Summary

Halogenated Agent Delivery in Porcine Model of Acute Respiratory Distress Syndrome via an Intensive Care Unit Type Device

Published: September 24, 2020
doi:

Summary

We describe a model of hydrochloric acid-induced acute respiratory distress syndrome (ARDS) in piglets receiving sedation with halogenated agents, isoflurane and sevoflurane, through a device used for inhaled intensive care sedation. This model can be used to investigate the biological mechanisms of halogenated agents on lung injury and repair.

Abstract

Acute respiratory distress syndrome (ARDS) is a common cause of hypoxemic respiratory failure and death in critically ill patients, and there is an urgent need to find effective therapies. Preclinical studies have shown that inhaled halogenated agents may have beneficial effects in animal models of ARDS. The development of new devices to administer halogenated agents using modern intensive care unit (ICU) ventilators has significantly simplified the dispensing of halogenated agents to ICU patients. Because previous experimental and clinical research suggested potential benefits of halogenated volatiles, such as sevoflurane or isoflurane, for lung alveolar epithelial injury and inflammation, two pathophysiologic landmarks of diffuse alveolar damage during ARDS, we designed an animal model to understand the mechanisms of the effects of halogenated agents on lung injury and repair. After general anesthesia, tracheal intubation, and the initiation of mechanical ventilation, ARDS was induced in piglets via the intratracheal instillation of hydrochloric acid. Then, the piglets were sedated with inhaled sevoflurane or isoflurane using an ICU-type device, and the animals were ventilated with lung-protective mechanical ventilation during a 4 h period. During the study period, blood and alveolar samples were collected to evaluate arterial oxygenation, the permeability of the alveolar-capillary membrane, alveolar fluid clearance, and lung inflammation. Mechanical ventilation parameters were also collected throughout the experiment. Although this model induced a marked decrease in arterial oxygenation with altered alveolar-capillary permeability, it is reproducible and is characterized by a rapid onset, good stability over time, and no fatal complications.

We have developed a piglet model of acid aspiration that reproduces most of the physiological, biological, and pathological features of clinical ARDS, and it will be helpful to further our understanding of the potential lung-protective effects of halogenated agents delivered through devices used for inhaled ICU sedation.

Introduction

Acute respiratory distress syndrome (ARDS) is a common cause of hypoxemic respiratory failure and death in critically ill patients1. It is characterized by both diffuse alveolar epithelial and endothelial injuries, leading to increased permeability and pulmonary edema, altered alveolar fluid clearance (AFC), and worsened respiratory distress2. The resorption of alveolar edema and recovery from ARDS require epithelial fluid transport through the alveoli to remain intact, which suggests that a therapy improving AFC could be useful3,4. Although lung-protective ventilation and a restrictive strategy for intravenous fluid therapy have proven beneficial in improving outcomes2,5, they are still associated with high mortality and morbidity6. Therefore, there is an urgent need to develop effective therapies for the syndrome and to better understand the precise mechanisms through which such therapies might work.

Halogenated anesthetics, such as isoflurane or sevoflurane, have been widely used for general anesthesia in the operating room. Sevoflurane is associated with decreased inflammation in the lungs of patients undergoing thoracic surgery and with a decrease in postoperative pulmonary complications, such as ARDS7. Similar results have been found in a meta-analysis of patients after cardiac surgery8. Halogenated volatiles also have a bronchodilatory effect9,10 and perhaps some properties that protect several organs, such as the heart8,11 and the kidneys12,13,14. Recently, there has been growing interest in the clinical use of inhaled anesthetics as sedatives in the intensive care unit (ICU). Both animal and human studies support the protective effects of pretreatment with halogenated agents before prolonged ischemia of the liver15, the brain16, or the heart11. Halogenated agents also have potential pharmacokinetic and pharmacodynamic advantages over other intravenous agents for the sedation of critically ill patients, including a rapid onset of action and fast offset due to little accumulation in tissues. Inhaled halogenated agents decrease intubation times in comparison with intravenous sedation in patients undergoing cardiac surgery17. Several studies support the safety and efficacy of halogenated agents in the sedation of ICU patients18,19,20. In experimental models of ARDS, inhaled sevoflurane improves gas exchange21,22, reduces alveolar edema21,22, and attenuates both pulmonary and systemic inflammation23. Isoflurane also ameliorates lung repair after injury by maintaining the integrity of the alveolar-capillary barrier, possibly by modulating the expression of a key tight junction protein24,25,26. In addition, mouse macrophages that were cultured and treated with isoflurane had better phagocytic effects on neutrophils than macrophages that were not treated with isoflurane27.

