing K. Colonic Oxygen Microbubbles Augment Systemic Oxygenation and CO2 Removal in a Porcine Smoke Inhalation Model of Severe Hypoxia. bioRxiv. 2021.
33.
Zurück zum Zitat Manitsopoulos N, Orfanos SE, Kotanidou A, Nikitopoulou I, Siempos I, Magkou C, Dimopoulou I, Zakynthinos SG, Armaganidis A, Maniatis NA. Inhibition of HMGCoA reductase by simvastatin protects mice from injurious mechanical ventilation. Respir Res. 2015;16(1):1–9.CrossRef Manitsopoulos N, Orfanos SE, Kotanidou A, Nikitopoulou I, Siempos I, Magkou C, Dimopoulou I, Zakynthinos SG, Armaganidis A, Maniatis NA. Inhibition of HMGCoA reductase by simvastatin protects mice from injurious mechanical ventilation. Respir Res. 2015;16(1):1–9.CrossRef
34.
Zurück zum Zitat Scheiermann J, Klinman DM. Suppressive oligonucleotides inhibit inflammation in a murine model of mechanical ventilator induced lung injury. J Thorac Dis. 2016;8(9):2434.PubMedPubMedCentralCrossRef Scheiermann J, Klinman DM. Suppressive oligonucleotides inhibit inflammation in a murine model of mechanical ventilator induced lung injury. J Thorac Dis. 2016;8(9):2434.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Wilson MR, Patel BV, Takata M. Ventilation with “clinically relevant” high tidal volumes does not promote stretch-induced injury in the lungs of healthy mice. Crit Care Med. 2012;40(10):2850.PubMedPubMedCentralCrossRef Wilson MR, Patel BV, Takata M. Ventilation with “clinically relevant” high tidal volumes does not promote stretch-induced injury in the lungs of healthy mice. Crit Care Med. 2012;40(10):2850.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Yildiz C, Palaniyar N, Otulakowski G, Khan MA, Post M, Kuebler WM, Tanswell K, Belcastro R, Masood A, Engelberts D, Kavanagh BP. Mechanical ventilation induces neutrophil extracellular trap formation. Anesthesiology. 2015;122(4):864–75.PubMedCrossRef Yildiz C, Palaniyar N, Otulakowski G, Khan MA, Post M, Kuebler WM, Tanswell K, Belcastro R, Masood A, Engelberts D, Kavanagh BP. Mechanical ventilation induces neutrophil extracellular trap formation. Anesthesiology. 2015;122(4):864–75.PubMedCrossRef
37.
Zurück zum Zitat Li LF, Lee CS, Liu YY, Chang CH, Lin CW, Chiu LC, Kao KC, Chen NH, Yang CT. Activation of Src-dependent Smad3 signaling mediates the neutrophilic inflammation and oxidative stress in hyperoxia-augmented ventilator-induced lung injury. Respir Res. 2015;16(1):1–14.CrossRef Li LF, Lee CS, Liu YY, Chang CH, Lin CW, Chiu LC, Kao KC, Chen NH, Yang CT. Activation of Src-dependent Smad3 signaling mediates the neutrophilic inflammation and oxidative stress in hyperoxia-augmented ventilator-induced lung injury. Respir Res. 2015;16(1):1–14.CrossRef
38.
Zurück zum Zitat Shi CS, Huang TH, Lin CK, Li JM, Chen MH, Tsai ML, Chang CC. VEGF production by Ly6C+high monocytes contributes to ventilator-induced lung injury. PLoS ONE. 2016;11(10):e0165317.PubMedPubMedCentralCrossRef Shi CS, Huang TH, Lin CK, Li JM, Chen MH, Tsai ML, Chang CC. VEGF production by Ly6C+high monocytes contributes to ventilator-induced lung injury. PLoS ONE. 2016;11(10):e0165317.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Lopez-Aguilar J, Quilez M, Marti-Sistac O, Garcia-Martin C, Fuster G, Puig F, Flores C, Villar J, Artigas A, Blanch L. Early physiological and biological features in three animal models of induced acute lung injury. Intensive Care Med. 2009;36(2):347–55.PubMedCrossRef Lopez-Aguilar J, Quilez M, Marti-Sistac O, Garcia-Martin C, Fuster G, Puig F, Flores C, Villar J, Artigas A, Blanch L. Early physiological and biological features in three animal models of induced acute lung injury. Intensive Care Med. 2009;36(2):347–55.PubMedCrossRef
40.
Zurück zum Zitat Araos J, Alegría L, García P, Damiani F, Tapia P, Soto D, Salomon T, Rodriguez F, Amthauer M, Erranz B, Castro G, Carreño P, Medina T, Retamal J, Cruces P, Bugedo G, Bruhn A. Extracorporeal membrane oxygenation improves survival in a novel 24-hour pig model of severe acute respiratory distress syndrome. Am J Transl Res. 2016;8(6):2826.PubMedPubMedCentral Araos J, Alegría L, García P, Damiani F, Tapia P, Soto D, Salomon T, Rodriguez F, Amthauer M, Erranz B, Castro G, Carreño P, Medina T, Retamal J, Cruces P, Bugedo G, Bruhn A. Extracorporeal membrane oxygenation improves survival in a novel 24-hour pig model of severe acute respiratory distress syndrome. Am J Transl Res. 2016;8(6):2826.PubMedPubMedCentral
