151
|
Mane A, Isaac N. Synopsis of Clinical Acute Respiratory Distress Syndrome (ARDS). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:323-331. [PMID: 34019275 DOI: 10.1007/978-3-030-68748-9_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The entity of acute respiratory distress syndrome (ARDS) is an acute inflammatory lung condition associated with lung damage and increased vascular permeability. In the ICU, ARDS was reported to be the cause of 10.4% of admissions. The syndrome is associated with conditions such as sepsis, burns, trauma, and many others. The Berlin Definition which is the most up-to-date definition defines ARDS as an early onset of severe and refractory hypoxemia, PaO2/FiO2 ratio less than 300 mmHg, bilateral infiltrates on chest x-ray, and alveolar edema not explained by a cardiogenic cause or fluid overload.The entity of ARDS and its treatment have been studied for many years to better understand it and help find therapies. However, the mainstay of medical management is supportive with specific strategies for mechanical ventilation. No specific drug therapy is available at present.In this chapter, the history, clinical picture, and therapeutic approaches to ARDS will be discussed. We include lung-protective ventilation, prone positioning, use of neuromuscular blockade, corticosteroids, as well as discussion of studies done on this important clinical and morbid condition. We emphasize that there are ongoing trials and research being done to better identify patients earlier in their clinical course so that supportive care with lung-protective ventilation and a conservative fluid approach can be implemented. We also mention promising therapies such as cell-based therapies which would help in decreasing lung inflammation.
Collapse
Affiliation(s)
- Archana Mane
- Department of Anesthesiology, Albany Medical Center, Albany, NY, USA.
| | - Naldine Isaac
- Department of Anesthesiology, Albany Medical Center, Albany, NY, USA
| |
Collapse
|
152
|
Hol L, Nijbroek SGLH, Schultz MJ. Perioperative Lung Protection: Clinical Implications. Anesth Analg 2020; 131:1721-1729. [PMID: 33186160 DOI: 10.1213/ane.0000000000005187] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In the past, it was common practice to use a high tidal volume (VT) during intraoperative ventilation, because this reduced the need for high oxygen fractions to compensate for the ventilation-perfusion mismatches due to atelectasis in a time when it was uncommon to use positive end-expiratory pressure (PEEP) in the operating room. Convincing and increasing evidence for harm induced by ventilation with a high VT has emerged over recent decades, also in the operating room, and by now intraoperative ventilation with a low VT is a well-adopted approach. There is less certainty about the level of PEEP during intraoperative ventilation. Evidence for benefit and harm of higher PEEP during intraoperative ventilation is at least contradicting. While some PEEP may prevent lung injury through reduction of atelectasis, higher PEEP is undeniably associated with an increased risk of intraoperative hypotension that frequently requires administration of vasoactive drugs. The optimal level of inspired oxygen fraction (FIO2) during surgery is even more uncertain. The suggestion that hyperoxemia prevents against surgical site infections has not been confirmed in recent research. In addition, gas absorption-induced atelectasis and its association with adverse outcomes like postoperative pulmonary complications actually makes use of a high FIO2 less attractive. Based on the available evidence, we recommend the use of a low VT of 6-8 mL/kg predicted body weight in all surgery patients, and to restrict use of a high PEEP and high FIO2 during intraoperative ventilation to cases in which hypoxemia develops. Here, we prefer to first increase FIO2 before using high PEEP.
Collapse
Affiliation(s)
| | | | - Marcus J Schultz
- Department of Intensive Care.,Department of Intensive Care and Anesthesiology, Laboratory of Experimental Intensive Care and Anesthesiology (L·E·I·C·A), Amsterdam University Medical Centers, Location 'Amsterdam Medical Center', Amsterdam, the Netherlands.,Department of Intensive Care, Mahidol Oxford Tropical Medicine Research Unit (MORU), Mahidol University, Bangkok, Thailand.,Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
153
|
Del Castillo R, Martinez D, Sarria GJ, Pinillos L, Garcia B, Castillo L, Carhuactocto A, Giordano FA, Sarria GR. Low-dose radiotherapy for COVID-19 pneumonia treatment: case report, procedure, and literature review. Strahlenther Onkol 2020; 196:1086-1093. [PMID: 32816059 PMCID: PMC7439803 DOI: 10.1007/s00066-020-01675-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 07/27/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND The COVID-19 pandemic outbreak has set the emergency services in developing countries on major alert, as the installed response capacities are easily overwhelmed by the constantly increasing high demand. The deficit of intensive care unit beds and ventilators in countries like Peru is forcing practitioners to seek preventive or early interventional strategies to prevent saturating these chronically neglected facilities. CASE PRESENTATION A 64-year-old patient is reported after presenting with COVID-19 pneumonia and rapidly progressing to deteriorated ventilatory function. Compassionate treatment with a single 1‑Gy dose to the bilateral whole-lung volume was administered, with gradual daily improvement of ventilatory function and decrease in serum inflammatory markers and oxygen support needs, including intubation. No treatment-related toxicity developed. Procedures of transport, disinfection, and treatment planning and delivery are described. CONCLUSION Whole-lung low-dose radiotherapy seems to be a promising approach for avoiding or delaying invasive respiratory support. Delivered low doses are far from meeting toxicity ranges. On-going prospective trials will elucidate the effectiveness of this approach.
Collapse
Affiliation(s)
| | - David Martinez
- Department of Radiation Oncology, Clinica Delgado-AUNA, Lima, Peru
| | - Gustavo J Sarria
- Department of Radiation Oncology, Clinica Delgado-AUNA, Lima, Peru
| | - Luis Pinillos
- Department of Radiation Oncology, Clinica Delgado-AUNA, Lima, Peru
| | - Bertha Garcia
- Department of Radiation Oncology, Clinica Delgado-AUNA, Lima, Peru
| | - Luis Castillo
- Department of Critical Care, Clinica Delgado-AUNA, Lima, Peru
| | | | - Frank A Giordano
- Department of Radiation Oncology, University Hospital Bonn, Universitätsklinikum Bonn, Venusberg Campus 1, Building 55, 53127, Bonn, Germany
| | - Gustavo R Sarria
- Department of Radiation Oncology, University Hospital Bonn, Universitätsklinikum Bonn, Venusberg Campus 1, Building 55, 53127, Bonn, Germany.
| |
Collapse
|
154
|
Abstract
PURPOSE OF REVIEW Most clinical trials of lung-protective ventilation have tested one-size-fits-all strategies with mixed results. Data are lacking on how best to tailor mechanical ventilation to patient-specific risk of lung injury. RECENT FINDINGS Risk of ventilation-induced lung injury is determined by biological predisposition to biophysical lung injury and physical mechanical perturbations that concentrate stress and strain regionally within the lung. Recent investigations have identified molecular subphenotypes classified as hyperinflammatory and hypoinflammatory acute respiratory distress syndrome (ARDS), which may have dissimilar risk for ventilation-induced lung injury. Mechanically, gravity-dependent atelectasis has long been recognized to decrease total aerated lung volume available for tidal ventilation, a concept termed the 'ARDS baby lung'. Recent studies have demonstrated that the aerated baby lung also has nonuniform stress/strain distribution, with potentially injurious forces concentrated in zones of heterogeneity where aerated alveoli are adjacent to flooded or atelectatic alveoli. The preponderance of evidence also indicates that current standard-of-care tidal volume management is not universally protective in ARDS. When considering escalation of lung-protective interventions, potential benefits of the intervention should be weighed against tradeoffs of accompanying cointerventions required, for example, deeper sedation or neuromuscular blockade. A precision medicine approach to lung-protection would weigh. SUMMARY A precision medicine approach to lung-protective ventilation requires weighing four key factors in each patient: biological predisposition to biophysical lung injury, mechanical predisposition to biophysical injury accounting for spatial mechanical heterogeneity within the lung, anticipated benefits of escalating lung-protective interventions, and potential unintended adverse effects of mandatory cointerventions.
Collapse
|
155
|
Lim SY, Cho YJ, Kim DJ, Kim JS, Jheon S, Chung JH, Lee JH. Effects of Ultralow-Tidal-Volume Ventilation under Veno-Venous Extracorporeal Membrane Oxygenation in a Porcine Model with Ventilator-Induced Lung Injury. MEMBRANES 2020; 10:membranes10120379. [PMID: 33260539 PMCID: PMC7761070 DOI: 10.3390/membranes10120379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022]
Abstract
Low-tidal-volume ventilation decreases mortality in acute respiratory distress syndrome (ARDS) patients. This study investigated the effects of ultralow tidal ventilation under veno-venous extracorporeal membrane oxygenator (ECMO) support in pigs with ARDS. Eight pigs were intubated and inoculated with methicillin-resistant Staphylococcus aureus through bronchoscopy. Ultralow tidal ventilation (3 mL/kg) under extracorporeal membrane oxygenator (ECMO) support was applied to one group and high tidal ventilation (15 mL/kg) was applied to another group to maintain comparable oxygenation for 12 h without ECMO support. Each group had similar arterial blood gas values and hemodynamic variables at baseline and during the experiment. The high-tidal-volume ventilation group showed a gradual decline in arterial oxygen levels, and repeated ANOVA showed significant differences in oxygenation change over time in the ultralow tidal ventilation group. Inflammatory cytokine levels in the bronchoalveolar lavage fluid and lung ultrasound scores were similar between two groups. Histologic analysis showed that both groups developed pneumonia after 12 h; however, the ultralow tidal ventilation group had a lower lung injury score assessed by the pathologist. We developed the first ultralow-tidal-volume ventilation porcine model under veno-venous ECMO support. The ultralow-tidal-volume ventilation strategy can mitigate mechanical ventilator-associated lung injury.
Collapse
Affiliation(s)
- Sung Yoon Lim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (S.Y.L.); (Y.-J.C.)
| | - Young-Jae Cho
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (S.Y.L.); (Y.-J.C.)
| | - Dong Jung Kim
- Department of Cardiovascular and Thoracic Surgery, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (D.J.K.); (J.S.K.); (S.J.)
| | - Jun Sung Kim
- Department of Cardiovascular and Thoracic Surgery, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (D.J.K.); (J.S.K.); (S.J.)
| | - Sanghoon Jheon
- Department of Cardiovascular and Thoracic Surgery, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (D.J.K.); (J.S.K.); (S.J.)
| | - Jin Haeng Chung
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam 13620, Korea;
| | - Jae Ho Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (S.Y.L.); (Y.-J.C.)
- Correspondence: ; Tel.: +82-31-787-7011; Fax: +82-31-787-4050
| |
Collapse
|
156
|
Spraider P, Martini J, Abram J, Putzer G, Glodny B, Hell T, Barnes T, Enk D. Individualized flow-controlled ventilation compared to best clinical practice pressure-controlled ventilation: a prospective randomized porcine study. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:662. [PMID: 33239039 PMCID: PMC7686826 DOI: 10.1186/s13054-020-03325-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/04/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Flow-controlled ventilation is a novel ventilation method which allows to individualize ventilation according to dynamic lung mechanic limits based on direct tracheal pressure measurement at a stable constant gas flow during inspiration and expiration. The aim of this porcine study was to compare individualized flow-controlled ventilation (FCV) and current guideline-conform pressure-controlled ventilation (PCV) in long-term ventilation. METHODS Anesthetized pigs were ventilated with either FCV or PCV over a period of 10 h with a fixed FiO2 of 0.3. FCV settings were individualized by compliance-guided positive end-expiratory pressure (PEEP) and peak pressure (Ppeak) titration. Flow was adjusted to maintain normocapnia and the inspiration to expiration ratio (I:E ratio) was set at 1:1. PCV was performed with a PEEP of 5 cm H2O and Ppeak was set to achieve a tidal volume (VT) of 7 ml/kg. The respiratory rate was adjusted to maintain normocapnia and the I:E ratio was set at 1:1.5. Repeated measurements during observation period were assessed by linear mixed-effects model. RESULTS In FCV (n = 6), respiratory minute volume was significantly reduced (6.0 vs 12.7, MD - 6.8 (- 8.2 to - 5.4) l/min; p < 0.001) as compared to PCV (n = 6). Oxygenation was improved in the FCV group (paO2 119.8 vs 96.6, MD 23.2 (9.0 to 37.5) Torr; 15.97 vs 12.87, MD 3.10 (1.19 to 5.00) kPa; p = 0.010) and CO2 removal was more efficient (paCO2 40.1 vs 44.9, MD - 4.7 (- 7.4 to - 2.0) Torr; 5.35 vs 5.98, MD - 0.63 (- 0.99 to - 0.27) kPa; p = 0.006). Ppeak and driving pressure were comparable in both groups, whereas PEEP was significantly lower in FCV (p = 0.002). Computed tomography revealed a significant reduction in non-aerated lung tissue in individualized FCV (p = 0.026) and no significant difference in overdistended lung tissue, although a significantly higher VT was applied (8.2 vs 7.6, MD 0.7 (0.2 to 1.2) ml/kg; p = 0.025). CONCLUSION Our long-term ventilation study demonstrates the applicability of a compliance-guided individualization of FCV settings, which resulted in significantly improved gas exchange and lung tissue aeration without signs of overinflation as compared to best clinical practice PCV.