However, the precise biological pathways and mechanisms accounting for the lung-protective properties of volatile anesthetics remain largely unknown to date, requiring further investigation18. Additional studies are also warranted to investigate the precise effects of sevoflurane on lung injury and to verify whether experimental evidence can be translated to patients. The first randomized control trial from our team found that the administration of inhaled sevoflurane in patients with ARDS was associated with oxygenation improvement and decreased levels of both pro-inflammatory cytokines and lung epithelial injury markers, as assessed by plasma and alveolar soluble receptors for advanced glycation end products (sRAGE)28. As sRAGE is now considered as a marker of alveolar type 1 cell injury and a key mediator of alveolar inflammation, these results could suggest some beneficial effects of sevoflurane on the lung alveolar epithelial injury21,29,30.

The use of halogenated agents for inhaled ICU sedation has long required operating room anesthesia ventilators and gas vaporizers to be deployed in the ICU. Since then, anesthetic reflectors suitable for the use with modern critical care ventilators have been developed for specific use in the ICU31. These devices feature modified heat and moisture exchanging filters inserted between the Y-piece of the respiratory circuit and the endotracheal tube. They allow the administration of halogenated agents, with isoflurane and sevoflurane being the most frequently used, and they consist of a porous polypropylene evaporator rod, into which a liquid agent, delivered by a specific syringe pump, is released. The halogenated agent is absorbed during expiration by a reflecting medium contained in the device and it is released during the next inspiration, allowing recirculation of approximately 90% of the expired halogenated agent31,32. Recently, a miniaturized version of the device was developed with an instrumental dead space of 50 mL, making it even more suitable for use during ultra-protective ventilation in ARDS patients, with tidal volumes that could be as low as 200 mL31. Such a miniaturized device has never been studied in an experimental piglet model of ARDS.

Because previous research supports the promising roles of halogenated volatiles in lung alveolar inflammation and injury during ARDS, we designed an experimental animal model to achieve a translational understanding of the mechanisms of the effects of halogenated agents on lung injury and repair33,34,35. In this study, we developed a model of hydrochloric acid (HCl)-induced ARDS in piglets in whom inhaled sedation can be delivered using the miniaturized version of the anesthetic conserving device, an ICU-type device. This large animal model of ARDS could be used to further our understanding of the potential lung-protective effects of inhaled halogenated agents.

Protocol

The study protocol was approved by the Animal Ethics Committee of the French Ministère de l’Education Nationale, de l’Enseignement Supérieur et de la Recherche (approval number 01505.03) before being registered at preclinicaltrials.eu (Pre-clinical registry identifier PCTE0000129). All procedures were performed in the Centre International de Chirurgie Endoscopique, Université Clermont Auvergne, Clermont-Ferrand, France, in accordance with the Animal Research: Reportin…

Representative Results

For this experiment, 25 piglets were anesthetized and divided in two groups: 12 piglets in the untreated group (SHAM group) and 13 piglets in the acid-injured group (HCl group). No piglet died before the end of the experiment. A two-way repeated-measures analysis of variance (RM-ANOVA) indicated a significant time by group interaction (P < 10−4) with a detrimental effect of HCl-induced ARDS on PaO2/FiO2, compared to sham animals without ARDS (Figure 3</stron…

Discussion

This article describes a reproducible experimental model of ARDS induced by the intratracheal instillation of HCl in piglets to investigate the lung-protective effects of halogenated volatiles, such as sevoflurane or isoflurane, delivered using an anesthetic conserving device.

The primary goal of this study was to develop an experimental model of ARDS in which volatile agents could be delivered by an anesthetic conserving device, such as those used in ICU patients. Although some effects of hal…

Declarações

The authors have nothing to disclose.

Acknowledgements

The authors would like to thank the staff from the GreD, the Université Clermont Auvergne, and the Centre International de Chirurgie Endoscopique (all in Clermont-Ferrand, France).

