1
|
Nosaka N, Borges V, Martinon D, Crother TR, Arditi M, Shimada K. Hypothermia protects against ventilator-induced lung injury by limiting IL-1β release and NETs formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.02.610778. [PMID: 40236184 PMCID: PMC11996356 DOI: 10.1101/2024.09.02.610778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Although mechanical ventilation is a critical intervention for acute respiratory distress syndrome (ARDS), it can trigger an IL-1β-associated complication known as ventilator-induced lung injury. In mice, we found that LPS and high-volume ventilation, LPS-HVV, leads to hypoxemia with neutrophil extracellular traps (NETs) formation in the alveoli. Furthermore, Il1r1 -/- LPS-HVV mice did not develop hypoxemia and had reduced NETs, indicating that IL-1R1 signaling is important for NETs formation and hypoxemia. Therapeutic hypothermia (TH) is known to reduce the release of inflammatory mediators. In LPS-HVV mice, TH (32 °C body temperature) prevented hypoxemia development, reducing albumin leakage, IL-1β, gasdermin D (GSDMD) and NETs formation. We also observed that LPS-primed macrophages, when stimulated at 32°C with ATP or nigericin, release less IL-1β associated with reduced GSDMD cleavage. Thus, hypothermia is an important modulating factor in the NLRP3 inflammasome activation, IL-1β release and NETs formation, preventing LPS-HVV-induced acute respiratory failure.
Collapse
|
2
|
Manji A, Wang L, Pape CM, McCaig LA, Troitskaya A, Batnyam O, McDonald LJ, Appleton CT, Veldhuizen RA, Gill SE. Effect of aging on pulmonary cellular responses during mechanical ventilation. JCI Insight 2025; 10:e185834. [PMID: 39946196 PMCID: PMC11949020 DOI: 10.1172/jci.insight.185834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 01/28/2025] [Indexed: 03/25/2025] Open
Abstract
Acute respiratory distress syndrome (ARDS) results in substantial morbidity and mortality, especially in elderly people. Mechanical ventilation, a common supportive treatment for ARDS, is necessary for maintaining gas exchange but can also propagate injury. We hypothesized that aging leads to alterations in surfactant function, inflammatory signaling, and microvascular permeability within the lung during mechanical ventilation. Young and aged male mice were mechanically ventilated, and surfactant function, inflammation, and vascular permeability were assessed. Additionally, single-cell RNA-Seq was used to delineate cell-specific transcriptional changes. The results showed that, in aged mice, surfactant dysfunction and vascular permeability were significantly augmented, while inflammation was less pronounced. Differential gene expression and pathway analyses revealed that alveolar macrophages in aged mice showed a blunted inflammatory response, while aged endothelial cells exhibited altered cell-cell junction formation. In vitro functional analysis revealed that aged endothelial cells had an impaired ability to form a barrier. These results highlight the complex interplay between aging and mechanical ventilation, including an age-related predisposition to endothelial barrier dysfunction, due to altered cell-cell junction formation, and decreased inflammation, potentially due to immune exhaustion. It is concluded that age-related vascular changes may underlie the increased susceptibility to injury during mechanical ventilation in elderly patients.
Collapse
Affiliation(s)
- Aminmohamed Manji
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Physiology and Pharmacology
| | - Lefeng Wang
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Medicine, and
| | - Cynthia M. Pape
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Medicine, and
| | - Lynda A. McCaig
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Medicine, and
| | - Alexandra Troitskaya
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Physiology and Pharmacology
| | - Onon Batnyam
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
| | - Leah J.J. McDonald
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | | | - Ruud A.W. Veldhuizen
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Physiology and Pharmacology
- Department of Medicine, and
| | - Sean E. Gill
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Physiology and Pharmacology
- Department of Medicine, and
| |
Collapse
|
3
|
Brokken T, Hütten MC, Ophelders DRMG, van Gorp C, Wolfs TGAM, Wald M. Optimized lung expansion ventilation modulates ventilation-induced lung injury in preterm lambs. Pediatr Pulmonol 2024; 59:2891-2900. [PMID: 38958257 DOI: 10.1002/ppul.27153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 06/03/2024] [Accepted: 06/14/2024] [Indexed: 07/04/2024]
Abstract
INTRODUCTION Preterm infants close to viability commonly require mechanical ventilation (MV) for respiratory distress syndrome. Despite commonly used lung-sparing ventilation techniques, rapid lung expansion during MV induces lung injury, a risk factor for bronchopulmonary dysplasia. This study investigates whether ventilation with optimized lung expansion is feasible and whether it can further minimize lung injury. Therefore, optimized lung expansion ventilation (OLEV) was compared to conventional volume targeted ventilation. METHODS Twenty preterm lambs were surgically delivered after 132 days of gestation. Nine animals were randomized to receive OLEV for 24 h, and seven received standard MV. Four unventilated animals served as controls (NV). Lungs were sampled for histological analysis at the end of the experimental period. RESULTS Ventilation with OLEV was feasible, resulting in a significantly higher mean ventilation pressure (0.7-1.3 mbar). Temporary differences in oxygenation between OLEV and MV did not reach clinically relevant levels. Ventilation in general tended to result in higher lung injury scores compared to NV, without differences between OLEV and MV. While pro-inflammatory tumor necrosis factor-α messenger RNA (mRNA) levels increased in both ventilation groups compared to NV, only animals in the MV group showed a higher number of CD45-positive cells in the lung. In contrast, mean (standard deviations) surfactant protein-B mRNA levels were significantly lower in OLEV, 0.63 (0.38) compared to NV 1.03 (0.32) (p = .023, one-way analysis of variance). CONCLUSION In conclusion, a small reduction in pulmonary inflammation after 24 h of support with OLEV suggests potential to reduce preterm lung injury.
Collapse
Affiliation(s)
- Tim Brokken
- Divisions of Neonatology, MosaKids Children's Hospital, Maastricht University Medical Center, Maastricht, The Netherlands
- GROW School for Oncology and Reproduction, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Matthias C Hütten
- Divisions of Neonatology, MosaKids Children's Hospital, Maastricht University Medical Center, Maastricht, The Netherlands
- GROW School for Oncology and Reproduction, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Daan R M G Ophelders
- Divisions of Neonatology, MosaKids Children's Hospital, Maastricht University Medical Center, Maastricht, The Netherlands
- GROW School for Oncology and Reproduction, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Charlotte van Gorp
- Divisions of Neonatology, MosaKids Children's Hospital, Maastricht University Medical Center, Maastricht, The Netherlands
- GROW School for Oncology and Reproduction, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Tim G A M Wolfs
- Divisions of Neonatology, MosaKids Children's Hospital, Maastricht University Medical Center, Maastricht, The Netherlands
- GROW School for Oncology and Reproduction, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Martin Wald
- Divisions of Neonatology, Department of Pediatrics and Adolescent Medicine, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
4
|
Al-Khalisy H, Nieman GF, Kollisch-Singule M, Andrews P, Camporota L, Shiber J, Manougian T, Satalin J, Blair S, Ghosh A, Herrmann J, Kaczka DW, Gaver DP, Bates JHT, Habashi NM. Time-Controlled Adaptive Ventilation (TCAV): a personalized strategy for lung protection. Respir Res 2024; 25:37. [PMID: 38238778 PMCID: PMC10797864 DOI: 10.1186/s12931-023-02615-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 11/25/2023] [Indexed: 01/22/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) alters the dynamics of lung inflation during mechanical ventilation. Repetitive alveolar collapse and expansion (RACE) predisposes the lung to ventilator-induced lung injury (VILI). Two broad approaches are currently used to minimize VILI: (1) low tidal volume (LVT) with low-moderate positive end-expiratory pressure (PEEP); and (2) open lung approach (OLA). The LVT approach attempts to protect already open lung tissue from overdistension, while simultaneously resting collapsed tissue by excluding it from the cycle of mechanical ventilation. By contrast, the OLA attempts to reinflate potentially recruitable lung, usually over a period of seconds to minutes using higher PEEP used to prevent progressive loss of end-expiratory lung volume (EELV) and RACE. However, even with these protective strategies, clinical studies have shown that ARDS-related mortality remains unacceptably high with a scarcity of effective interventions over the last two decades. One of the main limitations these varied interventions demonstrate to benefit is the observed clinical and pathologic heterogeneity in ARDS. We have developed an alternative ventilation strategy known as the Time Controlled Adaptive Ventilation (TCAV) method of applying the Airway Pressure Release Ventilation (APRV) mode, which takes advantage of the heterogeneous time- and pressure-dependent collapse and reopening of lung units. The TCAV method is a closed-loop system where the expiratory duration personalizes VT and EELV. Personalization of TCAV is informed and tuned with changes in respiratory system compliance (CRS) measured by the slope of the expiratory flow curve during passive exhalation. Two potentially beneficial features of TCAV are: (i) the expiratory duration is personalized to a given patient's lung physiology, which promotes alveolar stabilization by halting the progressive collapse of alveoli, thereby minimizing the time for the reopened lung to collapse again in the next expiration, and (ii) an extended inspiratory phase at a fixed inflation pressure after alveolar stabilization gradually reopens a small amount of tissue with each breath. Subsequently, densely collapsed regions are slowly ratcheted open over a period of hours, or even days. Thus, TCAV has the potential to minimize VILI, reducing ARDS-related morbidity and mortality.
Collapse
Affiliation(s)
| | - Gary F Nieman
- SUNY Upstate Medical University, 750 E. Adams St., Syracuse, NY, 13210, USA
| | | | - Penny Andrews
- R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, MD, USA
| | - Luigi Camporota
- Health Centre for Human and Applied Physiological Sciences, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Joseph Shiber
- University of Florida College of Medicine, Jacksonville, FL, USA
| | | | - Joshua Satalin
- SUNY Upstate Medical University, 750 E. Adams St., Syracuse, NY, 13210, USA.
| | - Sarah Blair
- SUNY Upstate Medical University, 750 E. Adams St., Syracuse, NY, 13210, USA
| | - Auyon Ghosh
- SUNY Upstate Medical University, 750 E. Adams St., Syracuse, NY, 13210, USA
| | | | | | | | | | - Nader M Habashi
- R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, MD, USA
| |
Collapse
|
5
|
Nieman GF, Kaczka DW, Andrews PL, Ghosh A, Al-Khalisy H, Camporota L, Satalin J, Herrmann J, Habashi NM. First Stabilize and then Gradually Recruit: A Paradigm Shift in Protective Mechanical Ventilation for Acute Lung Injury. J Clin Med 2023; 12:4633. [PMID: 37510748 PMCID: PMC10380509 DOI: 10.3390/jcm12144633] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is associated with a heterogeneous pattern of injury throughout the lung parenchyma that alters regional alveolar opening and collapse time constants. Such heterogeneity leads to atelectasis and repetitive alveolar collapse and expansion (RACE). The net effect is a progressive loss of lung volume with secondary ventilator-induced lung injury (VILI). Previous concepts of ARDS pathophysiology envisioned a two-compartment system: a small amount of normally aerated lung tissue in the non-dependent regions (termed "baby lung"); and a collapsed and edematous tissue in dependent regions. Based on such compartmentalization, two protective ventilation strategies have been developed: (1) a "protective lung approach" (PLA), designed to reduce overdistension in the remaining aerated compartment using a low tidal volume; and (2) an "open lung approach" (OLA), which first attempts to open the collapsed lung tissue over a short time frame (seconds or minutes) with an initial recruitment maneuver, and then stabilize newly recruited tissue using titrated positive end-expiratory pressure (PEEP). A more recent understanding of ARDS pathophysiology identifies regional alveolar instability and collapse (i.e., hidden micro-atelectasis) in both lung compartments as a primary VILI mechanism. Based on this understanding, we propose an alternative strategy to ventilating the injured lung, which we term a "stabilize lung approach" (SLA). The SLA is designed to immediately stabilize the lung and reduce RACE while gradually reopening collapsed tissue over hours or days. At the core of SLA is time-controlled adaptive ventilation (TCAV), a method to adjust the parameters of the airway pressure release ventilation (APRV) modality. Since the acutely injured lung at any given airway pressure requires more time for alveolar recruitment and less time for alveolar collapse, SLA adjusts inspiratory and expiratory durations and inflation pressure levels. The TCAV method SLA reverses the open first and stabilize second OLA method by: (i) immediately stabilizing lung tissue using a very brief exhalation time (≤0.5 s), so that alveoli simply do not have sufficient time to collapse. The exhalation duration is personalized and adaptive to individual respiratory mechanical properties (i.e., elastic recoil); and (ii) gradually recruiting collapsed lung tissue using an inflate and brake ratchet combined with an extended inspiratory duration (4-6 s) method. Translational animal studies, clinical statistical analysis, and case reports support the use of TCAV as an efficacious lung protective strategy.