Collapse
Affiliation(s)
- Patrick Spraider
- Department of Anaesthesia and Intensive Care Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Judith Martini
- Department of Anaesthesia and Intensive Care Medicine, Medical University of Innsbruck, Innsbruck, Austria.
| | - Julia Abram
- Department of Anaesthesia and Intensive Care Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Gabriel Putzer
- Department of Anaesthesia and Intensive Care Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Bernhard Glodny
- Department of Radiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Tobias Hell
- Department of Mathematics, Faculty of Mathematics, Computer Science and Physics, University of Innsbruck, Innsbruck, Austria
| | | | - Dietmar Enk
- Faculty of Medicine, University of Münster, Münster, Germany
| |
Collapse
|
157
|
Ordies S, Orlitova M, Heigl T, Sacreas A, Van Herck A, Kaes J, Saez B, Vanstapel A, Ceulemans L, Vanaudenaerde BM, Vos R, Verschakelen J, Verleden GM, Verleden SE, Van Raemdonck DE, Neyrinck AP. Flow-controlled ventilation during EVLP improves oxygenation and preserves alveolar recruitment. Intensive Care Med Exp 2020; 8:70. [PMID: 33237343 PMCID: PMC7686942 DOI: 10.1186/s40635-020-00360-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 11/17/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ex vivo lung perfusion (EVLP) is a widespread accepted platform for preservation and evaluation of donor lungs prior to lung transplantation (LTx). Standard lungs are ventilated using volume-controlled ventilation (VCV). We investigated the effects of flow-controlled ventilation (FCV) in a large animal EVLP model. Fourteen porcine lungs were mounted on EVLP after a warm ischemic interval of 2 h and randomized in two groups (n = 7/group). In VCV, 7 grafts were conventionally ventilated and in FCV, 7 grafts were ventilated by flow-controlled ventilation. EVLP physiologic parameters (compliance, pulmonary vascular resistance and oxygenation) were recorded hourly. After 6 h of EVLP, broncho-alveolar lavage (BAL) was performed and biopsies for wet-to-dry weight (W/D) ratio and histology were taken. The left lung was inflated, frozen in liquid nitrogen vapors and scanned with computed tomography (CT) to assess regional distribution of Hounsfield units (HU). RESULTS All lungs endured 6 h of EVLP. Oxygenation was better in FCV compared to VCV (p = 0.01) and the decrease in lung compliance was less in FCV (p = 0.03). W/D ratio, pathology and BAL samples did not differ between both groups (p = 0.16, p = 0.55 and p = 0.62). Overall, CT densities tended to be less pronounced in FCV (p = 0.05). Distribution of CT densities revealed a higher proportion of well-aerated lung parts in FCV compared to VCV (p = 0.01). CONCLUSIONS FCV in pulmonary grafts mounted on EVLP is feasible and leads to improved oxygenation and alveolar recruitment. This ventilation strategy might prolong EVLP over time, with less risk for volutrauma and atelectrauma.
Collapse
Affiliation(s)
- Sofie Ordies
- Unit of Anesthesiology and Algology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Anesthesiology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.,Leuven Lung Transplant Group, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Michaela Orlitova
- Unit of Anesthesiology and Algology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven, Leuven, Belgium.,Leuven Lung Transplant Group, Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Tobias Heigl
- Leuven Lung Transplant Group, Katholieke Universiteit Leuven, Leuven, Belgium.,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Annelore Sacreas
- Leuven Lung Transplant Group, Katholieke Universiteit Leuven, Leuven, Belgium.,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Anke Van Herck
- Leuven Lung Transplant Group, Katholieke Universiteit Leuven, Leuven, Belgium.,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Pneumology, University Hospitals Leuven, Leuven, Belgium
| | - Janne Kaes
- Leuven Lung Transplant Group, Katholieke Universiteit Leuven, Leuven, Belgium.,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Berta Saez
- Leuven Lung Transplant Group, Katholieke Universiteit Leuven, Leuven, Belgium.,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Pneumology, University Hospitals Leuven, Leuven, Belgium
| | - Arno Vanstapel
- Leuven Lung Transplant Group, Katholieke Universiteit Leuven, Leuven, Belgium.,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Laurens Ceulemans
- Leuven Lung Transplant Group, Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium.,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Bart M Vanaudenaerde
- Leuven Lung Transplant Group, Katholieke Universiteit Leuven, Leuven, Belgium.,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Robin Vos
- Leuven Lung Transplant Group, Katholieke Universiteit Leuven, Leuven, Belgium.,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Pneumology, University Hospitals Leuven, Leuven, Belgium
| | | | - Geert M Verleden
- Leuven Lung Transplant Group, Katholieke Universiteit Leuven, Leuven, Belgium.,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Pneumology, University Hospitals Leuven, Leuven, Belgium
| | - Stijn E Verleden
- Leuven Lung Transplant Group, Katholieke Universiteit Leuven, Leuven, Belgium.,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Dirk E Van Raemdonck
- Leuven Lung Transplant Group, Katholieke Universiteit Leuven, Leuven, Belgium.,Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium.,Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), Katholieke Universiteit Leuven, Leuven, Belgium
| | - Arne P Neyrinck
- Unit of Anesthesiology and Algology, Department of Cardiovascular Sciences, Katholieke Universiteit Leuven, Leuven, Belgium. .,Department of Anesthesiology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium. .,Leuven Lung Transplant Group, Katholieke Universiteit Leuven, Leuven, Belgium.
| |
Collapse
|
158
|
Death-associated Protein Kinase 1 Mediates Ventilator-induced Lung Injury in Mice by Promoting Alveolar Epithelial Cell Apoptosis. Anesthesiology 2020; 133:905-918. [PMID: 32930731 DOI: 10.1097/aln.0000000000003464] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
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
Collapse
|
159
|
Baumann P, Wiegert S, Greco F, Ersch J, Cannizzaro V. Strain-specific differences in lung tissue viscoelasticity of mechanically ventilated infant Sprague-Dawley and Wistar rats. Am J Physiol Lung Cell Mol Physiol 2020; 320:L220-L231. [PMID: 33207919 DOI: 10.1152/ajplung.00100.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Rats are often used in ventilator-induced lung injury (VILI) models. However, strain-specific susceptibility for VILI has not been elucidated yet. The aim of this study was to demonstrate strain-specific differences in VILI in infant Sprague-Dawley and Wistar rats. VILI was compared in 2-wk-old pups after 8 h of protective or injurious ventilation. Pups were ventilated with tidal volumes (VT) of ∼7 mL/kg and positive end-expiratory pressures (PEEP) of 6 cmH2O (VT7 PEEP6) or with VT of ∼21 mL/kg and PEEP 2 cmH2O (VT21 PEEP2). Interleukin-6, macrophage inflammatory protein-2 (MIP-2), inflammatory cells, and albumin in bronchoalveolar lavage fluid (BALF); histology; and low-frequency forced oscillation technique (LFOT) and pressure-volume (PV) maneuvers were assessed. Alveolar macrophages, neutrophils, and MIP-2 derived from BALF revealed more pronounced VILI after VT21 PEEP2 in both strains. LFOT and PV analyses demonstrated rat strain-specific differences both at baseline and particularly in response to VT21 PEEP2 ventilation. Sprague-Dawley rats showed higher airway and tissue resistance and elastance values with no difference in hysteresivity between ventilation strategies. Wister rats challenged by VT21 PEEP2 experienced significantly more energy dissipation when compared with VT7 PEEP6 ventilation. In conclusion, both rat strains are useful for VILI models. The degree of VILI severity depends on ventilation strategy and selected strain. However, fundamental and time-dependent differences in respiratory system mechanics exist and reflect different lung tissue viscoelasticity. Hence, strain-specific characteristics of the respiratory system need to be considered when planning and interpreting VILI studies with infant rats.
Collapse
Affiliation(s)
- Philipp Baumann
- Department of Intensive Care Medicine and Neonatology, University Children's Hospital Zurich, Zurich, Switzerland.,Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Susanne Wiegert
- Department of Intensive Care Medicine and Neonatology, University Children's Hospital Zurich, Zurich, Switzerland.,Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Francesco Greco
- Department of Intensive Care Medicine and Neonatology, University Children's Hospital Zurich, Zurich, Switzerland.,Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Joerg Ersch
- Department of Intensive Care Medicine and Neonatology, University Children's Hospital Zurich, Zurich, Switzerland.,Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Vincenzo Cannizzaro
- Department of Intensive Care Medicine and Neonatology, University Children's Hospital Zurich, Zurich, Switzerland.,Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland.,Department of Neonatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
160
|
Lian J, Lin J, Zakaria N, Yahaya BH. Acute Lung Injury: Disease Modelling and the Therapeutic Potential of Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1298:149-166. [PMID: 32424492 DOI: 10.1007/5584_2020_538] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acute lung injury (ALI) is a severe clinical condition with high morbidity and mortality that usually results in the development of multiple organ dysfunction. The complex pathophysiology of ALI seems to provide a wide range of targets that offer numerous therapeutic options. However, despite extensive studies of ALI pathophysiology and treatment, no effective pharmacotherapy is available. Increasing evidence from both preclinical and clinical studies supports the preventive and therapeutic effects of mesenchymal stem cells (MSCs) for treating ALI. As cell-based therapy poses the risk of occlusion in microvasculature or unregulated growth, MSC-derived extracellular vesicles (MSC-EVs) have been extensively studied as a new therapeutic strategy for non-cell based therapy. It is widely accepted that the therapeutic properties of MSCs are derived from soluble factors with paracrine or endocrine effects, and EVs are among the most important paracrine or endocrine vehicles that can deliver various soluble factors with a similar phenotype as the parent cell. Therapeutic effects of MSCs have been reported for various delivery approaches, diverse doses, multiple origins, and different times of administration, and MSC-EVs treatment may include but is not limited to these choices. The mechanisms by which MSCs and MSC-EVs may contribute to ALI treatment remain elusive and need further exploration. This review provides an overview of preclinical studies that support the application of MSC-EVs for treating ALI, and it discusses emerging opportunities and their associated challenges.
Collapse
Affiliation(s)
- Jie Lian
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, SAINS@Bertam, Penang, Malaysia.,Stem Cell and Biotherapy Technology Research Center of Henan Province, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Juntang Lin
- Stem Cell and Biotherapy Technology Research Center of Henan Province, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Norashikin Zakaria
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, SAINS@Bertam, Penang, Malaysia
| | - Badrul Hisham Yahaya
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, SAINS@Bertam, Penang, Malaysia.
| |
Collapse
|
161
|
Kuzkov VV, Lapin KS, Fot EV, Kirov MY. Ventilator-associated lung injury in the intensive care unit and operating room – what's new? MESSENGER OF ANESTHESIOLOGY AND RESUSCITATION 2020. [DOI: 10.21292/2078-5658-2020-17-5-47-61] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The prophylaxis of ventilator-associated lung injury (VALI) and postoperative pulmonary complications (PPC) is of utmost importance to reduce complications both in the perioperative period of major surgery and in the intensive care unit (ICU).Protective approach to mechanical ventilation comprises a wide range of measures reducing the damage of the lung tissue associated with the stress and strain phenomena. The implementation of the strategy of high positive end-expiratory pressure (PEEP) in combination with alveolar recruitment maneuver has numerous limitations and requires further personalized approaches.When lung injury is self-induced by a patient, it becomes an important contributor to VALI and should be timely diagnosed and prevented both before initiation of mechanical support and during the restoration of spontaneous breathing. This review highlights the key mechanisms of VALI and current understanding of protective ventilation. The concept of damaging energy as well as approaches to the personalized optimization of respiratory settings are discussed in detail. Particular attention is paid to the prognostication of the risk factors of VALI and PPC.
Collapse
Affiliation(s)
- V. V. Kuzkov
- Northern State Medical University; Severodvinsk Municipal Clinical Emergency Hospital no. 2
| | - K. S. Lapin
- Northern State Medical University; Severodvinsk Municipal Clinical Emergency Hospital no. 2
| | | | | |
Collapse
|
162
|
Chao KY, Lin YW, Chiang CE, Tseng CW, Mu SC. Sustained inflation: The lung recruitment maneuvers for neonates. Paediatr Respir Rev 2020; 36:142-150. [PMID: 32386887 DOI: 10.1016/j.prrv.2019.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/11/2019] [Accepted: 09/07/2019] [Indexed: 10/25/2022]
Abstract
Establishing effective respiration is vital in the transition from fetal to neonatal life. Respiratory support mainly facilitates and creates functional residual capacity and maintains adequate gas exchange. Sustained inflation (SI) delivers prolonged inflation and rapidly creates and establishes the functional residual capacity. The use of SI in preterm infants in the delivery room is still controversial. The optimum settings of SI remain unknown. Animal studies and clinical reports have demonstrated the advantages and disadvantages of SI. In this article, the current literature was reviewed to examine the efficacy of SI in infants.