Materials

Tracheal intubation
Endotracheal tube 6-mm Covidien 18860
Animal preparation
Central venous catheter 3-lumens catheter (7 French – 16 cm) Arrow CV-12703
Pulse contour cardiac output monitor PiCCO catheter (3-5 French – 20 cm) Getinge Pulsion Medical System catheter
Warm blankets WarmTouch5300 MedTronic 5300
Monitoring
External monitor IntelliVue MP40 Phillips MNT 142
Point-of-care blood gas analyzer Epoc® Blood Analysis System Siemens 20093
Pulse contour cardiac output monitor PiCCO Device PulsioFlex Monitor Getinge Pulsion Medical System PulsioFlex
Mechanical ventilation
Ventilator Engström Carestation General Electrics Engström
Halogenated anesthetics
Anaconda Syringe SedanaMedical 26022
Anesthetic conserving device AnaConDa-S SedanaMedical 26050
Charcoal filter FlurAbsorb SedanaMedical 26096
Filling Adaptaters SedanaMedical 26042
Ionomer membrane dryer line Nafion SedanaMedical 26053
Products
Propofol Mylan 66617123
Isoflurane Virbac QN01AB06
Pentobarbital PanPharma 68942457
Sevoflurane Abbvie N01AB08
Sufentanil Mylan 62404996