Collapse
Affiliation(s)
- Gary F. Nieman
- Department of Surgery, Upstate Medical University, Syracuse, NY 13210, USA;
| | - David W. Kaczka
- Departments of Anesthesia, Radiology and Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Penny L. Andrews
- Department of Medicine, R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, MD 21201, USA
| | - Auyon Ghosh
- Department of Medicine, Upstate Medical University, Syracuse, NY 13210, USA
| | - Hassan Al-Khalisy
- Brody School of Medicine, Department of Internal Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Luigi Camporota
- Department of Adult Critical Care, Guy’s and St Thomas’ NHS Foundation Trust, King’s Partners, St Thomas’ Hospital, London SE1 7EH, UK
| | - Joshua Satalin
- Department of Surgery, Upstate Medical University, Syracuse, NY 13210, USA;
| | - Jacob Herrmann
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Nader M. Habashi
- Department of Medicine, R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, MD 21201, USA
| |
Collapse
|
6
|
Barbeta E, Arrieta M, Motos A, Bobi J, Yang H, Yang M, Tanzella G, Di Ginnatale P, Nogas S, Vargas CR, Cabrera R, Battaglini D, Meli A, Kiarostami K, Vázquez N, Fernández-Barat L, Rigol M, Mellado-Artigas R, Frigola G, Camprubí-Rimblas M, Ferrer P, Martinez D, Artigas A, Ferrando C, Ferrer M, Torres A. A long-lasting porcine model of ARDS caused by pneumonia and ventilator-induced lung injury. Crit Care 2023; 27:239. [PMID: 37328874 PMCID: PMC10276390 DOI: 10.1186/s13054-023-04512-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/30/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND Animal models of acute respiratory distress syndrome (ARDS) do not completely resemble human ARDS, struggling translational research. We aimed to characterize a porcine model of ARDS induced by pneumonia-the most common risk factor in humans-and analyze the additional effect of ventilator-induced lung injury (VILI). METHODS Bronchoscopy-guided instillation of a multidrug-resistant Pseudomonas aeruginosa strain was performed in ten healthy pigs. In six animals (pneumonia-with-VILI group), pulmonary damage was further increased by VILI applied 3 h before instillation and until ARDS was diagnosed by PaO2/FiO2 < 150 mmHg. Four animals (pneumonia-without-VILI group) were protectively ventilated 3 h before inoculum and thereafter. Gas exchange, respiratory mechanics, hemodynamics, microbiological studies and inflammatory markers were analyzed during the 96-h experiment. During necropsy, lobar samples were also analyzed. RESULTS All animals from pneumonia-with-VILI group reached Berlin criteria for ARDS diagnosis until the end of experiment. The mean duration under ARDS diagnosis was 46.8 ± 7.7 h; the lowest PaO2/FiO2 was 83 ± 5.45 mmHg. The group of pigs that were not subjected to VILI did not meet ARDS criteria, even when presenting with bilateral pneumonia. Animals developing ARDS presented hemodynamic instability as well as severe hypercapnia despite high-minute ventilation. Unlike the pneumonia-without-VILI group, the ARDS animals presented lower static compliance (p = 0.011) and increased pulmonary permeability (p = 0.013). The highest burden of P. aeruginosa was found at pneumonia diagnosis in all animals, as well as a high inflammatory response shown by a release of interleukin (IL)-6 and IL-8. At histological examination, only animals comprising the pneumonia-with-VILI group presented signs consistent with diffuse alveolar damage. CONCLUSIONS In conclusion, we established an accurate pulmonary sepsis-induced ARDS model.
Collapse
Affiliation(s)
- Enric Barbeta
- Surgical Intensive Care Unit, Hospital Clínic de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona (UB), Barcelona, Spain
| | - Marta Arrieta
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona (UB), Barcelona, Spain
| | - Ana Motos
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain.
- University of Barcelona (UB), Barcelona, Spain.
| | - Joaquim Bobi
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona (UB), Barcelona, Spain
- Department of Cardiology, Erasmus MC, University Medical Center Rotterdam, 3015, Rotterdam, The Netherlands
- Cardiology Department, Institute Clinic Cardiovascular (ICCV), Hospital Clinic, Barcelona, Spain
| | - Hua Yang
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona (UB), Barcelona, Spain
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, China
| | - Minlan Yang
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona (UB), Barcelona, Spain
- Department of Infectious Diseases, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Giacomo Tanzella
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Department of Anesthesia and Intensive Care, IRCCS for Oncology and Neurosciences, San Martino Policlinico Hospital, Genoa, Italy
| | - Pierluigi Di Ginnatale
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Department of Anesthesiology, Critical Care Medicine and Emergency, SS. Annunziata Hospital, Chieti, Italy
| | - Stefano Nogas
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Department of Anesthesia and Intensive Care, IRCCS for Oncology and Neurosciences, San Martino Policlinico Hospital, Genoa, Italy
| | - Carmen Rosa Vargas
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona (UB), Barcelona, Spain
| | - Roberto Cabrera
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Denise Battaglini
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona (UB), Barcelona, Spain
- Department of Anesthesia and Intensive Care, IRCCS for Oncology and Neurosciences, San Martino Policlinico Hospital, Genoa, Italy
| | - Andrea Meli
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Department of Anesthesia and Intensive Care, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Kasra Kiarostami
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona (UB), Barcelona, Spain
| | - Nil Vázquez
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona (UB), Barcelona, Spain
| | - Laia Fernández-Barat
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona (UB), Barcelona, Spain
| | - Montserrat Rigol
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona (UB), Barcelona, Spain
- Cardiology Department, Institute Clinic Cardiovascular (ICCV), Hospital Clinic, Barcelona, Spain
| | - Ricard Mellado-Artigas
- Surgical Intensive Care Unit, Hospital Clínic de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Gerard Frigola
- Critical Care Center, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Marta Camprubí-Rimblas
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Critical Care Center, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Pau Ferrer
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Daniel Martinez
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Department of Pathology, Hospital Clinic, Barcelona, Spain
| | - Antonio Artigas
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Critical Care Center, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Carlos Ferrando
- Surgical Intensive Care Unit, Hospital Clínic de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona (UB), Barcelona, Spain
| | - Miquel Ferrer
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona (UB), Barcelona, Spain
- Pneumology Service, Respiratory Institute, Hospital Clinic of Barcelona, Villarroel st. 170, 08036, Barcelona, Spain
| | - Antoni Torres
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain.
- University of Barcelona (UB), Barcelona, Spain.
- Pneumology Service, Respiratory Institute, Hospital Clinic of Barcelona, Villarroel st. 170, 08036, Barcelona, Spain.
| |
Collapse
|
7
|
Ramcharran H, Bates JHT, Satalin J, Blair S, Andrews PL, Gaver DP, Gatto LA, Wang G, Ghosh AJ, Robedee B, Vossler J, Habashi NM, Daphtary N, Kollisch-Singule M, Nieman GF. Protective ventilation in a pig model of acute lung injury: timing is as important as pressure. J Appl Physiol (1985) 2022; 133:1093-1105. [PMID: 36135956 PMCID: PMC9621707 DOI: 10.1152/japplphysiol.00312.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/26/2022] [Accepted: 09/19/2022] [Indexed: 11/22/2022] Open
Abstract
Ventilator-induced lung injury (VILI) is a significant risk for patients with acute respiratory distress syndrome (ARDS). Management of the patient with ARDS is currently dominated by the use of low tidal volume mechanical ventilation, the presumption being that this mitigates overdistension (OD) injury to the remaining normal lung tissue. Evidence exists, however, that it may be more important to avoid cyclic recruitment and derecruitment (RD) of lung units, although the relative roles of OD and RD in VILI remain unclear. Forty pigs had a heterogeneous lung injury induced by Tween instillation and were randomized into four groups (n = 10 each) with higher (↑) or lower (↓) levels of OD and/or RD imposed using airway pressure release ventilation (APRV). OD was increased by setting inspiratory airway pressure to 40 cmH2O and lessened with 28 cmH2O. RD was attenuated using a short duration of expiration (∼0.45 s) and increased with a longer duration (∼1.0 s). All groups developed mild ARDS following injury. RD ↑ OD↑ caused the greatest degree of lung injury as determined by [Formula: see text]/[Formula: see text] ratio (226.1 ± 41.4 mmHg). RD ↑ OD↓ ([Formula: see text]/[Formula: see text]= 333.9 ± 33.1 mmHg) and RD ↓ OD↑ ([Formula: see text]/[Formula: see text] = 377.4 ± 43.2 mmHg) were both moderately injurious, whereas RD ↓ OD↓ ([Formula: see text]/[Formula: see text] = 472.3 ± 22.2 mmHg; P < 0.05) was least injurious. Both tidal volume and driving pressure were essentially identical in the RD ↑ OD↓ and RD ↓ OD↑ groups. We, therefore, conclude that considerations of expiratory time may be at least as important as pressure for safely ventilating the injured lung.NEW & NOTEWORTHY In a large animal model of ARDS, recruitment/derecruitment caused greater VILI than overdistension, whereas both mechanisms together caused severe lung damage. These findings suggest that eliminating cyclic recruitment and derecruitment during mechanical ventilation should be a preeminent management goal for the patient with ARDS. The airway pressure release ventilation (APRV) mode of mechanical ventilation can achieve this if delivered with an expiratory duration (TLow) that is brief enough to prevent derecruitment at end expiration.
Collapse
Affiliation(s)
| | | | | | - Sarah Blair
- SUNY Upstate Medical University, Syracuse, New York
| | | | | | | | - Guirong Wang
- SUNY Upstate Medical University, Syracuse, New York
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Nieman G, Kollisch-Singule M, Ramcharran H, Satalin J, Blair S, Gatto LA, Andrews P, Ghosh A, Kaczka DW, Gaver D, Bates J, Habashi NM. Unshrinking the baby lung to calm the VILI vortex. Crit Care 2022; 26:242. [PMID: 35934707 PMCID: PMC9357329 DOI: 10.1186/s13054-022-04105-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/12/2022] [Indexed: 02/07/2023] Open
Abstract
A hallmark of ARDS is progressive shrinking of the ‘baby lung,’ now referred to as the ventilator-induced lung injury (VILI) ‘vortex.’ Reducing the risk of the VILI vortex is the goal of current ventilation strategies; unfortunately, this goal has not been achieved nor has mortality been reduced. However, the temporal aspects of a mechanical breath have not been considered. A brief expiration prevents alveolar collapse, and an extended inspiration can recruit the atelectatic lung over hours. Time-controlled adaptive ventilation (TCAV) is a novel ventilator approach to achieve these goals, since it considers many of the temporal aspects of dynamic lung mechanics.