Collapse
Affiliation(s)
- Ke-Yun Chao
- Department of Respiratory Therapy, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan; School of Physical Therapy, Graduate Institute of Rehabilitation Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Wen Lin
- Department of Nursing, Fu Jen Catholic University Hospital, Fu Jen University, New Taipei City, Taiwan
| | - Chen-En Chiang
- Department of Respiratory Therapy, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Chi-Wei Tseng
- Department of Respiratory Therapy, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Shu-Chi Mu
- Department of Pediatrics, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan; Medical College, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
163
|
Sattari S, Mariano CA, Vittalbabu S, Velazquez JV, Postma J, Horst C, Teh E, Nordgren TM, Eskandari M. Introducing a Custom-Designed Volume-Pressure Machine for Novel Measurements of Whole Lung Organ Viscoelasticity and Direct Comparisons Between Positive- and Negative-Pressure Ventilation. Front Bioeng Biotechnol 2020; 8:578762. [PMID: 33195138 PMCID: PMC7643401 DOI: 10.3389/fbioe.2020.578762] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
Abstract
Asthma, emphysema, COVID-19 and other lung-impacting diseases cause the remodeling of tissue structural properties and can lead to changes in conducting pulmonary volume, viscoelasticity, and air flow distribution. Whole organ experimental inflation tests are commonly used to understand the impact of these modifications on lung mechanics. Here we introduce a novel, automated, custom-designed device for measuring the volume and pressure response of lungs, surpassing the capabilities of traditional machines and built to range size-scales to accommodate both murine and porcine tests. The software-controlled system is capable of constructing standardized continuous volume-pressure curves, while accounting for air compressibility, yielding consistent and reproducible measures while eliminating the need for pulmonary degassing. This device uses volume-control to enable viscoelastic whole lung macromechanical insights from rate dependencies and pressure-time curves. Moreover, the conceptual design of this device facilitates studies relating the phenomenon of diaphragm breathing and artificial ventilation induced by pushing air inside the lungs. System capabilities are demonstrated and validated via a comparative study between ex vivo murine lungs and elastic balloons, using various testing protocols. Volume-pressure curve comparisons with previous pressure-controlled systems yield good agreement, confirming accuracy. This work expands the capabilities of current lung experiments, improving scientific investigations of healthy and diseased pulmonary biomechanics. Ultimately, the methodologies demonstrated in the manufacturing of this system enable future studies centered on investigating viscoelasticity as a potential biomarker and improvements to patient ventilators based on direct assessment and comparisons of positive- and negative-pressure mechanics.
Collapse
Affiliation(s)
- Samaneh Sattari
- Department of Mechanical Engineering, University of California, Riverside, Riverside, CA, United States
| | - Crystal A Mariano
- Department of Mechanical Engineering, University of California, Riverside, Riverside, CA, United States
| | - Swathi Vittalbabu
- Department of Mechanical Engineering, University of California, Riverside, Riverside, CA, United States
| | - Jalene V Velazquez
- BREATHE Center at the School of Medicine, University of California, Riverside, Riverside, CA, United States.,Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | | | - Caleb Horst
- CellScale Biomaterials Testing, Waterloo, ON, Canada
| | - Eric Teh
- CellScale Biomaterials Testing, Waterloo, ON, Canada
| | - Tara M Nordgren
- BREATHE Center at the School of Medicine, University of California, Riverside, Riverside, CA, United States.,Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Mona Eskandari
- Department of Mechanical Engineering, University of California, Riverside, Riverside, CA, United States.,BREATHE Center at the School of Medicine, University of California, Riverside, Riverside, CA, United States.,Department of Bioengineering, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
164
|
Montrief T, Ramzy M, Long B, Gottlieb M, Hercz D. COVID-19 respiratory support in the emergency department setting. Am J Emerg Med 2020; 38:2160-2168. [PMID: 33046288 PMCID: PMC7413866 DOI: 10.1016/j.ajem.2020.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/26/2020] [Accepted: 08/01/2020] [Indexed: 01/25/2023] Open
Abstract
Introduction Severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2), which causes the coronavirus disease 2019 (COVID-19), may result in severe complications, multiorgan dysfunction, acute respiratory failure, and death. SARS-CoV-2 is highly contagious and places healthcare workers at significant risk, especially during aerosol-generating procedures, including airway management. Objective This narrative review outlines the underlying respiratory pathophysiology of patients with COVID-19 and discusses approaches to airway management in the emergency department (ED) based on current literature. Discussion Patients presenting with SARS-CoV-2 infection are at high risk for acute respiratory failure requiring airway management. Among hospitalized patients, 10–20% require intensive care unit admission, and 3–10% require intubation and mechanical ventilation. While providing respiratory support for these patients, proper infection control measures, including adherence to personal protective equipment policies, are necessary to prevent nosocomial transmission to healthcare workers. A structured approach to respiratory failure in these patients includes the use of exogenous oxygen via nasal cannula or non-rebreather, as well as titrated high-flow nasal cannula and non-invasive ventilation. This review offers several guiding principles and resources designed to be adapted in conjunction with local workplace policies for patients requiring respiratory support. Conclusions While the fundamental principles of acute respiratory failure management are similar between COVID-19 and non-COVID-19 patients, there are some notable differences, including a focus on provider safety. This review provides an approach to airway management and respiratory support in the patient with COVID-19.
Collapse
Affiliation(s)
- Tim Montrief
- Department of Emergency Medicine, Jackson Memorial Health System, Miami, Florida, United States of America
| | - Mark Ramzy
- Department of Emergency Medicine, Maimonides Medical Center, Brooklyn, NY, United States of America
| | - Brit Long
- Department of Emergency Medicine, Brooke Army Medical Center, San Antonio, TX, United States of America.
| | - Michael Gottlieb
- Department of Emergency Medicine, Rush University Medical Center, Chicago, IL, United States of America
| | - Dan Hercz
- Department of Emergency Medicine, Jackson Memorial Hospital, Miami, FL, United States of America
| |
Collapse
|
165
|
An evaluation of manual tidal volume and respiratory rate delivery during simulated resuscitation. Am J Emerg Med 2020; 45:446-450. [PMID: 33077312 DOI: 10.1016/j.ajem.2020.09.091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 11/20/2022] Open
Abstract
INTRODUCTION Excessive minute ventilation during cardiac arrest may cause lung injury and decrease the effectiveness of cardiopulmonary resuscitation (CPR). However, little is known about how clinicians deliver tidal volumes and respiratory rates during CPR. METHODS In this cross-sectional study, licensed practitioners attending an American Heart Association (AHA) Advanced Cardiac Life Support (ACLS) course performed CPR and manual ventilation on a high-fidelity simulator during the megacode portion of the course. Delivered tidal volumes and respiratory rates were measured on a monitor. During the first scenario, results were not displayed to participants, but were displayed during the second scenario. RESULTS Fifty-two clinicians participated in this study. Average height was 169 (157,178) cm. Pre-monitor display tidal volumes delivered were larger in male participants compared to female participants (684.6 ± 134.4 vs 586.7 ± 167.6 ml, P = 0.05). Those using medium-sized gloves delivered smaller tidal volumes than those using small or large gloves. Twenty-two (42.3%) delivered tidal volume in the range of 5-8 ml/kg of predicted body weight for the simulation manikin, and 35 (67.3%) delivered tidal volumes with >20% variability among breaths. All participants met the target respiratory rate around 10 breaths/min. CONCLUSION Tidal volume delivery varied greatly during manual ventilation and fewer than half participants delivered tidal volume at 5-8 ml/kg to the manikin. Sex and glove size appeared to impact tidal volume delivery when the participants were unaware of what they were delivering. Participants were able to meet the target respiratory rate around 10 without audio or visual feedback.
Collapse
|
166
|
Asim M, Amin F, El-Menyar A. Multiple organ dysfunction syndrome: Contemporary insights on the clinicopathological spectrum. Qatar Med J 2020; 2020:22. [PMID: 33628712 PMCID: PMC7884906 DOI: 10.5339/qmj.2020.22] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 03/03/2020] [Indexed: 12/27/2022] Open
Abstract
Multiorgan dysfunction syndrome (MODS) remains a major complication and challenge to treat patients with critical illness in different intensive care unit settings. The exact mechanism and pathophysiology of MODS is complex and remains unexplored. We reviewed the literature from January 2011 to August 2019 to analyze the underlying mechanisms, prognostic factors, MODS scoring systems, organ systems dysfunctions, and the management of MODS. We used the search engines PubMed, MEDLINE, Scopus, and Google Scholar with the keywords "multiple organ dysfunction syndrome," "intensive care units," "multiorgan failure," "MODS scoring system," and "MODS management." The initial search yielded 3550 abstracts, of which 91 articles were relevant to the scope of the present article. A better understanding of a disease course will help differentiate the signs of an intense inflammatory response from the early onset of sepsis and minimize the inappropriate use of medications. This, in turn, will promote organtargeted therapy and prevent occurrence and progression of MODS.
Collapse
Affiliation(s)
- Mohammad Asim
- Department of Surgery, Clinical Research, Trauma Surgery Section, Hamad General Hospital, Doha, Qatar
| | - Farhana Amin
- Sri Ramaswamy Memorial Medical College Hospital & Research Center, Tamil Nadu, India
| | | |
Collapse
|
167
|
Morrell ED, Grazioli S, Hung C, Kajikawa O, Kosamo S, Stapleton RD, Gharib SA, Amado-Rodríguez L, Albaiceta G, Wurfel MM, Matute-Bello G. Alveolar CCN1 is associated with mechanical stretch and acute respiratory distress syndrome severity. Am J Physiol Lung Cell Mol Physiol 2020; 319:L825-L832. [PMID: 32936024 DOI: 10.1152/ajplung.00073.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The cellular communication network factor 1 (CCN1) is a matricellular protein that can modulate multiple tissue responses, including inflammation and repair. We have previously shown that adenoviral overexpression of Ccn1 is sufficient to cause acute lung injury in mice. We hypothesized that CCN1 is present in the airspaces of lungs during the acute phase of lung injury, and higher concentrations are associated with acute respiratory distress syndrome (ARDS) severity. We tested this hypothesis by measuring 1) CCN1 in bronchoalveolar lavage fluid (BALF) and lung homogenates from mice subjected to ventilation-induced lung injury (VILI), 2) Ccn1 gene expression and protein levels in MLE-12 cells (alveolar epithelial cell line) subjected to mechanical stretch, and 3) CCN1 in BALF from mechanically ventilated humans with and without ARDS. BALF CCN1 concentrations and whole lung CCN1 protein levels were significantly increased in mice with VILI (n = 6) versus noninjured controls (n = 6). Ccn1 gene expression and CCN1 protein levels were increased in MLE-12 cells cultured under stretch conditions. Subjects with ARDS (n = 77) had higher BALF CCN1 levels compared with mechanically ventilated subjects without ARDS (n = 45) (P < 0.05). In subjects with ARDS, BALF CCN1 concentrations were associated with higher total protein, sRAGE, and worse [Formula: see text]/[Formula: see text] ratios (all P < 0.05). CCN1 is present in the lungs of mice and humans during the acute inflammatory phase of lung injury, and concentrations are higher in patients with increased markers of severity. Alveolar epithelial cells may be an important source of CCN1 under mechanical stretch conditions.
Collapse
Affiliation(s)
- Eric D Morrell
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, Washington.,Veterans Affairs Puget Sound Healthcare System, Seattle, Washington
| | - Serge Grazioli
- Division of Neonatal and Pediatric Intensive Care, Department of Pediatrics, University Hospitals of Geneva, Geneva, Switzerland
| | - Chi Hung
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, Washington
| | - Osamu Kajikawa
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, Washington
| | - Susanna Kosamo
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, Washington
| | | | - Sina A Gharib
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, Washington
| | - Laura Amado-Rodríguez
- Centro de Investigación Biomédica En Red-Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Cardiac Intensive Care Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Guillermo Albaiceta
- Centro de Investigación Biomédica En Red-Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Cardiac Intensive Care Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Mark M Wurfel
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, Washington
| | - Gustavo Matute-Bello
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, Washington.,Veterans Affairs Puget Sound Healthcare System, Seattle, Washington
| |
Collapse
|
168
|
Jones E, Gould A, Pillay TD, Khorasanee R, Sykes R, Bazo-Alvarez JC, Cox C, Shurovi B, Isted A, Simpson T, Jennings M, Breeze R, Khaliq W. Subcutaneous Emphysema, Pneumomediastinum, and Pneumothorax in Critically Ill Patients With Coronavirus Disease 2019: A Retrospective Cohort Study. Crit Care Explor 2020; 2:e0210. [PMID: 33063043 PMCID: PMC7515614 DOI: 10.1097/cce.0000000000000210] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
IMPORTANCE Management of severe coronavirus disease 2019 relies on advanced respiratory support modalities including invasive mechanical ventilation, continuous positive airway pressure, and noninvasive ventilation, all of which are associated with the development of subcutaneous emphysema, pneumomediastinum, and pneumothorax (herein collectively termed barotrauma). OBJECTIVES To assess the occurrence rate of barotrauma in severe coronavirus disease 2019 and to explore possible associated factors. DESIGN SETTING AND PARTICIPANTS A retrospective, single-center cohort study with nested case series, conducted at University Hospital Lewisham: a 450-bed general hospital in London, United Kingdom. All patients with confirmed coronavirus disease 2019 admitted to the critical care department from March 12, to April 12, 2020, were included. MAIN OUTCOMES AND MEASURES Patients were retrospectively screened for radiological evidence of barotrauma. Admission characteristics, modalities of respiratory support, and outcomes were compared between barotrauma and nonbarotrauma groups. Respiratory parameters in the period preceding barotrauma identification were recorded. RESULTS Of 83 admissions with coronavirus disease 2019, eight suffered barotrauma (occurrence rate 9.6%; 95% CI 4.3%-18.1%). Barotrauma cases had longer illness duration prior to critical care admission (10 vs 7 d; interquartile range, 8-14 and 6-10, respectively; p = 0.073) and were more often treated with continuous positive airway pressure or noninvasive ventilation as the initial modality of advanced respiratory support (87.5% vs 36.0%; p = 0.007). Patients managed with continuous positive airway pressure or noninvasive ventilation prior to the development of barotrauma had median minute ventilation of 16.2-19.9 and 21.3-22.7 L/min, respectively. Compared with the nonbarotrauma group, a higher proportion of patients with barotrauma had died (62.5% vs 43.2%), and a lower proportion of patients had been discharged (25.0% vs 53.3%) at 3-month follow-up. CONCLUSIONS AND RELEVANCE Barotrauma appears to be a common complication of severe coronavirus disease 2019. Determining whether high minute ventilation while using continuous positive airway pressure or noninvasive ventilation predisposes patients to barotrauma requires further investigation.