41.
Zurück zum Zitat Sacks D, Baxter B, Campbell BCV, Carpenter JS, Cognard C, Dippel D, Eesa M, Fischer U, Hausegger K, Hirsch JA, Shazam Hussain M, Jansen O, Jayaraman MV, Khalessi AA, Kluck BW, Lavine S, Meyers PM, Ramee S, Rüfenacht DA, Schirmer CM, Vorwerk D. Multisociety consensus quality improvement revised consensus statement for endovascular therapy of acute ischemic stroke. Int J Stroke. 2018;39(6):612–32. Sacks D, Baxter B, Campbell BCV, Carpenter JS, Cognard C, Dippel D, Eesa M, Fischer U, Hausegger K, Hirsch JA, Shazam Hussain M, Jansen O, Jayaraman MV, Khalessi AA, Kluck BW, Lavine S, Meyers PM, Ramee S, Rüfenacht DA, Schirmer CM, Vorwerk D. Multisociety consensus quality improvement revised consensus statement for endovascular therapy of acute ischemic stroke. Int J Stroke. 2018;39(6):612–32.
42.
Zurück zum Zitat Sinclair SE, Altemeier WA, Matute-Bello G, Chi EY. Augmented lung injury due to interaction between hyperoxia and mechanical ventilation. Crit Care Med. 2004;32(12):2496–501.PubMedCrossRef Sinclair SE, Altemeier WA, Matute-Bello G, Chi EY. Augmented lung injury due to interaction between hyperoxia and mechanical ventilation. Crit Care Med. 2004;32(12):2496–501.PubMedCrossRef
43.
Zurück zum Zitat Joelsson JP, Ingthorsson S, Kricker J, Gudjonsson T, Karason S. Ventilator-induced lung-injury in mouse models: is there a trap? Lab Anim Res. 2021;37(1):1–11.CrossRef Joelsson JP, Ingthorsson S, Kricker J, Gudjonsson T, Karason S. Ventilator-induced lung-injury in mouse models: is there a trap? Lab Anim Res. 2021;37(1):1–11.CrossRef
44.
Zurück zum Zitat Wolf GK, Gómez-Laberge C, Rettig JS, Vargas SO, Smallwood CD, Prabhu SP, et al. Mechanical ventilation guided by electrical impedance tomography in experimental acute lung injury. Crit Care Med. 2013;41(5):1296–304.PubMedCrossRef Wolf GK, Gómez-Laberge C, Rettig JS, Vargas SO, Smallwood CD, Prabhu SP, et al. Mechanical ventilation guided by electrical impedance tomography in experimental acute lung injury. Crit Care Med. 2013;41(5):1296–304.PubMedCrossRef
45.
Zurück zum Zitat Acute Respiratory Distress Syndrome Network, Brower RG, Matthay MA, Morris A, Schoenfeld D, Thompson BT, Wheeler A. Ventilation with Lower Tidal Volumes as Compared with Traditional Tidal Volumes for Acute Lung Injury and the Acute Respiratory Distress Syndrome. N Engl J Med. 2000;342(18): 1301–1308. Acute Respiratory Distress Syndrome Network, Brower RG, Matthay MA, Morris A, Schoenfeld D, Thompson BT, Wheeler A. Ventilation with Lower Tidal Volumes as Compared with Traditional Tidal Volumes for Acute Lung Injury and the Acute Respiratory Distress Syndrome. N Engl J Med. 2000;342(18): 1301–1308.
46.
Zurück zum Zitat Montgomery AB, Stager MA, Carrico CJ, Hudson LD. Causes of mortality in patients with the adult respiratory distress syndrome. Am Rev Respir Dis. 1985;132(3):485–9.PubMed Montgomery AB, Stager MA, Carrico CJ, Hudson LD. Causes of mortality in patients with the adult respiratory distress syndrome. Am Rev Respir Dis. 1985;132(3):485–9.PubMed
47.
Zurück zum Zitat Villar J, Schultz MJ, Kacmarek RM. The LUNG SAFE: a biased presentation of the prevalence of ARDS! Crit Care. 2016;20(1):1–3.CrossRef Villar J, Schultz MJ, Kacmarek RM. The LUNG SAFE: a biased presentation of the prevalence of ARDS! Crit Care. 2016;20(1):1–3.CrossRef
Metadaten
Titel
A long-lasting porcine model of ARDS caused by pneumonia and ventilator-induced lung injury
verfasst von
Enric Barbeta
Marta Arrieta
Ana Motos
Joaquim Bobi
Hua Yang
Minlan Yang
Giacomo Tanzella
Pierluigi Di Ginnatale
Stefano Nogas
Carmen Rosa Vargas
Roberto Cabrera
Denise Battaglini
Andrea Meli
Kasra Kiarostami
Nil Vázquez
Laia Fernández-Barat
Montserrat Rigol
Ricard Mellado-Artigas
Gerard Frigola
Marta Camprubí-Rimblas
Pau Ferrer
Daniel Martinez
Antonio Artigas
Carlos Ferrando
Miquel Ferrer
Antoni Torres
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Critical Care / Ausgabe 1/2023
Elektronische ISSN: 1364-8535
DOI
https://doi.org/10.1186/s13054-023-04512-8