Collapse
Affiliation(s)
- Gary Nieman
- Department of Surgery, SUNY Upstate Medical Center, SUNY Upstate, 750 East Adams St., Syracuse, NY, 13210, USA
| | - Michaela Kollisch-Singule
- Department of Surgery, SUNY Upstate Medical Center, SUNY Upstate, 750 East Adams St., Syracuse, NY, 13210, USA
| | - Harry Ramcharran
- Department of Surgery, SUNY Upstate Medical Center, SUNY Upstate, 750 East Adams St., Syracuse, NY, 13210, USA
| | - Joshua Satalin
- Department of Surgery, SUNY Upstate Medical Center, SUNY Upstate, 750 East Adams St., Syracuse, NY, 13210, USA.
| | - Sarah Blair
- Department of Surgery, SUNY Upstate Medical Center, SUNY Upstate, 750 East Adams St., Syracuse, NY, 13210, USA
| | - Louis A Gatto
- Department of Surgery, SUNY Upstate Medical Center, SUNY Upstate, 750 East Adams St., Syracuse, NY, 13210, USA
| | - Penny Andrews
- Department of Medicine, University of Maryland, Baltimore, MD, USA
| | - Auyon Ghosh
- Department of Surgery, SUNY Upstate Medical Center, SUNY Upstate, 750 East Adams St., Syracuse, NY, 13210, USA
| | - David W Kaczka
- Departments of Anesthesia, Biomedical Engineering, and Radiology, University of Iowa, Iowa City, IA, USA
| | - Donald Gaver
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, USA
| | - Jason Bates
- Department of Medicine, University of Vermont, Burlington, VT, USA
| | - Nader M Habashi
- Department of Medicine, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
9
|
Keskinidou C, Vassiliou AG, Dimopoulou I, Kotanidou A, Orfanos SE. Mechanistic Understanding of Lung Inflammation: Recent Advances and Emerging Techniques. J Inflamm Res 2022; 15:3501-3546. [PMID: 35734098 PMCID: PMC9207257 DOI: 10.2147/jir.s282695] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 05/04/2022] [Indexed: 12/12/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening lung injury characterized by an acute inflammatory response in the lung parenchyma. Hence, it is considered as the most appropriate clinical syndrome to study pathogenic mechanisms of lung inflammation. ARDS is associated with increased morbidity and mortality in the intensive care unit (ICU), while no effective pharmacological treatment exists. It is very important therefore to fully characterize the underlying pathobiology and the related mechanisms, in order to develop novel therapeutic approaches. In vivo and in vitro models are important pre-clinical tools in biological and medical research in the mechanistic and pathological understanding of the majority of diseases. In this review, we will present data from selected experimental models of lung injury/acute lung inflammation, which have been based on clinical disorders that can lead to the development of ARDS and related inflammatory lung processes in humans, including ventilation-induced lung injury (VILI), sepsis, ischemia/reperfusion, smoke, acid aspiration, radiation, transfusion-related acute lung injury (TRALI), influenza, Streptococcus (S.) pneumoniae and coronaviruses infection. Data from the corresponding clinical conditions will also be presented. The mechanisms related to lung inflammation that will be covered are oxidative stress, neutrophil extracellular traps, mitogen-activated protein kinase (MAPK) pathways, surfactant, and water and ion channels. Finally, we will present a brief overview of emerging techniques in the field of omics research that have been applied to ARDS research, encompassing genomics, transcriptomics, proteomics, and metabolomics, which may recognize factors to help stratify ICU patients at risk, predict their prognosis, and possibly, serve as more specific therapeutic targets.
Collapse
Affiliation(s)
- Chrysi Keskinidou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Alice G Vassiliou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Ioanna Dimopoulou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Anastasia Kotanidou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Stylianos E Orfanos
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| |
Collapse
|
10
|
Rajanathan R, Pedersen TM, Thomsen MB, Botker HE, Matchkov VV. Phenylephrine-Induced Cardiovascular Changes in the Anesthetized Mouse: An Integrated Assessment of in vivo Hemodynamics Under Conditions of Controlled Heart Rate. Front Physiol 2022; 13:831724. [PMID: 35250634 PMCID: PMC8891648 DOI: 10.3389/fphys.2022.831724] [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: 12/08/2021] [Accepted: 01/27/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Investigating the cardiovascular system is challenging due to its complex regulation by humoral and neuronal factors. Despite this complexity, many existing research methods are limited to the assessment of a few parameters leading to an incomplete characterization of cardiovascular function. Thus, we aim to establish a murine in vivo model for integrated assessment of the cardiovascular system under conditions of controlled heart rate. Utilizing this model, we assessed blood pressure, cardiac output, stroke volume, total peripheral resistance, and electrocardiogram (ECG). Hypothesis We hypothesize that (i) our in vivo model can be utilized to investigate cardiac and vascular responses to pharmacological intervention with the α1-agonist phenylephrine, and (ii) we can study cardiovascular function during artificial pacing of the heart, modulating cardiac function without a direct vascular effect. Methods We included 12 mice that were randomly assigned to either vehicle or phenylephrine intervention through intraperitoneal administration. Mice were anesthetized with isoflurane and intubated endotracheally for mechanical ventilation. We measured blood pressure via a solid-state catheter in the aortic arch, blood flow via a probe on the ascending aorta, and ECG from needle electrodes on the extremities. Right atrium was electrically paced at a frequency ranging from 10 to 11.3 Hz before and after either vehicle or phenylephrine administration. Results Phenylephrine significantly increased blood pressure, stroke volume, and total peripheral resistance compared to the vehicle group. Moreover, heart rate was significantly decreased following phenylephrine administration. Pacing significantly decreased stroke volume and cardiac output both prior to and after drug administration. However, phenylephrine-induced changes in blood pressure and total peripheral resistance were maintained with increasing pacing frequencies compared to the vehicle group. Total peripheral resistance was not significantly altered with increasing pacing frequencies suggesting that the effect of phenylephrine is primarily of vascular origin. Conclusion In conclusion, this in vivo murine model is capable of distinguishing between changes in peripheral vascular and cardiac functions. This study underlines the primary effect of phenylephrine on vascular function with secondary changes to cardiac function. Hence, this in vivo model is useful for the integrated assessment of the cardiovascular system.
Collapse
Affiliation(s)
- Rajkumar Rajanathan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- *Correspondence: Rajkumar Rajanathan,
| | | | - Morten B. Thomsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hans Erik Botker
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
11
|
von Knethen A, Heinicke U, Laux V, Parnham MJ, Steinbicker AU, Zacharowski K. Antioxidants as Therapeutic Agents in Acute Respiratory Distress Syndrome (ARDS) Treatment-From Mice to Men. Biomedicines 2022; 10:98. [PMID: 35052778 PMCID: PMC8773193 DOI: 10.3390/biomedicines10010098] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/26/2021] [Accepted: 12/31/2021] [Indexed: 12/16/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a major cause of patient mortality in intensive care units (ICUs) worldwide. Considering that no causative treatment but only symptomatic care is available, it is obvious that there is a high unmet medical need for a new therapeutic concept. One reason for a missing etiologic therapy strategy is the multifactorial origin of ARDS, which leads to a large heterogeneity of patients. This review summarizes the various kinds of ARDS onset with a special focus on the role of reactive oxygen species (ROS), which are generally linked to ARDS development and progression. Taking a closer look at the data which already have been established in mouse models, this review finally proposes the translation of these results on successful antioxidant use in a personalized approach to the ICU patient as a potential adjuvant to standard ARDS treatment.
Collapse
Affiliation(s)
- Andreas von Knethen
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany
| | - Ulrike Heinicke
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Volker Laux
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany
| | - Michael J Parnham
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt, Germany
| | - Andrea U Steinbicker
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Kai Zacharowski
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| |
Collapse
|
12
|
Yuki K, Koutsogiannaki S. Translational Role of Rodent Models to Study Ventilator-Induced Lung Injury. TRANSLATIONAL PERIOPERATIVE AND PAIN MEDICINE 2021; 8:404-415. [PMID: 34993270 PMCID: PMC8729883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Mechanical ventilation is an important part of medical care in intensive care units and operating rooms to support respiration. While it is a critical component of medical care, it is well known that mechanical ventilation itself can be injurious to the lungs. Despite a large number of clinical and preclinical studies that have been done so far, there still exists a gap of knowledge regarding how to ventilate patients mechanically without increasing lung injury. Here, we will review what we have learned so far from preclinical and clinical studies and consider how to use preclinical models of ventilation-induced lung injury that better recapitulate the clinical scenarios.
Collapse
Affiliation(s)
- Koichi Yuki
- Cardiac Anesthesia Division, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, USA,Department of Anaesthesia, Harvard Medical School, USA,Corresponding Authors: Sophia Koutsogiannaki, Ph.D and Koichi Yuki, M.D., Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children’s Hospital, USA, ;
| | - Sophia Koutsogiannaki
- Cardiac Anesthesia Division, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, USA,Department of Anaesthesia, Harvard Medical School, USA,Corresponding Authors: Sophia Koutsogiannaki, Ph.D and Koichi Yuki, M.D., Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children’s Hospital, USA, ;
| |
Collapse
|
13
|
Joelsson JP, Ingthorsson S, Kricker J, Gudjonsson T, Karason S. Ventilator-induced lung-injury in mouse models: Is there a trap? Lab Anim Res 2021; 37:30. [PMID: 34715943 PMCID: PMC8554750 DOI: 10.1186/s42826-021-00108-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/20/2021] [Indexed: 12/15/2022] Open
Abstract
Ventilator-induced lung injury (VILI) is a serious acute injury to the lung tissue that can develop during mechanical ventilation of patients. Due to the mechanical strain of ventilation, damage can occur in the bronchiolar and alveolar epithelium resulting in a cascade of events that may be fatal to the patients. Patients requiring mechanical ventilation are often critically ill, which limits the possibility of obtaining patient samples, making VILI research challenging. In vitro models are very important for VILI research, but the complexity of the cellular interactions in multi-organ animals, necessitates in vivo studies where the mouse model is a common choice. However, the settings and duration of ventilation used to create VILI in mice vary greatly, causing uncertainty in interpretation and comparison of results. This review examines approaches to induce VILI in mouse models for the last 10 years, to our best knowledge, summarizing methods and key parameters presented across the studies. The results imply that a more standardized approach is warranted.
Collapse
Affiliation(s)
- Jon Petur Joelsson
- Stem Cell Research Unit, BioMedical Center, School of Health Sciences, University of Iceland, Reykjavik, Iceland. .,Department of Laboratory Hematology, Landspitali-University Hospital, Reykjavik, Iceland. .,EpiEndo Pharmaceuticals, Seltjarnarnes, Iceland.
| | - Saevar Ingthorsson
- Department of Laboratory Hematology, Landspitali-University Hospital, Reykjavik, Iceland.,Faculty of Nursing, University of Iceland, Reykjavik, Iceland
| | | | - Thorarinn Gudjonsson
- Stem Cell Research Unit, BioMedical Center, School of Health Sciences, University of Iceland, Reykjavik, Iceland.,Department of Laboratory Hematology, Landspitali-University Hospital, Reykjavik, Iceland.,EpiEndo Pharmaceuticals, Seltjarnarnes, Iceland
| | - Sigurbergur Karason
- Stem Cell Research Unit, BioMedical Center, School of Health Sciences, University of Iceland, Reykjavik, Iceland.,Intensive Care Unit, Landspitali-University Hospital, Reykjavik, Iceland
| |
Collapse
|
14
|
Koh MW, Baldi RF, Soni S, Handslip R, Tan YY, O’Dea KP, Malesevic M, McAuley DF, O’Kane CM, Patel BV, Takata M, Wilson MR. Secreted Extracellular Cyclophilin A Is a Novel Mediator of Ventilator-induced Lung Injury. Am J Respir Crit Care Med 2021; 204:421-430. [PMID: 33848447 PMCID: PMC8480235 DOI: 10.1164/rccm.202009-3545oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
Rationale: Mechanical ventilation is a mainstay of intensive care but contributes to the mortality of patients through ventilator-induced lung injury. eCypA (extracellular CypA [cyclophilin A]) is an emerging inflammatory mediator and metalloproteinase inducer, and the gene responsible for its expression has recently been linked to coronavirus disease (COVID-19). Objectives: To explore the involvement of eCypA in the pathophysiology of ventilator-induced lung injury. Methods: Mice were ventilated with a low or high Vt for up to 3 hours, with or without blockade of eCypA signaling, and lung injury and inflammation were evaluated. Human primary alveolar epithelial cells were exposed to in vitro stretching to explore the cellular source of eCypA, and CypA concentrations were measured in BAL fluid from patients with acute respiratory distress syndrome to evaluate the clinical relevance. Measurements and Main Results: High-Vt ventilation in mice provoked a rapid increase in soluble CypA concentration in the alveolar space but not in plasma. In vivo ventilation and in vitro stretching experiments indicated the alveolar epithelium as the likely major source. In vivo blockade of eCypA signaling substantially attenuated physiological dysfunction, macrophage activation, and MMPs (matrix metalloproteinases). Finally, we found that patients with acute respiratory distress syndrome showed markedly elevated concentrations of eCypA within BAL fluid. Conclusions: CypA is upregulated within the lungs of injuriously ventilated mice (and critically ill patients), where it plays a significant role in lung injury. eCypA represents an exciting novel target for pharmacological intervention.