Referências

  1. ARDS Definition Task Force et al. Acute respiratory distress syndrome: the Berlin Definition. JAMA: The Journal of the American Medical Association. 307 (23), 2526-2533 (2012).
  2. Thompson, B. T., Chambers, R. C., Liu, K. D. Acute Respiratory Distress Syndrome. The New England Journal of Medicine. 377 (6), 562-572 (2017).
  3. Ware, L. B., Matthay, M. A. Alveolar fluid clearance is impaired in the majority of patients with acute lung injury and the acute respiratory distress syndrome. American Journal of Respiratory and Critical Care Medicine. 163 (6), 1376-1383 (2001).
  4. McAuley, D. F., Frank, J. A., Fang, X., Matthay, M. A. Clinically relevant concentrations of beta2-adrenergic agonists stimulate maximal cyclic adenosine monophosphate-dependent airspace fluid clearance and decrease pulmonary edema in experimental acid-induced lung injury. Critical Care Medicine. 32 (7), 1470-1476 (2004).
  5. Fan, E., et al. An official American thoracic society/European society of intensive care medicine/society of critical care medicine clinical practice guideline: Mechanical ventilation in adult patients with acute respiratory distress syndrome. American Journal of Respiratory and Critical Care Medicine. 195 (9), 1253-1263 (2017).
  6. Bellani, G., et al. Epidemiology, patterns of care, and mortality for patients with acute respiratory distress syndrome in intensive care units in 50 countries. JAMA: The Journal of the American Medical Association. 315 (8), 788-800 (2016).
  7. De Conno, E., et al. Anesthetic-induced improvement of the inflammatory response to one-lung ventilation. Anesthesiology. 110 (6), 1316-1326 (2009).
  8. Uhlig, C., et al. Effects of volatile anesthetics on mortality and postoperative pulmonary and other complications in patients undergoing surgery: A systematic review and meta-analysis. Anesthesiology: The Journal of the American Society of Anesthesiologists. 124 (6), 1230-1245 (2016).
  9. Campagna, J. A., Miller, K. W., Forman, S. A. Mechanisms of actions of inhaled anesthetics. The New England Journal of Medicine. 348 (21), 2110-2124 (2003).
  10. Dikmen, Y., Eminoglu, E., Salihoglu, Z., Demiroluk, S. Pulmonary mechanics during isoflurane, sevoflurane and desflurane anaesthesia. Anaesthesia. 58 (8), 745-748 (2003).
  11. Hert, S. G. D., et al. Choice of primary anesthetic regimen can influence intensive care unit length of stay after coronary surgery with cardiopulmonary bypass. Anesthesiology. 101 (1), 9-20 (2004).
  12. Hashiguchi, H., et al. Isoflurane protects renal function against ischemia and reperfusion through inhibition of protein kinases, JNK and ERK. Anesthesia and Analgesia. , 1584-1589 (2005).
  13. Fukazawa, K., Lee, H. T. Volatile anesthetics and AKI: risks, mechanisms, and a potential therapeutic window. Journal of the American Society of Nephrology: JASN. 25 (5), 884-892 (2014).
  14. Obal, D., Rascher, K., Favoccia, C., Dettwiler, S., Schlack, W. Post-conditioning by a short administration of desflurane reduced renal reperfusion injury after differing of ischaemia times in rats. British Journal of Anaesthesia. 97 (6), 783-791 (2006).
  15. Lv, X., et al. Isoflurane preconditioning at clinically relevant doses induce protective effects of heme oxygenase-1 on hepatic ischemia reperfusion in rats. BMC Gastroenterology. 11, 31 (2011).
  16. Sakai, H., et al. Isoflurane provides long-term protection against focal cerebral ischemia in the rat. Anesthesiology. 106 (1), 92-99 (2007).
  17. Jerath, A., et al. Volatile-based short-term sedation in cardiac surgical patients: a prospective randomized controlled trial. Critical Care Medicine. 43 (5), 1062-1069 (2015).
  18. Jerath, A., Parotto, M., Wasowicz, M., Ferguson, N. D. Volatile Anesthetics. Is a New Player Emerging in Critical Care Sedation. American Journal of Respiratory and Critical Care Medicine. 193 (11), 1202-1212 (2016).
  19. Perbet, S., et al. A pharmacokinetic study of 48-hour sevoflurane inhalation using a disposable delivery system (AnaConDa®) in ICU patients. Minerva Anestesiologica. 80 (6), 655-665 (2014).
  20. Mesnil, M., et al. Long-term sedation in intensive care unit: a randomized comparison between inhaled sevoflurane and intravenous propofol or midazolam. Intensive Care Medicine. 37 (6), 933-941 (2011).
  21. Schläpfer, M., et al. Sevoflurane reduces severity of acute lung injury possibly by impairing formation of alveolar oedema. Clinical and Experimental Immunology. 168 (1), 125-134 (2012).
  22. Voigtsberger, S., et al. Sevoflurane ameliorates gas exchange and attenuates lung damage in experimental lipopolysaccharide-induced lung Injury. Anesthesiology. 111 (6), 1238-1248 (2009).
  23. Steurer, M., et al. The volatile anaesthetic sevoflurane attenuates lipopolysaccharide-induced injury in alveolar macrophages. Clinical and Experimental Immunology. 155 (2), 224-230 (2009).
  24. Englert, J. A., et al. Isoflurane Ameliorates Acute Lung Injury by Preserving Epithelial Tight Junction Integrity. Anesthesiology. 123 (2), 377-388 (2015).
  25. Li, Q. F., Zhu, Y. S., Jiang, H., Xu, H., Sun, Y. Isoflurane preconditioning ameliorates endotoxin-induced acute lung injury and mortality in rats. Anesthesia and Analgesia. 109 (5), 1591-1597 (2009).