Weitere Artikel der Ausgabe 1/2023

Critical Care 1/2023 Zur Ausgabe

Blutdrucksenkung schon im Rettungswagen bei akutem Schlaganfall?

31.05.2024 Apoplex Nachrichten

Der optimale Ansatz für die Blutdruckkontrolle bei Patientinnen und Patienten mit akutem Schlaganfall ist noch nicht gefunden. Ob sich eine frühzeitige Therapie der Hypertonie noch während des Transports in die Klinik lohnt, hat jetzt eine Studie aus China untersucht.

Ähnliche Überlebensraten nach Reanimation während des Transports bzw. vor Ort

29.05.2024 Reanimation im Kindesalter Nachrichten

Laut einer Studie aus den USA und Kanada scheint es bei der Reanimation von Kindern außerhalb einer Klinik keinen Unterschied für das Überleben zu machen, ob die Wiederbelebungsmaßnahmen während des Transports in die Klinik stattfinden oder vor Ort ausgeführt werden. Jedoch gibt es dabei einige Einschränkungen und eine wichtige Ausnahme.

Nicht Creutzfeldt Jakob, sondern Abführtee-Vergiftung

29.05.2024 Hyponatriämie Nachrichten

Eine ältere Frau trinkt regelmäßig Sennesblättertee gegen ihre Verstopfung. Der scheint plötzlich gut zu wirken. Auf Durchfall und Erbrechen folgt allerdings eine Hyponatriämie. Nach deren Korrektur kommt es plötzlich zu progredienten Kognitions- und Verhaltensstörungen.

Häusliche Gewalt in der orthopädischen Notaufnahme oft nicht erkannt

28.05.2024 Häusliche Gewalt Nachrichten

In der Notaufnahme wird die Chance, Opfer von häuslicher Gewalt zu identifizieren, von Orthopäden und Orthopädinnen offenbar zu wenig genutzt. Darauf deuten die Ergebnisse einer Fragebogenstudie an der Sahlgrenska-Universität in Schweden hin.

Update AINS

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.