Collapse
Affiliation(s)
- Marissa W. Koh
- Division of Anaesthetics, Pain Medicine and Intensive Care, Imperial College London, London, United Kingdom
| | - Rhianna F. Baldi
- Division of Anaesthetics, Pain Medicine and Intensive Care, Imperial College London, London, United Kingdom
| | - Sanooj Soni
- Division of Anaesthetics, Pain Medicine and Intensive Care, Imperial College London, London, United Kingdom
| | - Rhodri Handslip
- Division of Anaesthetics, Pain Medicine and Intensive Care, Imperial College London, London, United Kingdom
| | - Ying Ying Tan
- Division of Anaesthetics, Pain Medicine and Intensive Care, Imperial College London, London, United Kingdom
| | - Kieran P. O’Dea
- Division of Anaesthetics, Pain Medicine and Intensive Care, Imperial College London, London, United Kingdom
| | - Miroslav Malesevic
- Institute of Biochemistry and Biotechnology, Martin Luther University Halle-Wittenberg, Halle, Germany; and
| | - Daniel F. McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Cecilia M. O’Kane
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Brijesh V. Patel
- Division of Anaesthetics, Pain Medicine and Intensive Care, Imperial College London, London, United Kingdom
| | - Masao Takata
- Division of Anaesthetics, Pain Medicine and Intensive Care, Imperial College London, London, United Kingdom
| | - Michael R. Wilson
- Division of Anaesthetics, Pain Medicine and Intensive Care, Imperial College London, London, United Kingdom
| |
Collapse
|
15
|
Lin CK, Huang TH, Yang CT, Shi CS. Roles of lung-recruited monocytes and pulmonary Vascular Endothelial Growth Factor (VEGF) in resolving Ventilator-Induced Lung Injury (VILI). PLoS One 2021; 16:e0248959. [PMID: 33740009 PMCID: PMC7978382 DOI: 10.1371/journal.pone.0248959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 03/09/2021] [Indexed: 01/31/2023] Open
Abstract
Monocytes and vascular endothelial growth factor (VEGF) have profound effects on tissue injury and repair. In ventilator-induced lung injury (VILI), monocytes, the majority of which are Ly6C+high, and VEGF are known to initiate lung injury. However, their roles in post-VILI lung repair remain unclear. In this study, we used a two-hit mouse model of VILI to identify the phenotypes of monocytes recruited to the lungs during the resolution of VILI and investigated the contributions of monocytes and VEGF to lung repair. We found that the lung-recruited monocytes were predominantly Ly6C+low from day 1 after the insult. Meanwhile, contrary to inflammatory cytokines, pulmonary VEGF decreased upon VILI but subsequently increased significantly on days 7 and 14 after the injury. There was a strong positive correlation between VEGF expression and proliferation of alveolar epithelial cells in lung sections. The expression pattern of VEGF mRNA in lung-recruited monocytes was similar to that of pulmonary VEGF proteins, and the depletion of monocytes significantly suppressed the increase of pulmonary VEGF proteins on days 7 and 14 after VILI. In conclusion, during recovery from VILI, the temporal expression patterns of pulmonary growth factors are different from those of inflammatory cytokines, and the restoration of pulmonary VEGF by monocytes, which are mostly Ly6C+low, is associated with pulmonary epithelial proliferation. Lung-recruited monocytes and pulmonary VEGF may play crucial roles in post-VILI lung repair.
Collapse
Affiliation(s)
- Chin-Kuo Lin
- Division of Pulmonary Infection and Critical Care, Department of Pulmonary and Critical Care Medicine, Chiayi Chang Gung Memorial Hospital, Puzi City, Taiwan
- Graduate Institute of Clinical Medicine Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tzu-Hsiung Huang
- Department of Respiratory Therapy, Chiayi Chang Gung Memorial Hospital, Puzi City, Taiwan
| | - Cheng-Ta Yang
- Department of Thoracic Medicine, Taoyuan Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Respiratory Therapy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chung-Sheng Shi
- Graduate Institute of Clinical Medicine Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Colon and Rectal Surgery, Department of Surgery, Chiayi Chang Gung Memorial Hospital, Puzi City, Taiwan
| |
Collapse
|
16
|
Hajjar WM, Eldawlatly A, Alnassar SA, Ahmed I, Alghamedi A, Shakoor Z, Alrikabi AC, Hajjar AW, Ahmad AE. The effect of low versus high tidal volume ventilation on inflammatory markers in animal model undergoing lung ventilation: A prospective study. Saudi J Anaesth 2021; 15:1-6. [PMID: 33824635 PMCID: PMC8016054 DOI: 10.4103/sja.sja_650_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 06/30/2020] [Accepted: 07/05/2020] [Indexed: 11/23/2022] Open
Abstract
Background and Aims: Mechanical ventilation (MV) with high tidal volume (Vt.) may induce or aggravate lung injury in critically ill patients. It might also cause an overwhelming systemic inflammation leading to acute lung injury (ALI), diffuse alveolar damage (DAD) and multiple organ failure (MOF) with subsequent high mortality. The objective of this study was to compare the effects of different Vt. on the inflammatory markers of the broncho-alveolar lavage (BAL) fluid and lung biopsy in a group of animal model (Beagle dogs). Methods: A two-phased prospective study involving 30 Beagle dogs (15 dogs/phase), each phase divided into three groups (each 5 dogs/group). In the first phase each group received MV with Vt. of 8 (low), 10 (normal, control group), and 12 (high) ml/kg body weight (b.w.) respectively. BAL fluid was obtained at the time of induction of anesthesia immediately following tracheal intubation and one hour later following MV to count the macrophages, neutrophils and lymphocytes. In the second phase of the experiment, in addition to obtaining (BAL) fluid similar to the phase one, mini thoracotomy and lung biopsy obtained from the upper lobe of the right lung at same timings for histopathological examination study. Mann-Whitney-Wilcoxon test was used for statistical analysis of the data obtained. Results: BAL fluid analysis showed increase in the counts of macrophages and lymphocytes with Vt. of 12 ml/kg b.w. compared to the control group (10 ml/kg b.w.) (P < 0.05). in the second phase, similar findings obtained. The histopathological study of the lung tissue obtained in the second phase of the study from the group that received a high Vt. of 12 ml/kg b.w. showed significant inflammatory changes with presence of neutrophil infiltration and edema in the bronchial wall compared to the control group (10 ml/kg b.w.) (P < 0.05). Conclusions: The use of high Vt. in ventilated animal lung model may increase the risk of inflammation and subsequent damage in healthy lungs, these findings may help physicians to avoid using high Vt. in short-term mechanically ventilated patients in the operating room setting.
Collapse
Affiliation(s)
- Waseem M Hajjar
- Department of Surgery, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Abdelazeem Eldawlatly
- Department of Anesthesia, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Sami A Alnassar
- Department of Surgery, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Iftikhar Ahmed
- Department of Surgery, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Alaa Alghamedi
- Department of Surgery, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Zahid Shakoor
- Department of Pathology, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Ammar C Alrikabi
- Department of Pathology, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Adnan W Hajjar
- Department of Surgery, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Abdulaziz Ejaz Ahmad
- Department of Anesthesia, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
17
|
Low-Molecular-Weight Heparin Reduces Ventilation-Induced Lung Injury through Hypoxia Inducible Factor-1α in a Murine Endotoxemia Model. Int J Mol Sci 2020; 21:ijms21093097. [PMID: 32353952 PMCID: PMC7247708 DOI: 10.3390/ijms21093097] [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: 01/30/2020] [Revised: 04/02/2020] [Accepted: 04/27/2020] [Indexed: 12/18/2022] Open
Abstract
Patients with sepsis frequently require mechanical ventilation (MV) to survive. However, MV has been shown to induce the production of proinflammatory cytokines, causing ventilator-induced lung injury (VILI). It has been demonstrated that hypoxia-inducible factor (HIF)-1α plays a crucial role in inducing both apoptotic and inflammatory processes. Low-molecular-weight heparin (LMWH) has been shown to have anti-inflammatory activities. However, the effects of HIF-1α and LMWH on sepsis-related acute lung injury (ALI) have not been fully delineated. We hypothesized that LMWH would reduce lung injury, production of free radicals and epithelial apoptosis through the HIF-1α pathway. Male C57BL/6 mice were exposed to 6-mL/kg or 30-mL/kg MV for 5 h. Enoxaparin, 4 mg/kg, was administered subcutaneously 30 min before MV. We observed that MV with endotoxemia induced microvascular permeability; interleukin-6, tumor necrosis factor-α, macrophage inflammatory protein-2 and vascular endothelial growth factor protein production; neutrophil infiltration; oxidative loads; HIF-1α mRNA activation; HIF-1α expression; bronchial epithelial apoptosis; and decreased respiratory function in mice (p < 0.05). Endotoxin-induced augmentation of VILI and epithelial apoptosis were reduced in the HIF-1α-deficient mice and in the wild-type mice following enoxaparin administration (p < 0.05). Our data suggest that enoxaparin reduces endotoxin-augmented MV-induced ALI, partially by inhibiting the HIF-1α pathway.
Collapse
|
18
|
Yu Q, Wang D, Wen X, Tang X, Qi D, He J, Zhao Y, Deng W, Zhu T. Adipose-derived exosomes protect the pulmonary endothelial barrier in ventilator-induced lung injury by inhibiting the TRPV4/Ca 2+ signaling pathway. Am J Physiol Lung Cell Mol Physiol 2020; 318:L723-L741. [PMID: 32073873 PMCID: PMC7191475 DOI: 10.1152/ajplung.00255.2019] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Mechanical ventilation (MV) is the main supportive treatment of acute respiratory distress syndrome (ARDS), but it may lead to ventilator-induced lung injury (VILI). Large epidemiological studies have found that obesity was associated with lower mortality in mechanically ventilated patients with acute lung injury, which is known as “obesity paradox.” However, the effects of obesity on VILI are unknown. In the present study, wild-type mice were fed a high-fat diet (HFD) and ventilated with high tidal volume to investigate the effects of obesity on VILI in vivo, and pulmonary microvascular endothelial cells (PMVECs) were subjected to 18% cyclic stretching (CS) to further investigate its underlying mechanism in vitro. We found that HFD protects mice from VILI by alleviating the pulmonary endothelial barrier injury and inflammatory responses in mice. Adipose-derived exosomes can regulate distant tissues as novel adipokines, providing a new mechanism for cell-cell interactions. We extracted three adipose-derived exosomes, including HFD mouse serum exosome (S-Exo), adipose tissue exosome (AT-Exo), and adipose-derived stem cell exosome (ADSC-Exo), and further explored their effects on MV or 18% CS-induced VILI in vivo and in vitro. Administration of three exosomes protected against VILI by suppressing pulmonary endothelial barrier hyperpermeability, repairing the expression of adherens junctions, and alleviating inflammatory response in vivo and in vitro, accompanied by transient receptor potential vanilloid 4 (TRPV4)/Ca2+ pathway inhibition. Collectively, these data indicated that HFD-induced obesity plays a protective role in VILI by alleviating the pulmonary endothelial barrier injury and inflammatory response via adipose-derived exosomes, at least partially, through inhibiting the TRPV4/Ca2+ pathway.