  26. Reutershan, J., Chang, D., Hayes, J. K., Ley, K. Role of a reduction of cytokine levels in isoflurane-mediated protection from endotoxin-induced lung Injury. Anesthesiology. 105 (6), 1280-1281 (2006).
  27. Du, X., et al. Isoflurane promotes phagocytosis of apoptotic neutrophils through AMPK-mediated ADAM17/Mer signaling. PloS One. 12 (7), 0180213 (2017).
  28. Jabaudon, M., et al. Sevoflurane for Sedation in Acute Respiratory Distress Syndrome. A Randomized Controlled Pilot Study. American Journal of Respiratory and Critical Care Medicine. 195 (6), 792-800 (2017).
  29. Yue, T., et al. Postconditioning with a volatile anaesthetic in alveolar epithelial cells in vitro. The European Respiratory Journal: Official Journal of the European Society for Clinical Respiratory Physiology. 31 (1), 118-125 (2008).
  30. Blondonnet, R., Constantin, J. M., Sapin, V., Jabaudon, M. A pathophysiologic approach to biomarkers in acute respiratory distress syndrome. Disease Markers. 2016, 3501373 (2016).
  31. Farrell, R., Oomen, G., Carey, P. A technical review of the history, development and performance of the anaesthetic conserving device “AnaConDa” for delivering volatile anaesthetic in intensive and post-operative critical care. Journal of Clinical Monitoring and Computing. 32 (4), 595-604 (2018).
  32. Sturesson, L. W., Bodelsson, M., Jonson, B., Malmkvist, G. Anaesthetic conserving device AnaConDa: dead space effect and significance for lung protective ventilation. British Journal of Anaesthesia. 113 (3), 508-514 (2014).
  33. Blondonnet, R., et al. RAGE inhibition reduces acute lung injury in mice. Scientific Reports. 7 (1), 7208 (2017).
  34. Jabaudon, M., et al. Soluble receptor for advanced glycation end-products predicts impaired alveolar fluid clearance in acute respiratory distress syndrome. American Journal of Respiratory and Critical Care Medicine. 192 (2), 191-199 (2015).
  35. Jabaudon, M., et al. Soluble forms and ligands of the receptor for advanced glycation end-products in patients with acute respiratory distress syndrome: An observational prospective study. PloS One. 10 (8), 0135857 (2015).
  36. Kilkenny, C., Browne, W. J., Cuthill, I. C., Emerson, M., Altman, D. G. Improving bioscience research reporting: The ARRIVE guidelines for reporting animal research. PLoS Biology. 8, 1000412 (2010).
  37. Audard, J., et al. Inhibition of the receptor for advanced glycation end-products in acute respiratory distress syndrome: A randomised laboratory trial in piglets. Scientific Reports. 9 (1), 9227 (2019).
  38. Wu, C. W., et al. Intra-operative neural monitoring of thyroid surgery in a porcine model. Journal of Visualized Experiments. (144), e57919 (2019).
  39. Russ, M., et al. Lavage-induced surfactant depletion in pigs as a model of the acute respiratory distress syndrome (ARDS). Journal of visualized experiments. (115), e53610 (2016).
  40. Marumo, C. K., et al. Hemodynamic effects of PEEP in a porcine model of HCl-induced mild acute lung injury. Acta Anaesthesiologica Scandinavica. 53 (2), 190-202 (2009).
  41. Ambrosio, A. M., et al. Effects of positive end-expiratory pressure titration and recruitment maneuver on lung inflammation and hyperinflation in experimental acid aspiration-induced lung injury. Anesthesiology. 117 (6), 1322-1334 (2012).
  42. Sackey, P. V., Martling, C. R., Granath, F., Radell, P. J. Prolonged isoflurane sedation of intensive care unit patients with the Anesthetic Conserving Device. Critical Care Medicine. 32 (11), 2241-2246 (2004).
  43. Blanchard, F., et al. Minimal alveolar concentration for deep sedation (MAC-DS) in intensive care unit patients sedated with sevoflurane: A physiological study. Anaesthesia, Critical Care & Pain. , (2020).
  44. Verghese, G. M., Ware, L. B., Matthay, B. A., Matthay, M. A. Alveolar epithelial fluid transport and the resolution of clinically severe hydrostatic pulmonary edema. Journal of Applied Physiology. 87 (4), 1301-1312 (1999).
  45. Sakuma, T., et al. Alveolar fluid clearance in the resected human lung. American Journal of Respiratory and Critical Care Medicine. 150 (2), 305-310 (1994).
  46. Matthay, M. A., Wiener-Kronish, J. P. Intact epithelial barrier function is critical for the resolution of alveolar edema in humans. The American Review of Respiratory Disease. 142 (6), 1250-1257 (1990).
  47. Ware, L. B., Matthay, M. A. Alveolar fluid clearance is impaired in the majority of patients with acute lung injury and the acute respiratory distress syndrome. American Journal of Respiratory and Critical Care Medicine. 163 (6), 1376-1383 (2001).
  48. Ware, L. B., Golden, J. A., Finkbeiner, W. E., Matthay, M. A. Alveolar epithelial fluid transport capacity in reperfusion lung injury after lung transplantation. American Journal of Respiratory and Critical Care Medicine. 159 (3), 980-988 (1999).
  49. Constantin, J. M., et al. Response to recruitment maneuver influences net alveolar fluid clearance in acute respiratory distress syndrome. Anesthesiology. 106 (5), 944-951 (2007).
  50. Kemming, G. I., et al. Effects of perfluorohexan vapor on gas exchange, respiratory mechanics, and lung histology in pigs with lung injury after endotoxin infusion. Anesthesiology. 103 (3), 585-594 (2005).