Collapse
Affiliation(s)
- Eben Jones
- Department of Critical Care, University Hospital Lewisham, London, United Kingdom
| | - Allon Gould
- Department of General Medicine, University Hospital Lewisham, London, United Kingdom
| | - Timesh D Pillay
- Department of Respiratory Medicine, St Mary's Hospital, Imperial College London, London, United Kingdom
| | - Reza Khorasanee
- Department of Critical Care, University Hospital Lewisham, London, United Kingdom
| | - Richard Sykes
- Department of Critical Care, University Hospital Lewisham, London, United Kingdom
| | - Juan Carlos Bazo-Alvarez
- Instituto de Investigación (IIU), Universidad Católica Los Angeles de Chimbote, Chimbote, Perú
- Research Department of Primary Care and Population Health, University College London (UCL), London, United Kingdom
| | - Charlie Cox
- Department of Critical Care, University Hospital Lewisham, London, United Kingdom
| | - Badrun Shurovi
- Department of Critical Care, University Hospital Lewisham, London, United Kingdom
| | - Alexander Isted
- Department of Critical Care, University Hospital Lewisham, London, United Kingdom
| | - Thomas Simpson
- Department of Respiratory Medicine, University Hospital Lewisham, London, United Kingdom
| | - Mick Jennings
- Department of Critical Care, University Hospital Lewisham, London, United Kingdom
| | - Richard Breeze
- Department of Critical Care, University Hospital Lewisham, London, United Kingdom
| | - Waqas Khaliq
- Department of Critical Care, University Hospital Lewisham, London, United Kingdom
| |
Collapse
|
169
|
Vayvada M, Uygun Y, Cıtak S, Sarıbas E, Erkılıc A, Tasci E. Extracorporeal membrane oxygenation as a bridge to lung transplantation in a Turkish lung transplantation program: our initial experience. J Artif Organs 2020; 24:36-43. [PMID: 32852668 PMCID: PMC7450232 DOI: 10.1007/s10047-020-01204-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/12/2020] [Indexed: 11/07/2022]
Abstract
Lung transplantation is a life-saving treatment for patients with end-stage lung disease. Although the number of lung transplants has increased over the years, the number of available donor lungs has not increased at the same rate, leading to the death of transplant candidates on waiting lists. In this paper, we presented our initial experience with the use of extracorporeal membrane oxygenation (ECMO) as a bridge to lung transplantation. Between December 2016 and August 2018, we retrospectively reviewed the use of ECMO as a bridge to lung transplantation. Thirteen patients underwent preparative ECMO for bridging to lung transplantation, and seven patients successfully underwent bridging to lung transplantation. The average age of the patients was 45.7 years (range, 19–62 years). The ECMO support period lasted 3–55 days (mean, 18.7 days; median, 13 days). In seven patients, bridging to lung transplantation was performed successfully. The mean age of patients was 49.8 years (range 42–62). Bridging time was 3–55 days (mean, 19 days; median, 13 days). Two patients died in the early postoperative period. Five patients survived until discharge from the hospital. One-year survival was achieved in four patients. ECMO can be used safely for a long time to meet the physiological needs of critically ill patients. The use of ECMO as a bridge to lung transplantation is an acceptable treatment option to reduce the number of deaths on the waiting list. Despite the successful results achieved, this approach still involves risks and complications.
Collapse
Affiliation(s)
- Mustafa Vayvada
- Thoracic Surgery, Kartal Kosuyolu Training and Research Hospital, K Blok Cevizli, Kartal, Istanbul, Turkey.
| | - Yesim Uygun
- Infectious Diseases, Kartal Kosuyolu Training and Research Hospital, Istanbul, Turkey
| | - Sevinc Cıtak
- Thoracic Surgery, Kartal Kosuyolu Training and Research Hospital, K Blok Cevizli, Kartal, Istanbul, Turkey
| | - Ertan Sarıbas
- Chest Diseases, Kartal Kosuyolu Training and Research Hospital, Istanbul, Turkey
| | - Atakan Erkılıc
- Anesthesia and Reanimation, Kartal Kosuyolu Training and Research Hospital, Istanbul, Turkey
| | - Erdal Tasci
- Thoracic Surgery, Kartal Kosuyolu Training and Research Hospital, K Blok Cevizli, Kartal, Istanbul, Turkey
| |
Collapse
|
170
|
Cohen W, Mirzai S, Combs P, Rose R, Kagan V, Correia C, Gottlieb LJ, Song T. Postoperative extracorporeal membrane oxygenation can successfully support patients following upper airway reconstruction. Head Neck 2020; 42:E30-E34. [PMID: 32767409 DOI: 10.1002/hed.26371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/23/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Patients requiring intensive upper airway reconstruction are anatomically restricted in terms of the respiratory support they can receive. While intraoperative extracorporeal membrane oxygenation (ECMO) has been effectively utilized, little has been shown regarding the utility of ECMO for long-term support in these patients. METHODS We demonstrate how a patient with tongue and hypopharyngeal squamous cell carcinoma that necessitated upper airway reconstruction was supported with veno-venous (VV) ECMO due to postoperative respiratory failure and an inability to maintain a stable airway. RESULTS By initiating VV ECMO, we were able to decrease positive pressure ventilation and FiO2 , thereby minimizing ventilator-associated trauma and irritation to facilitate wound healing. Over time, ventilatory support was increased in parallel with decreasing ECMO support, allowing discharge to rehabilitation after 74 days of ECMO. CONCLUSION ECMO can effectively support patients with ongoing respiratory requirements following upper airway reconstruction when standard ventilatory techniques are inadequate or not feasible.
Collapse
Affiliation(s)
- William Cohen
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Saeid Mirzai
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Pamela Combs
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Rebecca Rose
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Viktoriya Kagan
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Camil Correia
- Section of Otolaryngology - Head and Neck Surgery, Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Lawrence J Gottlieb
- Section of Plastic and Reconstructive Surgery, Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Tae Song
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
171
|
Sine ventilation in lung injury models: a new perspective for lung protective ventilation. Sci Rep 2020; 10:11690. [PMID: 32678177 PMCID: PMC7366701 DOI: 10.1038/s41598-020-68614-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/29/2020] [Indexed: 11/18/2022] Open
Abstract
Mechanical ventilation is associated with the risk of ventilator induced lung injury. For reducing lung injury in mechanically ventilated patients, the application of small tidal volumes and positive end-expiratory pressures has become clinical standard. Recently, an approach based on linear airway pressure decline and decelerated expiratory flow during expiration implied lung protective capacities. We assumed that ventilation with a smoothed, i.e. sinusoidal airway pressure profile may further improve ventilation efficiency and lung protection. We compared the effects of mechanical ventilation with sinusoidal airway pressure profile (SINE) regarding gas exchange, respiratory system compliance and histology to conventional volume and pressure controlled ventilation (VCV and PCV) and to VCV with flow-controlled expiration (FLEX) in two rat models of lung injury, tween induced surfactant depletion and high tidal volume mechanical ventilation. In both lung injury models ventilation with SINE showed more efficient CO2 elimination and blood oxygenation, improved respiratory system compliance and resulted in lower alveolar wall thickness, compared to VCV, PCV and FLEX. Optimization of the airway pressure profile may provide a novel means of lung protective mechanical ventilation.
Collapse
|
172
|
Mal H, Santin G, Cantrelle C, Durand L, Legeai C, Cheisson G, Saint-Marcel L, Pipien I, Durin L, Bastien O, Dorent R. Effect of Lung-Protective Ventilation in Organ Donors on Lung Procurement and Recipient Survival. Am J Respir Crit Care Med 2020; 202:250-258. [DOI: 10.1164/rccm.201910-2067oc] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Hervé Mal
- Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Paris, France
- Inserm UMR1152, Université Paris 7 Denis Diderot, Paris, France
| | | | | | | | | | - Gaëlle Cheisson
- Hôpital Kremlin Bicêtre, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France; and
| | | | | | | | | | | |
Collapse
|
173
|
Allam M, Cai S, Ganesh S, Venkatesan M, Doodhwala S, Song Z, Hu T, Kumar A, Heit J, Coskun AF. COVID-19 Diagnostics, Tools, and Prevention. Diagnostics (Basel) 2020; 10:E409. [PMID: 32560091 PMCID: PMC7344926 DOI: 10.3390/diagnostics10060409] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/13/2020] [Accepted: 06/14/2020] [Indexed: 12/27/2022] Open
Abstract
The Coronavirus Disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), outbreak from Wuhan City, Hubei province, China in 2019 has become an ongoing global health emergency. The emerging virus, SARS-CoV-2, causes coughing, fever, muscle ache, and shortness of breath or dyspnea in symptomatic patients. The pathogenic particles that are generated by coughing and sneezing remain suspended in the air or attach to a surface to facilitate transmission in an aerosol form. This review focuses on the recent trends in pandemic biology, diagnostics methods, prevention tools, and policies for COVID-19 management. To meet the growing demand for medical supplies during the COVID-19 era, a variety of personal protective equipment (PPE) and ventilators have been developed using do-it-yourself (DIY) manufacturing. COVID-19 diagnosis and the prediction of virus transmission are analyzed by machine learning algorithms, simulations, and digital monitoring. Until the discovery of a clinically approved vaccine for COVID-19, pandemics remain a public concern. Therefore, technological developments, biomedical research, and policy development are needed to decipher the coronavirus mechanism and epidemiological characteristics, prevent transmission, and develop therapeutic drugs.
Collapse
Affiliation(s)
- Mayar Allam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA; (M.A.); (S.C.); (S.G.); (M.V.); (S.D.); (Z.S.); (T.H.); (A.K.); (J.H.); (C.S.G.)
| | - Shuangyi Cai
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA; (M.A.); (S.C.); (S.G.); (M.V.); (S.D.); (Z.S.); (T.H.); (A.K.); (J.H.); (C.S.G.)
| | - Shambavi Ganesh
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA; (M.A.); (S.C.); (S.G.); (M.V.); (S.D.); (Z.S.); (T.H.); (A.K.); (J.H.); (C.S.G.)
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Mythreye Venkatesan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA; (M.A.); (S.C.); (S.G.); (M.V.); (S.D.); (Z.S.); (T.H.); (A.K.); (J.H.); (C.S.G.)
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Saurabh Doodhwala
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA; (M.A.); (S.C.); (S.G.); (M.V.); (S.D.); (Z.S.); (T.H.); (A.K.); (J.H.); (C.S.G.)
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA 30318, USA
| | - Zexing Song
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA; (M.A.); (S.C.); (S.G.); (M.V.); (S.D.); (Z.S.); (T.H.); (A.K.); (J.H.); (C.S.G.)
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA 30318, USA
| | - Thomas Hu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA; (M.A.); (S.C.); (S.G.); (M.V.); (S.D.); (Z.S.); (T.H.); (A.K.); (J.H.); (C.S.G.)
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Aditi Kumar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA; (M.A.); (S.C.); (S.G.); (M.V.); (S.D.); (Z.S.); (T.H.); (A.K.); (J.H.); (C.S.G.)
| | - Jeremy Heit
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA; (M.A.); (S.C.); (S.G.); (M.V.); (S.D.); (Z.S.); (T.H.); (A.K.); (J.H.); (C.S.G.)
| | - COVID-19 Study Group
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA; (M.A.); (S.C.); (S.G.); (M.V.); (S.D.); (Z.S.); (T.H.); (A.K.); (J.H.); (C.S.G.)
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30313, USA
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Ahmet F. Coskun
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA; (M.A.); (S.C.); (S.G.); (M.V.); (S.D.); (Z.S.); (T.H.); (A.K.); (J.H.); (C.S.G.)
| |
Collapse
|
174
|
Kirov MY, Kuzkov VV. Protective ventilation from ICU to operating room: state of art and new horizons. Korean J Anesthesiol 2020; 73:179-193. [PMID: 32008277 PMCID: PMC7280889 DOI: 10.4097/kja.19499] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 01/26/2020] [Accepted: 01/27/2020] [Indexed: 12/16/2022] Open
Abstract
The prevention of ventilator-associated lung injury (VALI) and postoperative pulmonary complications (PPC) is of paramount importance for improving outcomes both in the operating room and in the intensive care unit (ICU). Protective respiratory support includes a wide spectrum of interventions to decrease pulmonary stress-strain injuries. The motto 'low tidal volume for all' should become routine, both during major surgery and in the ICU, while application of a high positive end-expiratory pressure (PEEP) strategy and of alveolar recruitment maneuvers requires a personalized approach and requires further investigation. Patient self-inflicted lung injury is an important type of VALI, which should be diagnosed and mitigated at the early stage, during restoration of spontaneous breathing. This narrative review highlights the strategies used for protective positive pressure ventilation. The emerging concepts of damaging energy and power, as well as pathways to personalization of the respiratory settings, are discussed in detail. In the future, individualized approaches to protective ventilation may involve multiple respiratory settings extending beyond low tidal volume and PEEP, implemented in parallel with quantifying the risk of VALI and PPC.