Collapse
Affiliation(s)
- Qian Yu
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Daoxin Wang
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoting Wen
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xumao Tang
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Di Qi
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing He
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Zhao
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wang Deng
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Zhu
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
19
|
Szabari MV, Takahashi K, Feng Y, Locascio JJ, Chao W, Carter EA, Vidal Melo MF, Musch G. Relation between Respiratory Mechanics, Inflammation, and Survival in Experimental Mechanical Ventilation. Am J Respir Cell Mol Biol 2019; 60:179-188. [PMID: 30199644 DOI: 10.1165/rcmb.2018-0100oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Low-tidal volume (Vt) ventilation might protect healthy lungs from volutrauma but lead to inflammation resulting from other mechanisms, namely alveolar derecruitment and the ensuing alveolar collapse and tidal reexpansion. We hypothesized that the different mechanisms of low- and high-volume injury would be reflected in different mechanical properties being associated with development of pulmonary inflammation and mortality: an increase of hysteresis, reflecting progressive alveolar derecruitment, at low Vt; an increase of elastance, as a result of overdistension, at higher Vt. Mice were allocated to "protective" (6 ml/kg) or "injurious" (15-20 ml/kg) Vt groups and ventilated for 16 hours or until death. We measured elastance and hysteresis; pulmonary IL-6, IL-1β, and MIP-2 (macrophage inflammatory protein 2); wet-to-dry ratio; and blood gases. Survival was greater in the protective group (60%) than in the injurious group (25%). Nonsurvivors showed increased pulmonary cytokines, particularly in the injurious group, with the increase of elastance reflecting IL-6 concentration. Survivors instead showed only modest increases of cytokines, independent of Vt and unrelated to the increase of elastance. No single lung strain threshold could discriminate survivors from nonsurvivors. Hysteresis increased faster in the protective group, but, contrary to our hypothesis, its change was inversely related to the concentration of cytokines. In this model, significant mortality associated with pulmonary inflammation occurred even for strain values as low as about 0.8. Low Vt improved survival. The accompanying increase of hysteresis was not associated with greater inflammation.
Collapse
Affiliation(s)
- Margit V Szabari
- 1 Department of Anesthesia, Critical Care and Pain Medicine.,2 Department of Medicine
| | | | - Yan Feng
- 1 Department of Anesthesia, Critical Care and Pain Medicine.,4 Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland; and
| | | | - Wei Chao
- 1 Department of Anesthesia, Critical Care and Pain Medicine.,4 Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland; and
| | - Edward A Carter
- 6 Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Guido Musch
- 1 Department of Anesthesia, Critical Care and Pain Medicine.,7 Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri
| |
Collapse
|
20
|
Bain W, Matute-Bello G. Should we shift the paradigm of preclinical models for ARDS therapies? Thorax 2019; 74:1109-1110. [PMID: 31624219 DOI: 10.1136/thoraxjnl-2019-213729] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2019] [Indexed: 11/03/2022]
Affiliation(s)
- William Bain
- Department of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Gustavo Matute-Bello
- Center for Lung Biology, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington, USA
- Veterans Administration Puget Sound Healthcare System, Seattle, WA, United States
| |
Collapse
|
21
|
Zhang W, Dai H, Lin F, Zhao C, Wang X, Zhang S, Ge W, Pei S, Pan L. Ly-6C high inflammatory-monocyte recruitment is regulated by p38 MAPK/MCP-1 activation and promotes ventilator-induced lung injury. Int Immunopharmacol 2019; 78:106015. [PMID: 31780369 DOI: 10.1016/j.intimp.2019.106015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/23/2019] [Accepted: 10/28/2019] [Indexed: 12/14/2022]
Abstract
Lymphocyte antigen 6Chigh (Ly-6Chigh) inflammatory monocytes, as novel mononuclear cells in the innate immune system, participate in infectious diseases. In this study, we investigated the potential role of these monocytes in ventilator-induced lung injury (VILI) and the possible mechanism involved in their migration to lung tissue. Our results showed that mechanical ventilation with high tidal volume (HTV) increased the accumulation of Ly-6Chigh inflammatory monocytes in lung tissues and that blocking C‑C chemokine receptor 2 (CCR2) could significantly reduce Ly-6Chigh inflammatory-monocyte migration and attenuate the degree of inflammation of lung tissues. In addition, inhibition of p38 mitogen-activated protein kinase (p38 MAPK) activity could decrease the secretion of monocyte chemoattractant protein 1 (MCP-1), which in turn decreased the migration of Ly-6Chigh inflammatory monocytes into lung tissue. We also demonstrated that high ventilation caused Ly-6Chigh inflammatory monocytes in the bone marrow to migrate into and aggregate in the lungs, creating inflammation, and that the mechanism was quite different from that of infectious diseases. Ly-6Chigh inflammatory monocytes might play a pro-inflammatory role in VILI, and blocking their infiltration into lung tissue might become a new target for the treatment of this injury.
Collapse
Affiliation(s)
- Weikang Zhang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China; Perioperative Medical Research Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
| | - Huijun Dai
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China; Perioperative Medical Research Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
| | - Fei Lin
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China; Perioperative Medical Research Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
| | - Chen Zhao
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China; Perioperative Medical Research Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
| | - Xiaoxia Wang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China; Perioperative Medical Research Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
| | - SuiSui Zhang
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China; Perioperative Medical Research Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
| | - Wanyun Ge
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China; Perioperative Medical Research Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
| | - Shenglin Pei
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China; Perioperative Medical Research Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
| | - Linghui Pan
- Department of Anesthesiology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China; Perioperative Medical Research Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China.
| |
Collapse
|
22
|
Boehm O, Rohner M, Ehrentraut H, Guenther U, Meyer R, Knuefermann P, Baumgarten G, Duerr GD, Velten M. Low-tidal-volume prevent ventilation induced inflammation in a mouse model of sepsis. Life Sci 2019; 240:117081. [PMID: 31756342 DOI: 10.1016/j.lfs.2019.117081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND GOAL OF THE STUDY Pulmonary inflammation, increased vascular permeability, and pulmonary edema, occur in response to primary pulmonary infections like pneumonia but are also evident in endotoxemia or sepsis. Mechanical ventilation augments pre-existing lung injury and inflammation resulting from exposure to microbial products. The objective of this study was to test the hypothesis that low-tidal-volume prevent ventilation induced lung injury in sepsis. MATERIALS AND METHODS 10-12-week-old male C57BL/6N-mice received an intraperitoneal (i.p.) injection with equipotent dosages of LPS, 1668-thioate, 1612-thioate, or PBS. 120 min after injection, mice were randomized to low- (LV, 7 ± 1 ml/kg) or high-tidal-volume (HV, 25 ± 1 ml/kg) ventilation. Hemodynamic and ventilatory parameters were recorded and inflammatory markers were analyzed form BAL that was generated after 90 minute ventilation. RESULTS AND DISCUSSION Arterial blood pressures declined during mechanical ventilation in all groups. pO2 decreased in LPS injected and CO2 increased in sham, LPS, and 1612-thioate administered mice at 45 min and in 1668-thioate injected mice after 90 minute LV ventilation compared to respective HV groups. BAL protein concentrations increased in HV ventilated and 1668- or 1612-thioat pre-treated mice. BAL TNF-α protein concentrations increased in both LPS- and 1668-thioate-injected and IL-1β protein concentrations only in LPS-injected and HV ventilated mice. Most notably, no increased protein concentrations were observed in any of the LV ventilated groups. CONCLUSION We conclude that low-tidal-volume ventilation may be a potential strategy for the prevention of ventilator induced lung injury in a murine model of systemic TLR agonist induced lung injury.
Collapse
Affiliation(s)
- Olaf Boehm
- Department of Anesthesiology and Intensive Care Medicine, Rheinische Friedrich-Wilhelms-University Medical Center, Bonn, Germany
| | - Marc Rohner
- Department of Anesthesiology and Intensive Care Medicine, Rheinische Friedrich-Wilhelms-University Medical Center, Bonn, Germany
| | - Heidi Ehrentraut
- Department of Anesthesiology and Intensive Care Medicine, Rheinische Friedrich-Wilhelms-University Medical Center, Bonn, Germany
| | - Ulf Guenther
- Department of Anesthesiology, Critical Care, Emergency Medicine and Pain Therapy, Klinikum Oldenburg, Medical Campus University of Oldenburg, Germany
| | - Rainer Meyer
- Institute of Physiology, Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | | | | | - Georg Daniel Duerr
- Department of Cardiac Surgery, Rheinische Friedrich-Wilhelms-University Medical Center, Bonn, Germany
| | - Markus Velten
- Department of Anesthesiology and Intensive Care Medicine, Rheinische Friedrich-Wilhelms-University Medical Center, Bonn, Germany.
| |
Collapse
|
23
|
Yehya N. Lessons learned in acute respiratory distress syndrome from the animal laboratory. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:503. [PMID: 31728356 DOI: 10.21037/atm.2019.09.33] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Since the description of the acute respiratory distress syndrome (ARDS) in 1967, investigators have struggled to reproduce the syndrome in the animal laboratory. While several different models of experimental acute lung injury (ALI) have been developed, none completely capture the inciting etiologies, initial inflammation, heterogeneity, and resolution of human ARDS. This potentially has contributed to the poor translation of potential therapeutics between animal ALI models and human ARDS. It was only recently that standardized criteria were suggested for what makes an ALI model comparable to human ARDS. Nevertheless, despite model heterogeneity, these models have contributed substantially to our understanding of the syndrome. From the initial studies identifying the risks of mechanical ventilation to the identification of potentially targetable inflammatory mediators, to modern studies focusing on regional heterogeneity and novel molecular pathways, animal models continue to inform our understanding of ARDS. This review will cover several major lessons learned from animal models of ALI, and provide some direction for future studies in this field.
Collapse
Affiliation(s)
- Nadir Yehya
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
24
|
Wang L, Zhang Y, Zhang N, Xia J, Zhan Q, Wang C. Potential role of M2 macrophage polarization in ventilator-induced lung fibrosis. Int Immunopharmacol 2019; 75:105795. [PMID: 31421547 DOI: 10.1016/j.intimp.2019.105795] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 12/24/2022]
Abstract
Mechanical ventilation (MV) is an essential life-support technique, but it can induce ventilator-induced lung injury (VILI) and subsequent pulmonary fibrosis. The mechanisms underlying this fibrosis are largely unknown. Because excessive polarization of M2 macrophages has increasingly been cited as possible inciting factor for tissue remodeling and organ fibrosis, we here hypothesize it might be involved in the development of pulmonary fibrosis after high tidal volume (VT) MV. In our prospective, randomized, controlled animal study, C57BL/6 mice were randomly placed in either a VILI group or sham group. After ventilation, surviving mice were allowed to recover for 0, 1, 3, 5, 7, or 14 days. 200 mice were involved in our in vivo experiment, and the results calculated here refer only to the surviving mice. The results clearly showed that high-VT MV caused early inflammation and a subsequent fibroproliferative response in mice without pre-existing lung disease. High-VT MV was also found to lead to a dramatic increase in the number of M2 macrophages in mouse bronchoalveolar lavage fluid (BALF) cell and lung tissues. Consistent with the progression of fibrosis, there were far more M2 macrophages at the 5th day after ventilation and remained dominant for 2 weeks. High-VT MV induced epithelial-mesenchymal transition (EMT) on day 7, accompanied by the increased expression of TGF-β1 and p-Smad2/3. In vitro experiments, the co-culture of M2 macrophage and MLE-12 cells resulted in a significant EMT and upregulation of TGF-β1 and p-Smad2/3 in MLE-12 cells. To summarize, our findings suggested the persistent tilt polarization toward M2 macrophages was associated with EMT during the course of ventilator-induced pulmonary fibrosis, which may play its roles through activation of epithelial TGF-β1/Smad2/3 signaling.