  51. Matute-Bello, G., et al. An official American Thoracic Society workshop report: features and measurements of experimental acute lung injury in animals. American Journal of Respiratory Cell and Molecular Biology. 44 (5), 725-738 (2011).
  52. Chiew, Y. S., et al. Physiological relevance and performance of a minimal lung model: an experimental study in healthy and acute respiratory distress syndrome model piglets. BMC Pulmonary Medicine. 12, 59 (2012).
  53. Hochhausen, N., et al. Optimizing PEEP by Electrical Impedance Tomography in a Porcine Animal Model of ARDS. Respiratory Care. 62 (3), 340-349 (2017).
  54. Fu, H., Sun, M., Miao, C. Effects of different concentrations of isoflurane pretreatment on respiratory mechanics, oxygenation and hemodynamics in LPS-induced acute respiratory distress syndrome model of juvenile piglets. Experimental Lung Research. 41 (8), 415-421 (2015).
  55. Yehya, N. Lessons learned in acute respiratory distress syndrome from the animal laboratory. Annals of Translational Medicine. 7 (19), 503 (2019).
  56. Hochhausen, N., et al. Comparison of two experimental ARDS models in pigs using electrical impedance tomography. PloS One. 14 (11), 0225218 (2019).
  57. Shaver, C. M., et al. Cell-free hemoglobin: a novel mediator of acute lung injury. American Journal of Physiology. Lung Cellular and Molecular Physiology. 310 (6), 532-541 (2016).
  58. Matthay, M. A., et al. Acute respiratory distress syndrome. Nature Reviews. Disease Primers. 5 (1), 18 (2019).
  59. Martin, T. R., Matute-Bello, G. Experimental models and emerging hypotheses for acute lung injury. Critical Care Clinics. 27 (3), 735-752 (2011).
  60. Light, R. W., Gary Lee, Y. C. . Textbook of Pleural Diseases Second Edition. , (2008).
  61. Laferriere-Langlois, P., d’Arogon, F., Manzanares, W. Halogenated volatile anesthetics in the intensive care unit: current knowledge on an upcoming practice. Minerva Anestesiologica. 83 (7), 737-748 (2017).
  62. Devlin, J. W., et al. Clinical Practice Guidelines for the Prevention and Management of Pain, Agitation/Sedation, Delirium, Immobility, and Sleep Disruption in Adult Patients in the ICU. Critical Care Medicine. 46 (9), 825-873 (2018).
  63. DAS-Taskforce 2015 et al. Evidence and consensus based guideline for the management of delirium, analgesia, and sedation in intensive care medicine. Revision 2015 (DAS-Guildeline 2015) – short version. German Medical Science: GMS e-journal. 13, (2015).
  64. O’Gara, B., Talmor, D. Lung protective properties of the volatile anesthetics. Intensive Care Medicine. 42 (9), 1487-1489 (2016).
  65. Murthy, S., Gomersall, C. D., Fowler, R. A. Care for critically ill patients with COVID-19. JAMA: The Journal of the American Medical Association. , (2020).
  66. Liao, X., Wang, B., Kang, Y. Novel coronavirus infection during the 2019-2020 epidemic: preparing intensive care units-the experience in Sichuan Province, China. Intensive Care Medicine. 46 (2), 357-360 (2020).
  67. Grasselli, G., Pesenti, A., Cecconi, M. Critical care utilization for the COVID-19 outbreak in Lombardy, Italy: Early experience and forecast during an emergency response. JAMA: The Journal of the American Medical Association. , (2020).
  68. Arentz, M., et al. Characteristics and outcomes of 21 critically ill patients with COVID-19 in Washington State. JAMA: The Journal of the American Medical Association. , (2020).
  69. Xu, Z., et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. The Lancet. Respiratory Medicine. 8 (4), 420-422 (2020).
  70. Ferrando, C., et al. but not propofol, reduces the lung inflammatory response and improves oxygenation in an acute respiratory distress syndrome model: a randomised laboratory study. European Journal of Anaesthesiology. 30 (8), 455-463 (2013).
  71. Voigtsberger, S., et al. Sevoflurane ameliorates gas exchange and attenuates lung damage in experimental lipopolysaccharide-induced lung injury. Anesthesiology. 111 (6), 1238-1248 (2009).
  72. Suter, D., et al. The immunomodulatory effect of sevoflurane in endotoxin-injured alveolar epithelial cells. Anesthesia and Analgesia. 104 (3), 638-645 (2007).
  73. Steurer, M., et al. The volatile anaesthetic sevoflurane attenuates lipopolysaccharide-induced injury in alveolar macrophages. Clinical and Experimental Immunology. 155 (2), 224-230 (2009).
  74. Blondonnet, R., et al. In vitro method to control concentrations of halogenated gases in cultured alveolar epithelial cells. Journal of Visualized Experiments. (144), e58554 (2018).
  75. Gargiulo, S., et al. Mice anesthesia, analgesia, and care, Part I: Anesthetic considerations in preclinical research. ILAR Journal / National Research Council, Institute of Laboratory Animal Resources. 53 (1), 55-69 (2012).
check_url/pt/61644?article_type=t

Play Video

Citar este artigo
Blondonnet, R., Paquette, B., Audard, J., Guler, R., Roman, F., Zhai, R., Belville, C., Blanchon, L., Godet, T., Futier, E., Bazin, J., Constantin, J., Sapin, V., Jabaudon, M. Halogenated Agent Delivery in Porcine Model of Acute Respiratory Distress Syndrome via an Intensive Care Unit Type Device. J. Vis. Exp. (163), e61644, doi:10.3791/61644 (2020).

View Video