Collapse
Affiliation(s)
- Mikhail Y. Kirov
- Department of Anesthesiology and Intensive Care Medicine, Northern State Medical University, Arkhangelsk, Russian Federation
| | - Vsevolod V. Kuzkov
- Department of Anesthesiology and Intensive Care Medicine, Northern State Medical University, Arkhangelsk, Russian Federation
| |
Collapse
|
175
|
Rajagopal K, Keller SP, Akkanti B, Bime C, Loyalka P, Cheema FH, Zwischenberger JB, El Banayosy A, Pappalardo F, Slaughter MS, Slepian MJ. Advanced Pulmonary and Cardiac Support of COVID-19 Patients: Emerging Recommendations From ASAIO-A "Living Working Document". ASAIO J 2020; 66:588-598. [PMID: 32358232 PMCID: PMC7217129 DOI: 10.1097/mat.0000000000001180] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The severe acute respiratory syndrome (SARS)-CoV-2 is an emerging viral pathogen responsible for the global coronavirus disease 2019 (COVID)-19 pandemic resulting in significant human morbidity and mortality. Based on preliminary clinical reports, hypoxic respiratory failure complicated by acute respiratory distress syndrome is the leading cause of death. Further, septic shock, late-onset cardiac dysfunction, and multiorgan system failure are also described as contributors to overall mortality. Although extracorporeal membrane oxygenation and other modalities of mechanical cardiopulmonary support are increasingly being utilized in the treatment of respiratory and circulatory failure refractory to conventional management, their role and efficacy as support modalities in the present pandemic are unclear. We review the rapidly changing epidemiology, pathophysiology, emerging therapy, and clinical outcomes of COVID-19; and based on these data and previous experience with artificial cardiopulmonary support strategies, particularly in the setting of infectious diseases, provide consensus recommendations from ASAIO. Of note, this is a "living document," which will be updated periodically, as additional information and understanding emerges.
Collapse
Affiliation(s)
- Keshava Rajagopal
- From the Departments of Clinical and Biomedical Sciences, University of Houston College of Medicine, Houston, TX
- Houston Heart, HCA Houston Healthcare, Houston, TX
| | - Steven P. Keller
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Bindu Akkanti
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, McGovern Medical School, University of Texas-Houston, Houston, TX
| | - Christian Bime
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Arizona College of Medicine - Tucson and Banner Health, Tucson, AZ
| | | | - Faisal H. Cheema
- From the Departments of Clinical and Biomedical Sciences, University of Houston College of Medicine, Houston, TX
- Houston Heart, HCA Houston Healthcare, Houston, TX
- HCA Research Institute, Nashville, TN
| | - Joseph B. Zwischenberger
- Department of Surgery, University of Kentucky College of Medicine and Medical Center, Lexington, KY
| | - Aly El Banayosy
- Nazih Zuhdi Transplant Institute, Integris Baptist Medical Center, Oklahoma City, OK
| | | | - Mark S. Slaughter
- Department of Cardiovascular and Thoracic Surgery, University of Louisville School of Medicine and Jewish Hospital, Louisville, KY
| | - Marvin J. Slepian
- Department of Cardiovascular and Thoracic Surgery, University of Louisville School of Medicine and Jewish Hospital, Louisville, KY
| |
Collapse
|
176
|
Rajagopal K, Keller SP, Akkanti B, Bime C, Loyalka P, Cheema FH, Zwischenberger JB, El-Banayosy A, Pappalardo F, Slaughter MS, Slepian MJ. Advanced Pulmonary and Cardiac Support of COVID-19 Patients: Emerging Recommendations From ASAIO -a Living Working Document. Circ Heart Fail 2020; 13:e007175. [PMID: 32357074 PMCID: PMC7304497 DOI: 10.1161/circheartfailure.120.007175] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The severe acute respiratory syndrome-CoV-2 is an emerging viral pathogen responsible for the global coronavirus disease 2019 pandemic resulting in significant human morbidity and mortality. Based on preliminary clinical reports, hypoxic respiratory failure complicated by acute respiratory distress syndrome is the leading cause of death. Further, septic shock, late-onset cardiac dysfunction, and multiorgan system failure are also described as contributors to overall mortality. Although extracorporeal membrane oxygenation and other modalities of mechanical cardiopulmonary support are increasingly being utilized in the treatment of respiratory and circulatory failure refractory to conventional management, their role and efficacy as support modalities in the present pandemic are unclear. We review the rapidly changing epidemiology, pathophysiology, emerging therapy, and clinical outcomes of coronavirus disease 2019; and based on these data and previous experience with artificial cardiopulmonary support strategies, particularly in the setting of infectious diseases, provide consensus recommendations from American Society for Artificial Internal Organs. Of note, this is a living document, which will be updated periodically, as additional information and understanding emerges.
Collapse
Affiliation(s)
- Keshava Rajagopal
- University of Houston College of Medicine, Houston, TX
- Houston Heart, HCA Houston Healthcare, Houston, TX
| | - Steven P. Keller
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Bindu Akkanti
- University of Texas-Houston & Memorial Hermann-Texas Medical Center, Houston, TX
| | - Christian Bime
- University of Arizona College of Medicine and Banner Health, Tucson, AZ
| | | | - Faisal H. Cheema
- University of Houston College of Medicine, Houston, TX
- Houston Heart, HCA Houston Healthcare, Houston, TX
- HCA Research Institute, Nashville, TN
| | | | | | | | - Mark S. Slaughter
- University of Louisville School of Medicine and Jewish Hospital, Louisville, KY
| | - Marvin J. Slepian
- University of Arizona College of Medicine and Banner Health, Tucson, AZ
| |
Collapse
|
177
|
Liu J, Huang X, Hu S, Meng Z, He H. Individualized lung protective ventilation vs. conventional ventilation during general anesthesia in laparoscopic total hysterectomy. Exp Ther Med 2020; 19:3051-3059. [PMID: 32256792 PMCID: PMC7086193 DOI: 10.3892/etm.2020.8549] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 11/21/2019] [Indexed: 12/15/2022] Open
Abstract
Laparoscopic total hysterectomy is performed by carbon dioxide insufflation, Trendelenburg position and mechanical ventilation of patients under general anesthesia. However, this may induce pulmonary atelectasis and/or hyperdistention of the lungs. Multiple studies have indicated that mechanical ventilation with the use of low tidal volumes, moderate positive end-expiratory pressure (PEEP) and regular alveolar recruitment maneuvers may improve post-operative outcomes. However, the benefits of an individualized level of PEEP have not been clearly established. In the present study, it was hypothesized that a moderate fixed PEEP may not suit all patients and an individually-titrated PEEP during anesthesia may improve the peri-operative pulmonary oxygenation function. The aim of the present study was to compare the pulmonary oxygenation function and post-operative pulmonary complications (PPCs) in patients receiving individualized lung-protective mechanical ventilation (LPV) vs. conventional ventilation (CV) during laparoscopic total hysterectomy. The present study was a randomized double-blinded clinical trial on 87 patients who were randomly divided to receive CV or protective ventilation (PV). An optimal individualized PEEP value was determined using a static pulmonary compliance-directed PEEP titration procedure. Pulmonary oxygenation function, serum inflammatory factors, including interleukin-8 and Clara cell protein 16, the incidence of PPCs and the post-operative length of stay were also determined. Patients in the PV group exhibited improved pulmonary oxygenation function during and after the operation. The total percentage of PPCs during the first 7 days after surgery was significantly lower in the PV group compared with those in the CV group. In conclusion, as compared to CV, intra-operative individualized LPV significantly improved pulmonary oxygenation function and reduced the incidence of PPCs during the first 7 days after laparoscopic total hysterectomy (Clinical trial registration no. ChiCTR1900027738).
Collapse
Affiliation(s)
- Jing Liu
- Department of Anesthesiology, Huzhou Maternity and Child Healthcare Hospital, Huzhou, Zhejiang 313000, P.R. China
| | - Xinhua Huang
- Department of Anesthesiology, Huzhou Maternity and Child Healthcare Hospital, Huzhou, Zhejiang 313000, P.R. China
| | - Siping Hu
- Department of Anesthesiology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Zhipeng Meng
- Department of Anesthesiology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| | - Huanzhong He
- Department of Anesthesiology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang 313000, P.R. China
| |
Collapse
|
178
|
Weber J, Schmidt J, Straka L, Wirth S, Schumann S. Flow-controlled ventilation improves gas exchange in lung-healthy patients- a randomized interventional cross-over study. Acta Anaesthesiol Scand 2020; 64:481-488. [PMID: 31828755 DOI: 10.1111/aas.13526] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 11/04/2019] [Accepted: 11/27/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Flow-controlled ventilation (FCV) is a new ventilation mode that provides constant inspiratory and expiratory flow. FCV was shown to improve gas exchange and lung recruitment in porcine models of healthy and injured ventilated lungs. The primary aim of our study was to verify the influences of FCV on gas exchange, respiratory mechanics and haemodynamic variables in mechanically ventilated lung-healthy patients. METHODS After obtaining ethical approval and informed consent, we measured arterial blood gases, respiratory and haemodynamic variables during volume-controlled ventilation (VCV) and FCV in 20 consecutive patients before they underwent abdominal surgery. After baseline (BL) ventilation, patients were randomly assigned to either BL-VCV-FCV or BL-FCV-VCV. Thereby, BL ventilation settings were kept, except for the ventilation mode-related differences (FCV is supposed to be used with an I:E ratio of 1:1). RESULTS Compared to BL and VCV, PaO2 was higher [PaO2 : FCV: 38.2 (7.1), BL ventilation: 35.0 (5.8), VCV: 35.2 (7.0) kPa, P < .001] and PaCO2 lower [PaCO2 : FCV: 4.8 (0.5), BL ventilation: 5.1 (0.5), VCV: 5.1 (0.5) kPa, P < .001] during FCV. With comparable plateau pressure [BL: 14.9 (1.9), VCV: 15.3 (1.6), FCV: 15.2 (1.5) cm H2 O), P = .185], tracheal mean pressure was higher during FCV [BL: 10.2 (1.1), VCV: 10.4 (0.7), FCV: 11.5 (1.0) cm H2 O, P < .001]. Haemodynamic variables did not differ between ventilation phases. CONCLUSION Flow-controlled ventilation improves oxygenation and carbon dioxide elimination within a short time, compared to VCV with identical tidal volume, inspiratory plateau pressure and end-expiratory pressure.
Collapse
Affiliation(s)
- Jonas Weber
- Department of Anesthesiology and Critical Care Medical Center – University of Freiburg Faculty of Medicine University of Freiburg Freiburg Germany
| | - Johannes Schmidt
- Department of Anesthesiology and Critical Care Medical Center – University of Freiburg Faculty of Medicine University of Freiburg Freiburg Germany
| | - Leonie Straka
- Department of Anesthesiology and Critical Care Medical Center – University of Freiburg Faculty of Medicine University of Freiburg Freiburg Germany
| | - Steffen Wirth
- Department of Anesthesiology and Critical Care Medical Center – University of Freiburg Faculty of Medicine University of Freiburg Freiburg Germany
| | - Stefan Schumann
- Department of Anesthesiology and Critical Care Medical Center – University of Freiburg Faculty of Medicine University of Freiburg Freiburg Germany
| |
Collapse
|
179
|
Abstract
Ventilation-induced lung injury results from mechanical stress and strain that occur during tidal ventilation in the susceptible lung. Classical descriptions of ventilation-induced lung injury have focused on harm from positive pressure ventilation. However, injurious forces also can be generated by patient effort and patient–ventilator interactions. While the role of global mechanics has long been recognized, regional mechanical heterogeneity within the lungs also appears to be an important factor propagating clinically significant lung injury. The resulting clinical phenotype includes worsening lung injury and a systemic inflammatory response that drives extrapulmonary organ failures. Bedside recognition of ventilation-induced lung injury requires a high degree of clinical acuity given its indistinct presentation and lack of definitive diagnostics. Yet the clinical importance of ventilation-induced lung injury is clear. Preventing such biophysical injury remains the most effective management strategy to decrease morbidity and mortality in patients with acute respiratory distress syndrome and likely benefits others at risk.
Collapse
Affiliation(s)
- Purnema Madahar
- Center for Acute Respiratory Failure, Columbia University College of Physicians and Surgeons, New York City, NY, USA.,Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University College of Physicians and Surgeons, New York City, NY, USA.,Department of Medicine, New York-Presbyterian Hospital, New York City, NY, USA
| | - Jeremy R Beitler
- Center for Acute Respiratory Failure, Columbia University College of Physicians and Surgeons, New York City, NY, USA.,Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University College of Physicians and Surgeons, New York City, NY, USA.,Department of Medicine, New York-Presbyterian Hospital, New York City, NY, USA
| |
Collapse
|
180
|
Elenkov M, Ecker P, Lukitsch B, Janeczek C, Harasek M, Gföhler M. Estimation Methods for Viscosity, Flow Rate and Pressure from Pump-Motor Assembly Parameters. SENSORS 2020; 20:s20051451. [PMID: 32155844 PMCID: PMC7085755 DOI: 10.3390/s20051451] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/24/2020] [Accepted: 02/28/2020] [Indexed: 01/02/2023]
Abstract
Blood pumps have found applications in heart support devices, oxygenators, and dialysis systems, among others. Often, there is no room for sensors, or the sensors are simply unreliable when long-term operation is required. However, control systems rely on those hard-to-measure parameters, such as blood flow rate and pressure difference, thus their estimation takes a central role in the development process of such medical devices. The viscosity of the blood not only influences the estimation of those parameters but is often a parameter that is of great interest to both doctors and engineers. In this work, estimation methods for blood flow rate, pressure difference, and viscosity are presented using Gaussian process regression models. Different water–glycerol mixtures were used to model blood. Data was collected from a custom-built blood pump, designed for intracorporeal oxygenators in an in vitro test circuit. The estimation was performed from motor current and motor speed measurements and its accuracy was measured for: blood flow rate r2 = 0.98, root mean squared error (RMSE) = 46 mL.min−1; pressure difference r2 = 0.98, RMSE = 8.7 mmHg; and viscosity r2 = 0.98, RMSE = 0.049 mPa.s. The results suggest that the presented methods can be used to accurately predict blood flow rate, pressure, and viscosity online.