Collapse
Affiliation(s)
- Lu Wang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, China; Center for Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, China
| | - Yi Zhang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, China; Center for Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, China
| | - Nannan Zhang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, China; Center for Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, China
| | - Jingen Xia
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, China; Center for Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, China
| | - Qingyuan Zhan
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, China; Center for Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, China.
| | - Chen Wang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, China; Center for Respiratory Diseases, National Clinical Research Center for Respiratory Diseases, China; Chinese Academy of Medical Science, Peking Union Medical College, China.
| |
Collapse
|
25
|
Oakley C, Koh M, Baldi R, Soni S, O'Dea K, Takata M, Wilson M. Ventilation following established ARDS: a preclinical model framework to improve predictive power. Thorax 2019; 74:1120-1129. [PMID: 31278170 DOI: 10.1136/thoraxjnl-2019-213460] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/09/2019] [Accepted: 06/07/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND Despite advances in understanding the pathophysiology of acute respiratory distress syndrome, effective pharmacological interventions have proven elusive. We believe this is a consequence of existing preclinical models being designed primarily to explore biological pathways, rather than predict treatment effects. Here, we describe a mouse model in which both therapeutic intervention and ventilation were superimposed onto existing injury and explored the impact of β-agonist treatment, which is effective in simple models but not clinically. METHODS Mice had lung injury induced by intranasal lipopolysaccharide (LPS), which peaked at 48 hours post-LPS based on clinically relevant parameters including hypoxaemia and impaired mechanics. At this peak of injury, mice were treated intratracheally with either terbutaline or tumour necrosis factor (TNF) receptor 1-targeting domain antibody, and ventilated with moderate tidal volume (20 mL/kg) to induce secondary ventilator-induced lung injury (VILI). RESULTS Ventilation of LPS-injured mice at 20 mL/kg exacerbated injury compared with low tidal volume (8 mL/kg). While terbutaline attenuated VILI within non-LPS-treated animals, it was ineffective to reduce VILI in pre-injured mice, mimicking its lack of clinical efficacy. In contrast, anti-TNF receptor 1 antibody attenuated secondary VILI within pre-injured lungs, indicating that the model was treatable. CONCLUSIONS We propose adoption of a practical framework like that described here to reduce the number of ultimately ineffective drugs reaching clinical trials. Novel targets should be evaluated alongside interventions which have been previously tested clinically, using models that recapitulate the (lack of) clinical efficacy. Within such a framework, outperforming a failed pharmacologic should be a prerequisite for drugs entering trials.
Collapse
Affiliation(s)
- Charlotte Oakley
- Department of Anaesthetics, Pain Medicine & Intensive Care, Imperial College London, London, UK
| | - Marissa Koh
- Department of Anaesthetics, Pain Medicine & Intensive Care, Imperial College London, London, UK
| | - Rhianna Baldi
- Department of Anaesthetics, Pain Medicine & Intensive Care, Imperial College London, London, UK
| | - Sanooj Soni
- Department of Anaesthetics, Pain Medicine & Intensive Care, Imperial College London, London, UK
| | - Kieran O'Dea
- Department of Anaesthetics, Pain Medicine & Intensive Care, Imperial College London, London, UK
| | - Masao Takata
- Department of Anaesthetics, Pain Medicine & Intensive Care, Imperial College London, London, UK
| | - Michael Wilson
- Department of Anaesthetics, Pain Medicine & Intensive Care, Imperial College London, London, UK
| |
Collapse
|
26
|
Zhang N, Zhang Y, Wang L, Xia J, Liang S, Wang Y, Wang Z, Huang X, Li M, Zeng H, Zhan Q. Expression profiling analysis of long noncoding RNAs in a mouse model of ventilator-induced lung injury indicating potential roles in inflammation. J Cell Biochem 2019; 120:11660-11679. [PMID: 30784114 PMCID: PMC7983175 DOI: 10.1002/jcb.28446] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/01/2018] [Accepted: 12/06/2018] [Indexed: 01/24/2023]
Abstract
The key regulators of inflammation underlying ventilator-induced lung injury (VILI) remain poorly defined. Long noncoding RNAs (lncRNAs) have been implicated in the inflammatory response of many diseases; however, their roles in VILI remain unclear. We, therefore, performed transcriptome profiling of lncRNA and messenger RNA (mRNA) using RNA sequencing in lungs collected from mice model of VILI and control groups. Gene expression was analyzed through RNA sequencing and quantitative reverse transctiption polymerase chain reaction. A comprehensive bioinformatics analysis was used to characterize the expression profiles and relevant biological functions and for multiple comparisons among the controls and the injury models at different time points. Finally, lncRNA-mRNA coexpression networks were constructed and dysregulated lncRNAs were analyzed functionally. The mRNA transcript profiling, coexpression network analysis, and functional analysis of altered lncRNAs indicated enrichment in the regulation of immune system/inflammation processes, response to stress, and inflammatory pathways. We identified the lncRNA Gm43181 might be related to lung damage and neutrophil activation via chemokine receptor chemokine (C-X-C) receptor 2. In summary, our study provides an identification of aberrant lncRNA alterations involved in inflammation upon VILI, and lncRNA-mediated regulatory patterns may contribute to VILI inflammation.
Collapse
Affiliation(s)
- Nan‐Nan Zhang
- Center for Respiratory Diseases, China‐Japan Friendship HospitalBeijingChina,Department of Pulmonary and Critical Care MedicineChina‐Japan Friendship HospitalBeijingChina,National Clinical Research Center for Respiratory DiseasesBeijingChina,Graduate School of Peking Union Medical College, Chinese Academy of Medical SciencesBeijingChina
| | - Yi Zhang
- Center for Respiratory Diseases, China‐Japan Friendship HospitalBeijingChina,Department of Pulmonary and Critical Care MedicineChina‐Japan Friendship HospitalBeijingChina,National Clinical Research Center for Respiratory DiseasesBeijingChina
| | - Lu Wang
- Center for Respiratory Diseases, China‐Japan Friendship HospitalBeijingChina,Department of Pulmonary and Critical Care MedicineChina‐Japan Friendship HospitalBeijingChina,National Clinical Research Center for Respiratory DiseasesBeijingChina
| | - Jin‐Gen Xia
- Center for Respiratory Diseases, China‐Japan Friendship HospitalBeijingChina,Department of Pulmonary and Critical Care MedicineChina‐Japan Friendship HospitalBeijingChina,National Clinical Research Center for Respiratory DiseasesBeijingChina
| | - Shun‐Tao Liang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical UniversityBeijingChina
| | - Yan Wang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical SciencesBeijingChina
| | - Zhi‐Zhi Wang
- Graduate School of Peking Union Medical College, Chinese Academy of Medical SciencesBeijingChina
| | - Xu Huang
- Center for Respiratory Diseases, China‐Japan Friendship HospitalBeijingChina,Department of Pulmonary and Critical Care MedicineChina‐Japan Friendship HospitalBeijingChina,National Clinical Research Center for Respiratory DiseasesBeijingChina
| | - Min Li
- Center for Respiratory Diseases, China‐Japan Friendship HospitalBeijingChina,Department of Pulmonary and Critical Care MedicineChina‐Japan Friendship HospitalBeijingChina,National Clinical Research Center for Respiratory DiseasesBeijingChina
| | - Hui Zeng
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical UniversityBeijingChina
| | - Qing‐Yuan Zhan
- Center for Respiratory Diseases, China‐Japan Friendship HospitalBeijingChina,Department of Pulmonary and Critical Care MedicineChina‐Japan Friendship HospitalBeijingChina,National Clinical Research Center for Respiratory DiseasesBeijingChina,Graduate School of Peking Union Medical College, Chinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
27
|
Mellenthin MM, Seong SA, Roy GS, Bartolák-Suki E, Hamlington KL, Bates JHT, Smith BJ. Using injury cost functions from a predictive single-compartment model to assess the severity of mechanical ventilator-induced lung injuries. J Appl Physiol (1985) 2019; 127:58-70. [PMID: 31046518 DOI: 10.1152/japplphysiol.00770.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Identifying safe ventilation patterns for patients with acute respiratory distress syndrome remains challenging because of the delicate balance between gas exchange and selection of ventilator settings to prevent further ventilator-induced lung injury (VILI). Accordingly, this work seeks to link ventilator settings to graded levels of VILI to identify injury cost functions that predict injury by using a computational model to process pressures and flows measured at the airway opening. Pressure-volume loops were acquired over the course of ~2 h of mechanical ventilation in four different groups of BALB/c mice. A cohort of these animals were subjected to an injurious bronchoalveolar lavage before ventilation. The data were analyzed with a single-compartment model that predicts recruitment/derecruitment and tissue distension at each time step in measured pressure-volume loops. We compared several injury cost functions to markers of VILI-induced blood-gas barrier disruption. Of the cost functions considered, we conclude that mechanical power dissipation and strain heterogeneity are the best at distinguishing between graded levels of injury and are good candidates for forecasting the development of VILI. NEW & NOTEWORTHY This work uses a predictive single-compartment model and injury cost functions to assess graded levels of mechanical ventilator-induced lung injury. The most promising measures include strain heterogeneity and mechanical power dissipation.
Collapse
Affiliation(s)
| | - Siyeon A Seong
- College of Medicine, University of Vermont , Burlington, Vermont
| | - Gregory S Roy
- College of Medicine, University of Vermont , Burlington, Vermont
| | | | - Katharine L Hamlington
- College of Medicine, University of Vermont , Burlington, Vermont.,University of Colorado at Children's Hospital Colorado , Aurora, Colorado
| | - Jason H T Bates
- College of Medicine, University of Vermont , Burlington, Vermont
| | - Bradford J Smith
- Department of Bioengineering, University of Colorado Denver , Aurora, Colorado.,College of Medicine, University of Vermont , Burlington, Vermont
| |
Collapse
|
28
|
Wilson M, Takata M. Mechanical Ventilation in Mice: Does Longer Equal Better? Am J Respir Cell Mol Biol 2019; 60:137-138. [DOI: 10.1165/rcmb.2018-0308ed] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Michael Wilson
- Anaesthetics, Pain Medicine and Intensive CareImperial College LondonLondon, United Kingdomand
- Chelsea and Westminster HospitalLondon, United Kingdom
| | - Masao Takata
- Anaesthetics, Pain Medicine and Intensive CareImperial College LondonLondon, United Kingdomand
- Chelsea and Westminster HospitalLondon, United Kingdom
| |
Collapse
|
29
|
Driving-pressure-independent protective effects of open lung approach against experimental acute respiratory distress syndrome. Crit Care 2018; 22:228. [PMID: 30243301 PMCID: PMC6151188 DOI: 10.1186/s13054-018-2154-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 08/08/2018] [Indexed: 12/16/2022] Open
Abstract
Background The open lung approach (OLA) reportedly has lung-protective effects against acute respiratory distress syndrome (ARDS). Recently, lowering of the driving pressure (ΔP), rather than improvement in lung aeration per se, has come to be considered as the primary lung-protective mechanism of OLA. However, the driving pressure-independent protective effects of OLA have never been evaluated in experimental studies. We here evaluated whether OLA shows protective effects against experimental ARDS even when the ΔP is not lowered. Methods Lipopolysaccharide was intratracheally administered to rats to establish experimental ARDS. After 24 h, rats were mechanically ventilated and randomly allocated to the OLA or control group. In the OLA group, 5 cmH2O positive end-expiratory pressure (PEEP) and recruitment maneuver (RM) were applied. Neither PEEP nor RM was applied to the rats in the control group. Dynamic ΔP was kept at 15 cmH2O in both groups. After 6 h of mechanical ventilation, rats in both groups received RM to inflate reversible atelectasis of the lungs. Arterial blood gas analysis, lung computed tomography, histological evaluation, and comprehensive biochemical analysis were performed. Results OLA significantly improved lung aeration, arterial oxygenation, and gas exchange. Even after RM in both groups, the differences in these parameters between the two groups persisted, indicating that the atelectasis-induced respiratory dysfunction observed in the control group is not an easily reversible functional problem. Lung histological damage was severe in the dorsal dependent area in both groups, but was attenuated by OLA. White blood cell counts, protein concentrations, and tissue injury markers in the broncho-alveolar lavage fluid (BALF) were higher in the control than in the OLA group. Furthermore, levels of CXCL-7, a platelet-derived chemokine, were higher in the BALF from the control group, indicating that OLA protects the lungs by suppressing platelet activation. Conclusions OLA shows protective effects against experimental ARDS, even when the ΔP is not decreased. In addition to reducing ΔP, maintaining lung aeration seems to be important for lung protection in ARDS. Electronic supplementary material The online version of this article (10.1186/s13054-018-2154-2) contains supplementary material, which is available to authorized users.
Collapse
|
30
|
Abstract
Both acute and chronic lung injury are associated with up-regulation of the pulmonary expression of the purinergic receptors P2XR4 and P2XR7. Genetic deletion or blockade of P2XR7 attenuated pulmonary hyperinflammation, but simultaneous P2XR4 up-regulation compensated for P2XR7 deletion. Therefore, we tested the hypothesis whether genetic P2XR4 deletion would attenuate the pulmonary inflammatory response and thereby improve organ function after blunt chest trauma in mice with and without pretraumatic cigarette smoke (CS) exposure.After 3 weeks to 4 weeks of exposure to CS, anesthetized wildtype or P2XR4 mice (n = 32) underwent a blast wave-induced blunt chest trauma followed by 4 h of lung-protective mechanical ventilation, fluid resuscitation, and noradrenaline support to maintain mean arterial pressure >55 mm Hg. Hemodynamics, lung mechanics, gas exchange, and acid-base status were measured together with blood and tissue cytokine and chemokine concentrations, heme oxygenase-1, B-cell lymphoma-extra large (Bcl-xL), endogenous nuclear factor-κB inhibitor (IκBα) expression, nitrotyrosine formation, purinergic receptor expression, and histological scoring.Despite a significant increase in the histopathology score in both CS-exposed groups, neither CS exposure nor P2XR4 deletion had any significant effect on post-traumatic pulmonary function and inflammatory response. However, P2XR4 deletion was associated with attenuated impairment of glucose homeostasis and acid-base-status after CS exposure and chest trauma.In conclusion, genetic P2XR4 deletion failed to attenuate the acute post-traumatic pulmonary inflammatory response. The improved glucose homeostasis and acid-base-status after CS exposure in the P2XR4 group was possibly due to less alveolar hypoxia-induced right ventricular remodeling resulting in preserved liver metabolic capacity.
Collapse
|
31
|
High-Fat Feeding Protects Mice From Ventilator-Induced Lung Injury, Via Neutrophil-Independent Mechanisms. Crit Care Med 2017; 45:e831-e839. [PMID: 28426531 DOI: 10.1097/ccm.0000000000002403] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Obesity has a complex impact on acute respiratory distress syndrome patients, being associated with increased likelihood of developing the syndrome but reduced likelihood of dying. We propose that such observations are potentially explained by a model in which obesity influences the iatrogenic injury that occurs subsequent to intensive care admission. This study therefore investigated whether fat feeding protected mice from ventilator-induced lung injury. DESIGN In vivo study. SETTING University research laboratory. SUBJECTS Wild-type C57Bl/6 mice or tumor necrosis factor receptor 2 knockout mice, either fed a high-fat diet for 12-14 weeks, or age-matched lean controls. INTERVENTIONS Anesthetized mice were ventilated with injurious high tidal volume ventilation for periods up to 180 minutes. MEASUREMENTS AND MAIN RESULTS Fat-fed mice showed clear attenuation of ventilator-induced lung injury in terms of respiratory mechanics, blood gases, and pulmonary edema. Leukocyte recruitment and activation within the lungs were not significantly attenuated nor were a host of circulating or intra-alveolar inflammatory cytokines. However, intra-alveolar matrix metalloproteinase activity and levels of the matrix metalloproteinase cleavage product soluble receptor for advanced glycation end products were significantly attenuated in fat-fed mice. This was associated with reduced stretch-induced CD147 expression on lung epithelial cells. CONCLUSIONS Consumption of a high-fat diet protects mice from ventilator-induced lung injury in a manner independent of neutrophil recruitment, which we postulate instead arises through blunted up-regulation of CD147 expression and subsequent activation of intra-alveolar matrix metalloproteinases. These findings may open avenues for therapeutic manipulation in acute respiratory distress syndrome and could have implications for understanding the pathogenesis of lung disease in obese patients.
Collapse
|
32
|
Smith BJ, Bartolak-Suki E, Suki B, Roy GS, Hamlington KL, Charlebois CM, Bates JHT. Linking Ventilator Injury-Induced Leak across the Blood-Gas Barrier to Derangements in Murine Lung Function. Front Physiol 2017; 8:466. [PMID: 28736528 PMCID: PMC5500660 DOI: 10.3389/fphys.2017.00466] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 06/19/2017] [Indexed: 01/10/2023] Open
Abstract
Mechanical ventilation is vital to the management of acute respiratory distress syndrome, but it frequently leads to ventilator-induced lung injury (VILI). Understanding the pathophysiological processes involved in the development of VILI is an essential prerequisite for improving lung-protective ventilation strategies. The goal of this study was to relate the amount and nature of material accumulated in the airspaces to biomarkers of injury and the derecruitment behavior of the lung in VILI. Forty-nine BALB/c mice were mechanically ventilated with combinations of tidal volume and end-expiratory pressures to produce varying degrees of overdistension and atelectasis while lung function was periodically assessed. Total protein, serum protein, and E-Cadherin levels were measured in bronchoalveolar lavage fluid (BALF). Tissue injury was assessed by histological scoring. We found that both high tidal volume and zero positive end-expiratory pressure were necessary to produce significant VILI. Increased BALF protein content was correlated with increased lung derecruitability, elevated peak pressures, and histological evidence of tissue injury. Blood derived molecules were present in the BALF in proportion to histological injury scores and epithelial injury, reflected by E-Cadherin levels in BALF. We conclude that repetitive recruitment is an important factor in the pathogenesis of VILI that exacerbates injury associated with tidal overdistension. Furthermore, the dynamic mechanical behavior of the injured lung provides a means to assess both the degree of tissue injury and the nature and amount of blood-derived fluid and proteins that accumulate in the airspaces.
Collapse
Affiliation(s)
- Bradford J Smith
- Department of Bioengineering, Anschutz Medical Campus, University of Colorado DenverAurora, CO, United States
| | | | - Bela Suki
- Department of Biomedical Engineering, Boston UniversityBoston, MA, United States
| | - Gregory S Roy
- Department of Medicine, Vermont Lung Center, Larner College of Medicine at The University of VermontBurlington, VT, United States
| | - Katharine L Hamlington
- Department of Medicine, Vermont Lung Center, Larner College of Medicine at The University of VermontBurlington, VT, United States
| | - Chantel M Charlebois
- Department of Medicine, Vermont Lung Center, Larner College of Medicine at The University of VermontBurlington, VT, United States
| | - Jason H T Bates
- Department of Medicine, Vermont Lung Center, Larner College of Medicine at The University of VermontBurlington, VT, United States
| |
Collapse
|
33
|
One-hit Models of Ventilator-induced Lung Injury: Benign Inflammation versus Inflammation as a By-product. Anesthesiology 2017; 126:909-922. [PMID: 28277372 DOI: 10.1097/aln.0000000000001605] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND One important explanation for the detrimental effects of conventional mechanical ventilation is the biotrauma hypothesis that ventilation may trigger proinflammatory responses that subsequently cause lung injury. This hypothesis has frequently been studied in so-called one-hit models (overventilation of healthy lungs) that so far have failed to establish an unequivocal link between inflammation and hypoxemic lung failure. This study was designed to develop a one-hit biotrauma model. METHODS Mice (six per group) were ventilated for up to 7 h (positive end-expiratory pressure 2 cm H2O) and received 300 μl/h fluid support. Series_1: initial plateau pressures of 10, 24, 27, or 30 cm H2O. Series_2: ventilation with pressure release at 34 cm H2O and initial plateau pressure of 10, 24, 27, or 30 cm H2O. To study the significance of inflammation, the latter groups were also pretreated with the steroid dexamethasone. RESULTS Within 7 h, 20 of 24 mice ventilated with plateau pressure of 27 cm H2O or more died of a catastrophic lung failure characterized by strongly increased proinflammatory markers and a precipitous decrease in pulmonary compliance, blood pressure, and oxygenation. Pretreatment with dexamethasone reduced inflammation, but prolonged median survival time by 30 min. CONCLUSIONS Our findings demonstrate a sharp distinction between ventilation with 24 cm H2O that was well tolerated and ventilation with 27 cm H2O that was lethal for most animals due to catastrophic lung failure. In the former case, inflammation was benign and in the latter, a by-product that only accelerated lung failure. The authors suggest that biotrauma-when defined as a ventilation-induced and inflammation-dependent hypoxemia-is difficult to study in murine one-hit models of ventilation, at least not within 7 h. (Anesthesiology 2017; 126:909-22).
Collapse
|
34
|
|
35
|
Wilson MR, Wakabayashi K, Bertok S, Oakley CM, Patel BV, O'Dea KP, Cordy JC, Morley PJ, Bayliffe AI, Takata M. Inhibition of TNF Receptor p55 By a Domain Antibody Attenuates the Initial Phase of Acid-Induced Lung Injury in Mice. Front Immunol 2017; 8:128. [PMID: 28243236 PMCID: PMC5304467 DOI: 10.3389/fimmu.2017.00128] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 01/25/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Tumor necrosis factor-α (TNF) is strongly implicated in the development of acute respiratory distress syndrome (ARDS), but its potential as a therapeutic target has been hampered by its complex biology. TNF signals through two receptors, p55 and p75, which play differential roles in pulmonary edema formation during ARDS. We have recently shown that inhibition of p55 by a novel domain antibody (dAb™) attenuated ventilator-induced lung injury. In the current study, we explored the efficacy of this antibody in mouse models of acid-induced lung injury to investigate the longer consequences of treatment. METHODS We employed two acid-induced injury models, an acute ventilated model and a resolving spontaneously breathing model. C57BL/6 mice were pretreated intratracheally or intranasally with p55-targeting dAb or non-targeting "dummy" dAb, 1 or 4 h before acid instillation. RESULTS Acid instillation in the dummy dAb group caused hypoxemia, increased respiratory system elastance, pulmonary inflammation, and edema in both the ventilated and resolving models. Pretreatment with p55-targeting dAb significantly attenuated physiological markers of ARDS in both models. p55-targeting dAb also attenuated pulmonary inflammation in the ventilated model, with signs that altered cytokine production and leukocyte recruitment persisted beyond the very acute phase. CONCLUSION These results demonstrate that the p55-targeting dAb attenuates lung injury and edema formation in models of ARDS induced by acid aspiration, with protection from a single dose lasting up to 24 h. Together with our previous data, the current study lends support toward the clinical targeting of p55 for patients with, or at risk of ARDS.