Collapse
Affiliation(s)
- Martin Elenkov
- Institute of Engineering Design and Product Development, TU Wien, 1060 Vienna, Austria; (P.E.); (C.J.); (M.G.)
- Correspondence: ; Tel.: +43-1-58801-30764
| | - Paul Ecker
- Institute of Engineering Design and Product Development, TU Wien, 1060 Vienna, Austria; (P.E.); (C.J.); (M.G.)
- Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, 1060 Vienna, Austria; (B.L.); (M.H.)
| | - Benjamin Lukitsch
- Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, 1060 Vienna, Austria; (B.L.); (M.H.)
| | - Christoph Janeczek
- Institute of Engineering Design and Product Development, TU Wien, 1060 Vienna, Austria; (P.E.); (C.J.); (M.G.)
| | - Michael Harasek
- Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, 1060 Vienna, Austria; (B.L.); (M.H.)
| | - Margit Gföhler
- Institute of Engineering Design and Product Development, TU Wien, 1060 Vienna, Austria; (P.E.); (C.J.); (M.G.)
| |
Collapse
|
181
|
Bedside respiratory physiology to detect risk of lung injury in acute respiratory distress syndrome. Curr Opin Crit Care 2020; 25:3-11. [PMID: 30531534 DOI: 10.1097/mcc.0000000000000579] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW The most effective strategies for treating the patient with acute respiratory distress syndrome center on minimizing ventilation-induced lung injury (VILI). Yet, current standard-of-care does little to modify mechanical ventilation to patient-specific risk. This review focuses on evaluation of bedside respiratory mechanics, which when interpreted in patient-specific context, affords opportunity to individualize lung-protective ventilation in patients with acute respiratory distress syndrome. RECENT FINDINGS Four biophysical mechanisms of VILI are widely accepted: volutrauma, barotrauma, atelectrauma, and stress concentration. Resulting biotrauma, that is, local and systemic inflammation and endothelial activation, may be thought of as the final common pathway that propagates VILI-mediated multiorgan failure. Conventional, widely utilized techniques to assess VILI risk rely on airway pressure, flow, and volume changes, and remain essential tools for determining overdistension of aerated lung regions, particularly when interpreted cognizant of their limitations. Emerging bedside tools identify regional differences in mechanics, but further study is required to identify how they might best be incorporated into clinical practice. SUMMARY Quantifying patient-specific risk of VILI requires understanding each patient's pulmonary mechanics in context of biological predisposition. Tailoring support at bedside according to these factors affords the greatest opportunity to date for mitigating VILI and alleviating associated morbidity.
Collapse
|
182
|
|
183
|
Huang H, Feng H, Zhuge D. M1 Macrophage Activated by Notch Signal Pathway Contributed to Ventilator-Induced Lung Injury in Chronic Obstructive Pulmonary Disease Model. J Surg Res 2019; 244:358-367. [PMID: 31323391 DOI: 10.1016/j.jss.2019.06.060] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/10/2019] [Accepted: 06/14/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND Ventilator-induced lung injury (VILI) in chronic obstructive pulmonary disease (COPD) is still a problem. We intended to explore the role of macrophage polarity in VILI and the underlying mechanism. MATERIALS AND METHODS COPD model was created by cigarette smoke and ventilated. Macrophages were isolated, and the wet/dry (W/D) ratio was determined after modeling, and proteins in bronchoalveolar lavage fluid (BALF) were assessed by bicinchoninic acid assay. Histopathology was observed by Hematoxylin-Eosin staining. Tumor necrosis factor (TNF)-α and interleukin (IL)-6 levels were measured by enzyme-linked immunosorbent assay. Macrophage polarity was assessed by flow cytometry. Protein levels were measured by Western blot and mRNA by quantitative real-time polymerase chain reaction. RESULTS Pathology statement was worsened, and the W/D ratio, protein level in BALF, TNF-α level, and IL-6 levels were elevated in cigarette smoke-induced COPD model. Notch-1 intracellular domain, hairy and enhancer of split (Hes) 1, Hes5, hairy/enhancer-of-split related with YRPW motif protein 1, CD86, TNF-α, and inducible nitric oxide synthases expressions were raised, whereas CD206, IL-4, and IL-10 expressions were decreased in macrophages after ventilation, shifting macrophage polarity to M1 phenotype. After the inhibition of Notch signaling, pathology statement was improved, and the W/D ratio, protein level in BALF, TNF-α, IL-6, Notch-1 intracellular domain, Hes1, Hes5, hairy/enhancer-of-split related with YRPW motif protein 1, CD86, TNF-α, and inducible nitric oxide synthases expressions were decreased while CD206, IL-4, and IL-10 expressions were elevated after ventilation, shifting macrophage polarity to M2 phenotype partially. CONCLUSIONS Mechanical ventilation in cigarette-induced COPD could activate the Notch signal pathway and further shift the polarity of macrophage toward M1 phenotype, leading to VILI in cigarette-induced COPD.
Collapse
Affiliation(s)
- Hongping Huang
- Department of Eastern Respiratory Medicine, Linyi People's Hospital, Linyi, China
| | - Hui Feng
- Linyi People's Hospital Office, Linyi People's Hospital, Linyi, China.
| | - Dong Zhuge
- Department of Eastern General Internal Medicine, Linyi People's Hospital, Linyi, China
| |
Collapse
|
184
|
Arnal JM, Saoli M, Garnero A. Airway and transpulmonary driving pressures and mechanical powers selected by INTELLiVENT-ASV in passive, mechanically ventilated ICU patients. Heart Lung 2019; 49:427-434. [PMID: 31733881 DOI: 10.1016/j.hrtlng.2019.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/31/2019] [Accepted: 11/01/2019] [Indexed: 01/27/2023]
Abstract
BACKGROUND Driving pressure (ΔP) and mechanical power (MP) are predictors of the risk of ventilation- induced lung injuries (VILI) in mechanically ventilated patients. INTELLiVENT-ASV® is a closed-loop ventilation mode that automatically adjusts respiratory rate and tidal volume, according to the patient's respiratory mechanics. OBJECTIVES This prospective observational study investigated ΔP and MP (and also transpulmonary ΔP (ΔPL) and MP (MPL) for a subgroup of patients) delivered by INTELLiVENT-ASV. METHODS Adult patients admitted to the ICU were included if they were sedated and met the criteria for a single lung condition (normal lungs, COPD, or ARDS). INTELLiVENT-ASV was used with default target settings. If PEEP was above 16 cmH2O, the recruitment strategy used transpulmonary pressure as a reference, and ΔPL and MPL were computed. Measurements were made once for each patient. RESULTS Of the 255 patients included, 98 patients were classified as normal-lungs, 28 as COPD, and 129 as ARDS patients. The median ΔP was 8 (7 - 10), 10 (8 - 12), and 9 (8 - 11) cmH2O for normal-lungs, COPD, and ARDS patients, respectively. The median MP was 9.1 (4.9 - 13.5), 11.8 (8.6 - 16.5), and 8.8 (5.6 - 13.8) J/min for normal-lungs, COPD, and ARDS patients, respectively. For the 19 patients managed with transpulmonary pressure ΔPL was 6 (4 - 7) cmH2O and MPL was 3.6 (3.1 - 4.4) J/min. CONCLUSIONS In this short term observation study, INTELLiVENT-ASV selected ΔP and MP considered in safe ranges for lung protection. In a subgroup of ARDS patients, the combination of a recruitment strategy and INTELLiVENT-ASV resulted in an apparently safe ΔPL and MPL.
Collapse
Affiliation(s)
- Jean-Michel Arnal
- Service de Réanimation Polyvalente, Hôpital Sainte Musse, 54 Avenue Henri Sainte Claire Deville, 83056 Toulon, France; Department of Medical Research, Hamilton Medical AG, via Crusch 8, 7402 Bonaduz, Switzerland.
| | - Mathieu Saoli
- Service de Réanimation Polyvalente, Hôpital Sainte Musse, 54 Avenue Henri Sainte Claire Deville, 83056 Toulon, France
| | - Aude Garnero
- Service de Réanimation Polyvalente, Hôpital Sainte Musse, 54 Avenue Henri Sainte Claire Deville, 83056 Toulon, France
| |
Collapse
|
185
|
Applying Positive End-Expiratory Pressure During Mechanical Ventilation Causes Pulmonary Redox Imbalance and Inflammation in Rats. Shock 2019; 50:572-578. [PMID: 29194341 DOI: 10.1097/shk.0000000000001072] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Mechanical ventilation (MV) may induce or aggravate lung injury through the production of cytokines, inflammatory infiltration of neutrophils, and changes in the permeability of the alveolar-capillary barrier. The use of positive end-expiratory pressure (PEEP) helps improve gas exchanges avoiding alveolar collapse at the end of expiration. The present study aimed to analyze inflammatory response and redox imbalance in lungs of rats submitted to MV with and without PEEP. METHODS Eighteen Wistar rats were divided into three groups: control (CG), PEEP group (PG), and zero PEEP (ZEEP) group (ZG). PG and ZG were submitted to MV for 60 min with or without PEEP, respectively. Subsequently, the animals were euthanized, and blood, bronchoalveolar lavage fluid, and lungs were collected for analyses. RESULTS The number of neutrophils was higher in PG compared with CG. Leucocyte and neutrophil influx in bronchoalveolar lavage fluid was higher in PG compared with CG. PG showed an increase in alveolar area compared with the other groups. There were increases in the levels of chemokines, CCL3 and CCL5, in PG compared with CG. There were increases in oxidation of lipids and proteins in PG compared with other groups. There were increases in the activity of superoxide dismutase and catalase in PG compared with CG and ZG. However, there was a decrease in the ratio of glutathione to glutathione disulfide in PG compared with other groups. CONCLUSIONS MV with PEEP caused redox imbalance and inflammation in lungs of healthy rats.
Collapse
|
186
|
Rackley CR, MacIntyre NR. Low Tidal Volumes for Everyone? Chest 2019; 156:783-791. [DOI: 10.1016/j.chest.2019.06.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/13/2019] [Accepted: 06/06/2019] [Indexed: 01/03/2023] Open
|
187
|
Hyperoxia affects the lung tissue: A porcine histopathological and metabolite study using five hours of apneic oxygenation. Metabol Open 2019; 4:100018. [PMID: 32812938 PMCID: PMC7424812 DOI: 10.1016/j.metop.2019.100018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/12/2019] [Accepted: 09/12/2019] [Indexed: 12/14/2022] Open
Abstract
Background Oxygen is a liberally dosed medicine; however, too much oxygen can be harmful. In certain situations, treatment with high oxygen concentration is necessary, e.g. after cardiopulmonary resuscitation. The amount of oxygen and duration of hyperoxia causing pulmonary damage is not fully elucidated. The aim of this study was to investigate pathophysiological and metabolite changes in lung tissue during hyperoxia while the lungs were kept open under constant low pressure. Methods Seven pigs were exposed to 100% oxygen for five hours, using an apneic oxygenation technique with one long uninterrupted inspiration, while carbon dioxide was removed with an interventional lung assist. Arterial blood samples were collected every 30 minutes. Lung biopsies were obtained before and after hyperoxia. Microscopy and high-resolution magic angle spinning nuclear magnetic resonance spectroscopy were used to detect possible pathological and metabolite changes, respectively. Unsupervised multivariate analysis of variance and paired sample tests were performed. A two-tailed p-value ≤ 0.05 was considered significant. Results No significant changes in arterial pH, and partial pressure of carbon dioxide, and no clear histopathological changes were observed after hyperoxia. While blood glucose and lactate levels changed to a minor degree, their levels dropped significantly in the lung after hyperoxia (p ≤ 0.04). Reduced levels of antioxidants (p ≤ 0.05), tricarboxylic acid cycle and energy (p ≤ 0.04) metabolites and increased levels of several amino acids (p ≤ 0.05) were also detected. Conclusion Despite no histological changes, tissue metabolites were altered, indicating that exposure to hyperoxia affects lung tissue matrix on a molecular basis. No significant histopathological changes in lung tissue after five hours hyperoxia. Five hours hyperoxia induces significant metabolite changes in lung tissue. Hyperoxia affects cellular energy, Krebs cycle, and oxidant-antioxidant defense.
Collapse
|
188
|
Closer to Nature Through Dynamic Culture Systems. Cells 2019; 8:cells8090942. [PMID: 31438519 PMCID: PMC6769584 DOI: 10.3390/cells8090942] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/16/2019] [Accepted: 08/19/2019] [Indexed: 12/12/2022] Open
Abstract
Mechanics in the human body are required for normal cell function at a molecular level. It is now clear that mechanical stimulations play significant roles in cell growth, differentiation, and migration in normal and diseased cells. Recent studies have led to the discovery that normal and cancer cells have different mechanosensing properties. Here, we discuss the application and the physiological and pathological meaning of mechanical stimulations. To reveal the optimal conditions for mimicking an in vivo microenvironment, we must, therefore, discern the mechanotransduction occurring in cells.
Collapse
|
189
|
Ventilation-Like Mechanical Strain Modulates the Inflammatory Response of BEAS2B Epithelial Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2769761. [PMID: 31320981 PMCID: PMC6607724 DOI: 10.1155/2019/2769761] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/29/2019] [Accepted: 05/28/2019] [Indexed: 01/16/2023]
Abstract
Protective mechanical ventilation is aimed at preventing ventilator-induced lung injury while ensuring sufficient gas exchange. A new approach focuses on the temporal profile of the mechanical ventilation. We hypothesized that the temporal mechanical strain profile modulates inflammatory signalling. We applied cyclic strain with various temporal profiles to human bronchial epithelial cells (BEAS2B) and assessed proinflammatory response. The cells were subjected to sinusoidal, rectangular, or triangular strain profile and rectangular strain profile with prestrain set to 0, 25, 50, or 75% of the maximum stain, static strain, and strain resembling a mechanical ventilation-like profile with or without flow-controlled expiration. The BEAS2B response to mechanical load included altered mitochondrial activity, increased superoxide radical levels, NF-kappaB translocation, and release of interleukin-8. The response to strain was substantially modulated by the dynamics of the stimulation pattern. The rate of dynamic changes of the strain profile correlates with the degree of mechanical stress-induced cell response.
Collapse
|
190
|
Wu X, Zheng R, Zhuang Z. Effect of transpulmonary pressure-guided positive end-expiratory pressure titration on lung injury in pigs with acute respiratory distress syndrome. J Clin Monit Comput 2019; 34:151-159. [PMID: 30903412 PMCID: PMC6946758 DOI: 10.1007/s10877-019-00267-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 01/25/2019] [Indexed: 01/17/2023]
Abstract
To investigate the effect of positive end-expiratory pressure (PEEP) guided by transpulmonary pressure or with maximum oxygenation-directed PEEP on lung injury in a porcine model of acute respiratory distress syndrome (ARDS). The porcine model of ARDS was induced in 12 standard pigs by intratracheal infusion with normal saline. The pigs were then randomly divided into two groups who were ventilated with the lung-protective strategy of low tidal volume (VT) (6 ml/kg), using different methods to titrate PEEP level: transpulmonary pressure (TP group; n = 6) or maximum oxygenation (MO group; n = 6). Gas exchange, pulmonary mechanics, and hemodynamics were determined and pulmonary inflammatory response indices were measured after 4 h of ventilation. The titrated PEEP level in the TP group (6.12 ± 0.89 cmH2O) was significantly lower than that in the MO group (11.33 ± 2.07 cmH2O) (P < 0.05). The PaO2/FiO2 (P/F) after PEEP titration both improved in the TP and MO groups as compared with that at T0 (when the criteria for ARDS were obtained). The P/F in the TP group did not differ significantly from that in the MO group during the 4 h of ventilation (P > 0.05). Respiratory system compliance and lung compliance were significantly improved in the TP group compared to the MO group (P < 0.05). The VD/VT in the TP group was significantly lower than that in the MO group after 4 h of ventilation (P < 0.05). Central venous pressure increased and the cardiac index decreased significantly in the MO group as compared with the TP group (P < 0.05), whereas oxygen delivery did not differ significantly between the groups (P > 0.05). The pulmonary vascular permeability index and the extravascular lung water index in the TP group were significantly lower than those in the MO group (P < 0.05). The TP group had a lower lung wet to dry weight ratio, lung injury score, and MPO, TNF-, and IL-8 concentrations than the MO group (P < 0.05). In summary, in a pig model of ARDS, ventilation with low VT and transpulmonary pressure-guided PEEP adjustment was associated with improved compliance, reduced dead space ventilation, increased cardiac output, and relieved lung injury, as compared to maximum oxygenation-guide PEEP adjustment.
Collapse
Affiliation(s)
- Xiaoyan Wu
- Department of Critical Care Medicine, Clinical Medical College, YangZhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, Jiangsu, China
| | - Ruiqiang Zheng
- Department of Critical Care Medicine, Clinical Medical College, YangZhou University, Northern Jiangsu People's Hospital, Yangzhou, 225001, Jiangsu, China.
| | - Zhiqing Zhuang
- Department of Neurology, Clinical Medical College, Wutaishan Hospital, YangZhou University, Yangzhou, 225001, Jiangsu, China
| |
Collapse
|
191
|
Barnes T, Enk D. Ventilation for low dissipated energy achieved using flow control during both inspiration and expiration. TRENDS IN ANAESTHESIA AND CRITICAL CARE 2019. [DOI: 10.1016/j.tacc.2018.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
192
|
Non‑canonical Wnt signaling contributes to ventilator‑induced lung injury through upregulation of WISP1 expression. Int J Mol Med 2019; 43:1217-1228. [PMID: 30664165 PMCID: PMC6365043 DOI: 10.3892/ijmm.2019.4067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 12/17/2018] [Indexed: 12/25/2022] Open
Abstract
Mechanical ventilation may cause ventilator-induced lung injury (VILI). Canonical Wnt signaling has been reported to serve an important role in the pathogenesis of VILI. Bioinformatics analysis revealed that canonical and non-canonical Wnt signaling pathways were activated in VILI. However, the role of non-canonical Wnt signaling in the pathogenesis of VILI remains unclear. The present study aimed to analyze the potential role of non-canonical Wnt signaling in VILI pathogenesis. Lung injury was assessed via Evans blue albumin permeability and histological scoring, as well as by inflammatory cytokine expression and total protein concentration in bronchoalveolar lavage fluid. The relative protein expression of canonical and non-canonical Wnt signaling pathway components were examined via western blotting and immunohistochemistry. The results demonstrated that 6 h of mechanical ventilation at low tidal volume (LTV; 6 ml/kg) or moderate tidal volume (MTV; 12 ml/kg) induced lung injury in sensitive A/J mice. Ventilation with MTV increased the protein levels of Wnt-induced secreted protein 1 (WISP1), Rho-associated protein kinase 1 (ROCK1), phosphorylated (p)-Ras homolog gene family, member A and p-C-Jun N-terminal kinase (JNK). Inhibition of ROCK1 by Y27632 and JNK by SP600125 attenuated MTV-induced lung injury and decreased the expression of proteins involved in non-canonical Wnt signaling, including WISP1. In conclusion, non-canonical Wnt signaling participates in VILI by modulating WISP1 expression, which has been previously noted as critical for VILI development. Therefore, the non-canonical Wnt signaling pathway may provide a preventive and therapeutic target in VILI.
Collapse
|
193
|
Evans D, Shure D, Clark L, Criner GJ, Dres M, de Abreu MG, Laghi F, McDonagh D, Petrof B, Nelson T, Similowski T. Temporary transvenous diaphragm pacing vs. standard of care for weaning from mechanical ventilation: study protocol for a randomized trial. Trials 2019; 20:60. [PMID: 30654837 PMCID: PMC6337771 DOI: 10.1186/s13063-018-3171-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/31/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mechanical ventilation (MV) is a life-saving technology that restores or assists breathing. Like any treatment, MV has side effects. In some patients it can cause diaphragmatic atrophy, injury, and dysfunction (ventilator-induced diaphragmatic dysfunction, VIDD). Accumulating evidence suggests that VIDD makes weaning from MV difficult, which involves increased morbidity and mortality. METHODS AND ANALYSIS This paper describes the protocol of a randomized, controlled, open-label, multicenter trial that is designed to investigate the safety and effectiveness of a novel therapy, temporary transvenous diaphragm pacing (TTVDP), to improve weaning from MV in up to 88 mechanically ventilated adult patients who have failed at least two spontaneous breathing trials over at least 7 days. Patients will be randomized (1:1) to TTVDP (treatment) or standard of care (control) groups. The primary efficacy endpoint is time to successful extubation with no reintubation within 48 h. Secondary endpoints include maximal inspiratory pressure and ultrasound-measured changes in diaphragm thickness and diaphragm thickening fraction over time. In addition, observational data will be collected and analyzed, including 30-day mortality and time to discharge from the intensive care unit and from the hospital. The hypothesis to be tested postulates that more TTVDP patients than control patients will be successfully weaned from MV within the 30 days following randomization. DISCUSSION This study is the first large-scale clinical trial of a novel technology (TTVDP) aimed at accelerating difficult weaning from MV. The technology tested provides the first therapy directed specifically at VIDD, an important cause of delayed weaning from MV. Its results will help delineate the place of this therapeutic approach in clinical practice and help design future studies aimed at defining the indications and benefits of TTVDP. TRIAL REGISTRATION ClinicalTrials.gov, NCT03096639 . Registered on 30 March 2017.
Collapse
Affiliation(s)
- Douglas Evans
- Lungpacer Medical Incorporated, Burnaby, BC, Canada.,Lungpacer Medical, 260 Sierra Drive, Exton, PA, 19335, USA
| | | | - Linda Clark
- Lungpacer Medical Incorporated, Burnaby, BC, Canada
| | - Gerard J Criner
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Martin Dres
- Sorbonne Université, INSERM, UMRS1158 Neurophysiologie Respiratoire Expérimentale et Clinique and AP-HP, Groupe Hospitalier Pitié-Salpêtrière Charles Foix, Service de Pneumologie et Réanimation Médicale du Département R3S, Paris, France
| | - Marcelo Gama de Abreu
- Department of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Franco Laghi
- Division of Pulmonary and Critical Care Medicine, Hines Veterans Affairs Hospital Hines, Loyola University, Maywood, IL, USA
| | - David McDonagh
- Departments of Anesthesiology and Pain Management, Neurological surgery, Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Basil Petrof
- Meakins-Christie Laboratories, and Translational Research in Respiratory Diseases Program, McGill University Health Centre and Research Institute, Montreal, QC, Canada
| | | | - Thomas Similowski
- Sorbonne Université, INSERM, UMRS1158 Neurophysiologie Respiratoire Expérimentale et Clinique and AP-HP, Groupe Hospitalier Pitié-Salpêtrière Charles Foix, Service de Pneumologie et Réanimation Médicale du Département R3S, Paris, France.
| |
Collapse
|
194
|
Song X, Liu Y, Dong L, Wang Y. Stromal-Interacting Molecule 1 (Stim1)/Orai1 Modulates Endothelial Permeability in Ventilator-Induced Lung Injury. Med Sci Monit 2018; 24:9413-9423. [PMID: 30589833 PMCID: PMC6322368 DOI: 10.12659/msm.911268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Increased endothelial permeability is involved in ventilator-induced lung injury (VILI). Stim1/Orai1 mediates store-operated Ca2+ activation, which modulates endothelial permeability. However, the underlying mechanisms of the Stim1/Orai1 pathway in VILI are poorly understood. Material/Methods Wistar rats were exposed to low tidal volume (7 mL/kg) or high tidal volume (40 mL/kg) ventilation. Human Lung Microvascular Endothelial Cells (HULEC) were subjected to 8% or 18% cyclic stretching (CS). BTP2 pretreatment was performed. Lung wet/dry weight ratio, histological changes of lung injury, and bronchoalveolar lavage fluid (BALF) protein were measured. Endothelial permeability and intracellular calcium concentration were evaluated in HULECs. Protein expression was determined by Western blotting. Results High tidal volume mechanical ventilation-induced lung injury (such as severe congestion and hemorrhage) and BTP2 pretreatment protected lungs from injury. The expression of Stim1, Orai1, and PKCα, lung wet/dry weight ratio, and BALF protein level significantly increased in the high tidal volume group compared to the control group and low tidal volume group. Importantly, BTP2 pretreatment alleviated the above-mentioned effects. Compared with exposure to 8% CS, the protein levels of Stim1, Orai1, and PKCα in HULECs significantly increased after exposure to 18% CS for 4 h, whereas BTP2 pretreatment significantly inhibited the increase (P<0.05). BTP2 pretreatment also suppressed increase of endothelial permeability and the intracellular calcium induced by 18% CS (P<0.05). Conclusions When exposed to high tidal volume or large-magnitude CS, Stim1 and Orai1 expression are upregulated, which further activates calcium-sensitive PKCα and results in calcium overload, endothelial hyperpermeability, and, finally, lung injury.
Collapse
Affiliation(s)
- Xiumei Song
- Department of Anesthesiology, Qianfoshan Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Yang Liu
- Department of Anesthesiology, Qianfoshan Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Ling Dong
- Department of Anesthesiology, Qianfoshan Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Yuelan Wang
- Department of Anesthesiology, Qianfoshan Hospital, Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
195
|
Inflammatory Cellular Response to Mechanical Ventilation in Elastase-Induced Experimental Emphysema: Role of Preexisting Alveolar Macrophages Infiltration. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5721293. [PMID: 30662910 PMCID: PMC6313972 DOI: 10.1155/2018/5721293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 11/07/2018] [Accepted: 11/11/2018] [Indexed: 12/13/2022]
Abstract
An excessive pulmonary inflammatory response could explain the poor prognosis of chronic obstructive pulmonary disease (COPD) patients submitted to invasive mechanical ventilation. The aim of this study was to evaluate the response to normal tidal volume mechanical ventilation in an elastase-induced murine model of pulmonary emphysema. In this model, two time points, associated with different levels of lung inflammation but similar lung destruction, were analyzed. C57BL/6 mice received a tracheal instillation of 5 IU of porcine pancreatic elastase (Elastase mice) or the same volume of saline (Saline mice). Fourteen (D14) and 21 (D21) days after instillation, mice were anesthetized, intubated, and either mechanically ventilated (MV) or maintained on spontaneous ventilation (SV) during two hours. As compared with Saline mice, Elastase mice showed a similarly increased mean chord length and pulmonary compliance at D14 and D21, while bronchoalveolar lavage cellularity was comparable between groups. Lung mechanics was similarly altered during mechanical ventilation in Elastase and Saline mice. Activated alveolar macrophages CD11bmid were present in lung parenchyma in both Elastase SV mice and Elastase MV mice at D14 but were absent at D21 and in Saline mice, indicating an inflammatory state with elastase at D14 only. At D14, Elastase MV mice showed a significant increase in percentage of neutrophils in total lung, as compared with Elastase SV mice. Furthermore, alveolar macrophages of Elastase MV mice at D14 overexpressed Gr1, and monocytes showed a trend to overexpression of CD62L, compared with Elastase SV mice. In an elastase-induced model of pulmonary emphysema, normal tidal volume mechanical ventilation may produce an increase in the proportion of pulmonary neutrophils, and an activation of alveolar macrophages and pulmonary monocytes. This response seems to be observed only when the emphysema model shows an underlying inflammation (D14), reflected by the presence of activated alveolar macrophages CD11bmid.
Collapse
|
196
|
Linnane MP, Caruana LR, Tronstad O, Corley A, Spooner AJ, Barnett AG, Thomas PJ, Walsh JR. A comparison of the effects of manual hyperinflation and ventilator hyperinflation on restoring end-expiratory lung volume after endotracheal suctioning: A pilot physiologic study. J Crit Care 2018; 49:77-83. [PMID: 30388492 DOI: 10.1016/j.jcrc.2018.10.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 10/28/2022]
Abstract
PURPOSE Endotracheal suctioning (ES) of mechanically ventilated patients decreases end-expiratory lung volume (EELV). Manual hyperinflation (MHI) and ventilator hyperinflation (VHI) may restore EELV post-ES but it remains unknown which method is most effective. The primary aim was to compare the efficacy of MHI and VHI in restoring EELV post-ES. MATERIALS AND METHODS ES was performed on mechanically ventilated intensive care patients, followed by MHI or VHI, in a randomised crossover design. The washout period between interventions was 1 h. End-expiratory lung impedance (EELI), measured by electrical impedance tomography, was recorded at baseline, during ES, during hyperinflation and 1, 5, 15 and 30 min post-hyperinflation. RESULTS Nine participants were studied. ES decreased EELI by 1672z (95% CI, 1204 to 2140) from baseline. From baseline, MHI increased EELI by 1154z (95% CI, 977 to 1330) while VHI increased EELI by 769z (95% CI, 457 to 1080). Five minutes post-VHI, EELI remained 528z (95% CI, 4 to 1053) above baseline. Fifteen minutes post-MHI, EELI remained 351z (95% CI, 111 to 592) above baseline. At subsequent time-points, EELI returned to baseline. CONCLUSIONS MHI and VHI effectively restore EELV above baseline post-ES and should be considered post suctioning.
Collapse
Affiliation(s)
- Matthew P Linnane
- Critical Care Research Group, The Prince Charles Hospital and University of Queensland, Brisbane, QLD 4032, Australia; Physiotherapy Department, The Prince Charles Hospital, Brisbane, QLD 4032, Australia.
| | - Lawrence R Caruana
- Critical Care Research Group, The Prince Charles Hospital and University of Queensland, Brisbane, QLD 4032, Australia; Physiotherapy Department, The Prince Charles Hospital, Brisbane, QLD 4032, Australia.
| | - Oystein Tronstad
- Critical Care Research Group, The Prince Charles Hospital and University of Queensland, Brisbane, QLD 4032, Australia; Physiotherapy Department, The Prince Charles Hospital, Brisbane, QLD 4032, Australia.
| | - Amanda Corley
- Critical Care Research Group, The Prince Charles Hospital and University of Queensland, Brisbane, QLD 4032, Australia.
| | - Amy J Spooner
- Institute of Health and Biomedical Innovation, School of Public Health, Queensland University of Technology, Brisbane, QLD 4000, Australia.
| | - Adrian G Barnett
- Institute of Health and Biomedical Innovation, School of Public Health, Queensland University of Technology, Brisbane, QLD 4000, Australia.
| | - Peter J Thomas
- Physiotherapy Department, Royal Brisbane and Women's Hospital, Brisbane, QLD 4006, Australia.
| | - James R Walsh
- Physiotherapy Department, The Prince Charles Hospital, Brisbane, QLD 4032, Australia; School of Allied Health Sciences, Griffith University, Gold Coast, QLD 4215, Australia.
| |
Collapse
|
197
|
Barnes T, van Asseldonk D, Enk D. Minimisation of dissipated energy in the airways during mechanical ventilation by using constant inspiratory and expiratory flows - Flow-controlled ventilation (FCV). Med Hypotheses 2018; 121:167-176. [PMID: 30396474 DOI: 10.1016/j.mehy.2018.09.038] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/16/2018] [Accepted: 09/22/2018] [Indexed: 12/16/2022]
Abstract
It has been suggested that energy dissipation in the airways during mechanical ventilation is associated with an increased probability of ventilator induced lung injury (VILI). We hypothesise that energy dissipation in the airways may be minimised by ventilating with constant flow during both the inspiration and expiration phases of the respiratory cycle. We present a simple analysis and numerical calculations that support our hypothesis and show that for ventilation with minimum dissipated energy not only should the flows during inspiration and expiration be controlled to be constant and continuous, but the ventilation should also be undertaken with an I:E ratio that is close to 1:1.
Collapse
Affiliation(s)
- Tom Barnes
- University of Greenwich, Park Row, London SE10 9LE, United Kingdom.
| | - Dirk van Asseldonk
- Ventinova Medical, Meerenakkerplein 7, 5652 BJ Eindhoven, The Netherlands
| | - Dietmar Enk
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Münster (UKM), Albert-Schweitzer-Campus 1, 48149 Münster, Germany; University of Greenwich, Park Row, London SE10 9LE, United Kingdom
| |
Collapse
|
198
|
Richard C, Shabbir W, Ferraro P, Massé C, Berthiaume Y. Alveolar liquid clearance in lung injury: Evaluation of the impairment of the β 2-adrenergic agonist response in an ischemia-reperfusion lung injury model. Respir Physiol Neurobiol 2018; 259:104-110. [PMID: 30171906 DOI: 10.1016/j.resp.2018.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 07/27/2018] [Accepted: 08/27/2018] [Indexed: 01/05/2023]
Abstract
While alveolar liquid clearance (ALC) mediated by the β2-adrenergic receptor (β2-AR) plays an important role in lung edema resolution in certain models of lung injury, in more severe lung injury models, this response might disappear. Indeed, we have shown that in an ischemia-reperfusion-induced lung injury model, β2-agonists do not enhance ALC. The objective of this study was to determine if downregulation of the β2-AR could explain the lack of response to β2-agonists in this lung injury model. In an in vivo canine model of lung transplantation, we observed no change in β2-AR concentration or affinity in the injured transplanted lungs compared to the native lungs. Furthermore, we could not enhance ALC in transplanted lungs with dcAMP + aminophylline, a treatment that bypasses the β2-adrenergic receptor and is known to stimulate ALC in normal lungs. However, transplantation decreased αENaC expression in the lungs by 50%. We conclude that the lack of response to β2-agonists in ischemia-reperfusion-induced lung injury is not associated with significant downregulation of the β2-adrenergic receptors but is attributable to decreased expression of the ENaC channel, which is essential for sodium transport and alveolar liquid clearance in the lung.
Collapse
Affiliation(s)
- Chloé Richard
- Centre de recherche, Centre hospitalier de l'université de Montréal (CHUM), Canada
| | - Waheed Shabbir
- Institute of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Pasquale Ferraro
- Centre de recherche, Centre hospitalier de l'université de Montréal (CHUM), Canada; Département de chirurgie, Université de Montréal, Montréal, Québec, Canada
| | - Chantal Massé
- Centre de recherche, Centre hospitalier de l'université de Montréal (CHUM), Canada; Institut de recherches cliniques de Montréal (IRCM), Montréal, Quebec, Canada
| | - Yves Berthiaume
- Centre de recherche, Centre hospitalier de l'université de Montréal (CHUM), Canada; Département de médecine, Université de Montréal, Montréal, Québec, Canada; Institut de recherches cliniques de Montréal (IRCM), Montréal, Quebec, Canada.
| |
Collapse
|
199
|
Salvage therapies for refractory hypoxemia in ARDS. Respir Med 2018; 141:150-158. [PMID: 30053961 DOI: 10.1016/j.rmed.2018.06.030] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/15/2018] [Accepted: 06/29/2018] [Indexed: 02/06/2023]
Abstract
Acute Respiratory Distress Syndrome (ARDS) is a condition of varied etiology characterized by the acute onset (within 1 week of the inciting event) of hypoxemia, reduced lung compliance, diffuse lung inflammation and bilateral opacities on chest imaging attributable to noncardiogenic (increased permeability) pulmonary edema. Although multi-organ failure is the most common cause of death in ARDS, an estimated 10-15% of the deaths in ARDS are caused due to refractory hypoxemia, i.e.- hypoxemia despite lung protective conventional ventilator modes. In these cases, clinicians may resort to other measures with less robust evidence -referred to as "salvage therapies". These include proning, 48 h of paralysis early in the course of ARDS, various recruitment maneuvers, unconventional ventilator modes, inhaled pulmonary vasodilators, and Extracorporeal membrane oxygenation (ECMO). All the salvage therapies described have been associated with improved oxygenation, but with the exception of proning and 48 h of paralysis early in the course of ARDS, none of them have a proven mortality benefit. Based on the current evidence, no salvage therapy has been shown to be superior to the others and each of them is associated with its own risks and benefits. Hence, the order of application of these therapies varies in different institutions and should be applied following a risk-benefit analysis specific to the patient and local experience. This review explores the rationale, evidence, advantages and risks behind each of these strategies.
Collapse
|
200
|
Wang X, Luo B, Lu Y, Pang D, Zheng J, Mo J, Huang H, Feng J. The triggering receptor expressed by myeloid cells-1 activates TLR4-MyD88-NF-κB-dependent signaling to aggravate ventilation-induced lung inflammation and injury in mice. Cell Tissue Res 2018; 374:137-148. [PMID: 29869715 DOI: 10.1007/s00441-018-2853-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 04/17/2018] [Accepted: 05/04/2018] [Indexed: 01/09/2023]
Abstract
The triggering receptor expressed by myeloid cells-1 (TREM-1) plays an important role in infectious and autoimmune diseases but how it contributes to ventilation-induced lung injury (VILI) and inflammation is unclear. Here, we examine the possibility that TREM-1 activates signaling dependent on Toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (Myd88) and nuclear factor (NF)-κB, which leads in turn to VILI. In a mouse model of VILI, which we validated based on lung edema and histopathology as well as cytokine levels, we examine mRNA and protein levels of TREM-1, TLR4, MyD88, NF-κB and its inhibitory protein I-κB in animals subjected to ventilation at normal or high tidal volume. The extent of lung edema, injury and inflammation were higher in the high tidal volume animals, as were the expression levels of all proteins examined. Treatment with TREM-1 agonist aggravated these effects, whereas treatment with TREM-1 antagonist attenuated them. Our results suggest that aggravation of VILI by TREM-1 in mice may be associated with TLR4-MyD88-NF-κB-dependent signaling.
Collapse
Affiliation(s)
- Xiaoxia Wang
- Department of Anesthesiology, The Maternal and & Child Health Hospital, The Children's Hospital, The Obstetrics & Gynecology Hospital of Guangxi Zhuang Autonomous Region, Xiang Zhu Rd No. 59, Nanning, 530002, People's Republic of China
| | - Bijun Luo
- Department of Anesthesiology, The Maternal and & Child Health Hospital, The Children's Hospital, The Obstetrics & Gynecology Hospital of Guangxi Zhuang Autonomous Region, Xiang Zhu Rd No. 59, Nanning, 530002, People's Republic of China
| | - Yanyan Lu
- Department of Anesthesiology, The Maternal and & Child Health Hospital, The Children's Hospital, The Obstetrics & Gynecology Hospital of Guangxi Zhuang Autonomous Region, Xiang Zhu Rd No. 59, Nanning, 530002, People's Republic of China
| | - Dengge Pang
- Department of Anesthesiology, The Maternal and & Child Health Hospital, The Children's Hospital, The Obstetrics & Gynecology Hospital of Guangxi Zhuang Autonomous Region, Xiang Zhu Rd No. 59, Nanning, 530002, People's Republic of China
| | - Jianqiu Zheng
- Department of Anesthesiology, The Maternal and & Child Health Hospital, The Children's Hospital, The Obstetrics & Gynecology Hospital of Guangxi Zhuang Autonomous Region, Xiang Zhu Rd No. 59, Nanning, 530002, People's Republic of China
| | - Jianlan Mo
- Department of Anesthesiology, The Maternal and & Child Health Hospital, The Children's Hospital, The Obstetrics & Gynecology Hospital of Guangxi Zhuang Autonomous Region, Xiang Zhu Rd No. 59, Nanning, 530002, People's Republic of China
| | - Hui Huang
- Department of Anesthesiology, The Maternal and & Child Health Hospital, The Children's Hospital, The Obstetrics & Gynecology Hospital of Guangxi Zhuang Autonomous Region, Xiang Zhu Rd No. 59, Nanning, 530002, People's Republic of China
| | - Jifeng Feng
- Department of Anesthesiology, The Maternal and & Child Health Hospital, The Children's Hospital, The Obstetrics & Gynecology Hospital of Guangxi Zhuang Autonomous Region, Xiang Zhu Rd No. 59, Nanning, 530002, People's Republic of China.
| |
Collapse
|