Collapse
Affiliation(s)
- Michael R Wilson
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital , London , UK
| | - Kenji Wakabayashi
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK; Department of Intensive Care Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Szabolcs Bertok
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital , London , UK
| | - Charlotte M Oakley
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital , London , UK
| | - Brijesh V Patel
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital , London , UK
| | - Kieran P O'Dea
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital , London , UK
| | - Joanna C Cordy
- Biopharm Molecular Discovery, GlaxoSmithKline R&D , Stevenage , UK
| | - Peter J Morley
- Biopharm Molecular Discovery, GlaxoSmithKline R&D , Stevenage , UK
| | | | - Masao Takata
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital , London , UK
| |
Collapse
|
36
|
Wang T, Gross C, Desai AA, Zemskov E, Wu X, Garcia AN, Jacobson JR, Yuan JXJ, Garcia JGN, Black SM. Endothelial cell signaling and ventilator-induced lung injury: molecular mechanisms, genomic analyses, and therapeutic targets. Am J Physiol Lung Cell Mol Physiol 2016; 312:L452-L476. [PMID: 27979857 DOI: 10.1152/ajplung.00231.2016] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 12/08/2016] [Accepted: 12/11/2016] [Indexed: 12/13/2022] Open
Abstract
Mechanical ventilation is a life-saving intervention in critically ill patients with respiratory failure due to acute respiratory distress syndrome (ARDS). Paradoxically, mechanical ventilation also creates excessive mechanical stress that directly augments lung injury, a syndrome known as ventilator-induced lung injury (VILI). The pathobiology of VILI and ARDS shares many inflammatory features including increases in lung vascular permeability due to loss of endothelial cell barrier integrity resulting in alveolar flooding. While there have been advances in the understanding of certain elements of VILI and ARDS pathobiology, such as defining the importance of lung inflammatory leukocyte infiltration and highly induced cytokine expression, a deep understanding of the initiating and regulatory pathways involved in these inflammatory responses remains poorly understood. Prevailing evidence indicates that loss of endothelial barrier function plays a primary role in the development of VILI and ARDS. Thus this review will focus on the latest knowledge related to 1) the key role of the endothelium in the pathogenesis of VILI; 2) the transcription factors that relay the effects of excessive mechanical stress in the endothelium; 3) the mechanical stress-induced posttranslational modifications that influence key signaling pathways involved in VILI responses in the endothelium; 4) the genetic and epigenetic regulation of key target genes in the endothelium that are involved in VILI responses; and 5) the need for novel therapeutic strategies for VILI that can preserve endothelial barrier function.
Collapse
Affiliation(s)
- Ting Wang
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Christine Gross
- Vascular Biology Center, Augusta University, Augusta, Georgia
| | - Ankit A Desai
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Evgeny Zemskov
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Xiaomin Wu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Alexander N Garcia
- Department of Pharmacology University of Illinois at Chicago, Chicago, Illinois; and
| | - Jeffrey R Jacobson
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Jason X-J Yuan
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Joe G N Garcia
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Stephen M Black
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona;
| |
Collapse
|
37
|
Silva PL, Pelosi P, Rocco PRM. Optimal mechanical ventilation strategies to minimize ventilator-induced lung injury in non-injured and injured lungs. Expert Rev Respir Med 2016; 10:1243-1245. [PMID: 27766893 DOI: 10.1080/17476348.2016.1251842] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Pedro L Silva
- a Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics , Federal University of Rio de Janeiro , Rio de Janeiro , Brazil
| | - Paolo Pelosi
- b Department of Surgical Sciences and Integrated Diagnostics, IRCCS AOU San Martino IST , University of Genoa , Genoa , Italy
| | - Patricia R M Rocco
- a Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics , Federal University of Rio de Janeiro , Rio de Janeiro , Brazil
| |
Collapse
|
38
|
John AE, Wilson MR, Habgood A, Porte J, Tatler AL, Stavrou A, Miele G, Jolly L, Knox AJ, Takata M, Offermanns S, Jenkins RG. Loss of epithelial Gq and G11 signaling inhibits TGFβ production but promotes IL-33-mediated macrophage polarization and emphysema. Sci Signal 2016; 9:ra104. [PMID: 27811142 DOI: 10.1126/scisignal.aad5568] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Heterotrimeric guanine nucleotide-binding protein (G protein) signaling links hundreds of G protein-coupled receptors with four G protein signaling pathways. Two of these, one mediated by Gq and G11 (Gq/11) and the other by G12 and G13 (G12/13), are implicated in the force-dependent activation of transforming growth factor-β (TGFβ) in lung epithelial cells. Reduced TGFβ activation in alveolar cells leads to emphysema, whereas enhanced TGFβ activation promotes acute lung injury and idiopathic pulmonary fibrosis. Therefore, precise control of alveolar TGFβ activation is essential for alveolar homeostasis. We investigated the involvement of the Gq/11 and G12/13 pathways in epithelial cells in generating active TGFβ and regulating alveolar inflammation. Mice deficient in both Gαq and Gα11 developed inflammation that was primarily caused by alternatively activated (M2-polarized) macrophages, enhanced matrix metalloproteinase 12 (MMP12) production, and age-related alveolar airspace enlargement consistent with emphysema. Mice with impaired Gq/11 signaling had reduced stretch-mediated generation of TGFβ by epithelial cells and enhanced macrophage MMP12 synthesis but were protected from the effects of ventilator-induced lung injury. Furthermore, synthesis of the cytokine interleukin-33 (IL-33) was increased in these alveolar epithelial cells, resulting in the M2-type polarization of alveolar macrophages independently of the effect on TGFβ. Our results suggest that alveolar Gq/11 signaling maintains alveolar homeostasis and likely independently increases TGFβ activation in response to the mechanical stress of the epithelium and decreases epithelial IL-33 synthesis. Together, these findings suggest that disruption of Gq/11 signaling promotes inflammatory emphysema but protects against mechanically induced lung injury.
Collapse
Affiliation(s)
- Alison E John
- Division of Respiratory Medicine, University of Nottingham, Nottingham, U.K.
| | - Michael R Wilson
- Department of Anaesthetics, Pain Medicine and Intensive Care, Imperial College, London, U.K
| | - Anthony Habgood
- Division of Respiratory Medicine, University of Nottingham, Nottingham, U.K
| | - Joanne Porte
- Division of Respiratory Medicine, University of Nottingham, Nottingham, U.K
| | - Amanda L Tatler
- Division of Respiratory Medicine, University of Nottingham, Nottingham, U.K
| | - Anastasios Stavrou
- Division of Respiratory Medicine, University of Nottingham, Nottingham, U.K
| | | | - Lisa Jolly
- Division of Respiratory Medicine, University of Nottingham, Nottingham, U.K
| | - Alan J Knox
- Division of Respiratory Medicine, University of Nottingham, Nottingham, U.K
| | - Masao Takata
- Department of Anaesthetics, Pain Medicine and Intensive Care, Imperial College, London, U.K
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - R Gisli Jenkins
- Division of Respiratory Medicine, University of Nottingham, Nottingham, U.K
| |
Collapse
|
39
|
Low tidal volume ventilation with low PEEP during surgery may induce lung inflammation. BMC Anesthesiol 2016; 16:47. [PMID: 27473050 PMCID: PMC4967315 DOI: 10.1186/s12871-016-0209-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 07/15/2016] [Indexed: 12/31/2022] Open
Abstract
Background Compared to conventional tidal volume ventilation, low tidal-volume ventilation reduces mortality in cased of acute respiratory distress syndrome. The aim of the present study is to determine whether low tidal-volume ventilation reduces the production of inflammatory mediators in the lungs and improves physiological status during hepatic surgery. Methods We randomly assigned patients undergoing hepatectomy into 2 groups: conventional tidal-volume vs. low tidal-volume (12 vs. 6 mL•kg−1 ideal body weight) ventilation with a positive end-expiratory pressure of 3 cm H2O. Arterial blood and airway epithelial lining fluid were sampled immediately after intubation and every 3 h thereafter. Results Twenty-five patients were analyzed. No significant changes were found in hemodynamics or acid–base status during the study. Interleukin-8 was significantly elevated in epithelial lining fluid from the low tidal-volume group. Oxygenation evaluated immediately after admission to the post-surgical care unit was significantly worse in the low tidal-volume group. Conclusions Low tidal-volume ventilation with low positive end-expiratory pressure may lead to pulmonary inflammation during major surgery such as hepatectomy. Trial registration The effect of ventilatory tidal volume on lung injury during hepatectomy that requires transient liver blood flow interruption. UMIN000021371 (03/07/2016); retrospectively registered
Collapse
|
40
|
|
41
|
Herbert JA, Valentine MS, Saravanan N, Schneck MB, Pidaparti R, Fowler AA, Reynolds AM, Heise RL. Conservative fluid management prevents age-associated ventilator induced mortality. Exp Gerontol 2016; 81:101-9. [PMID: 27188767 DOI: 10.1016/j.exger.2016.05.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 05/09/2016] [Accepted: 05/13/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND Approximately 800 thousand patients require mechanical ventilation in the United States annually with an in-hospital mortality rate of over 30%. The majority of patients requiring mechanical ventilation are over the age of 65 and advanced age is known to increase the severity of ventilator-induced lung injury (VILI) and in-hospital mortality rates. However, the mechanisms which predispose aging ventilator patients to increased mortality rates are not fully understood. Ventilation with conservative fluid management decreases mortality rates in acute respiratory distress patients, but to date there has been no investigation of the effect of conservative fluid management on VILI and ventilator associated mortality rates. We hypothesized that age-associated increases in susceptibility and incidence of pulmonary edema strongly promote age-related increases in ventilator associated mortality. METHODS 2month old and 20month old male C57BL6 mice were mechanically ventilated with either high tidal volume (HVT) or low tidal volume (LVT) for up to 4h with either liberal or conservative fluid support. During ventilation, lung compliance, total lung capacity, and hysteresis curves were quantified. Following ventilation, bronchoalveolar lavage fluid was analyzed for total protein content and inflammatory cell infiltration. Wet to dry ratios were used to directly measure edema in excised lungs. Lung histology was performed to quantify alveolar barrier damage/destruction. Age matched non-ventilated mice were used as controls. RESULTS At 4h, both advanced age and HVT ventilation significantly increased markers of inflammation and injury, degraded pulmonary mechanics, and decreased survival rates. Conservative fluid support significantly diminished pulmonary edema and improved pulmonary mechanics by 1h in advanced age HVT subjects. In 4h ventilations, conservative fluid support significantly diminished pulmonary edema, improved lung mechanics, and resulted in significantly lower mortality rates in older subjects. CONCLUSION Our study demonstrates that conservative fluid alone can attenuate the age associated increase in ventilator associated mortality.
Collapse
Affiliation(s)
- Joseph A Herbert
- Department of Biomedical Engineering, Virginia Commonwealth University, 401 W Main St, PO Box 843067, Richmond, VA 23284, United States
| | - Michael S Valentine
- Department of Biomedical Engineering, Virginia Commonwealth University, 401 W Main St, PO Box 843067, Richmond, VA 23284, United States
| | - Nivi Saravanan
- Department of Biomedical Engineering, Virginia Commonwealth University, 401 W Main St, PO Box 843067, Richmond, VA 23284, United States
| | - Matthew B Schneck
- Department of Biomedical Engineering, Virginia Commonwealth University, 401 W Main St, PO Box 843067, Richmond, VA 23284, United States
| | | | - Alpha A Fowler
- Division of Pulmonary Disease and Critical Care Medicine, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, United States
| | - Angela M Reynolds
- Department of Mathematics and Applies Mathematics 1015 Floyd Avenue P.O. Box 842014 Richmond, VA 23284-2014, United States
| | - Rebecca L Heise
- Department of Biomedical Engineering, Virginia Commonwealth University, 401 W Main St, PO Box 843067, Richmond, VA 23284, United States; Department of Physiology and Biophysics VCU School of Medicine 1101 East Marshall Street P.O. Box 980551 Richmond, VA 23298-0551, United States.
| |
Collapse
|
42
|
Wagner K, Gröger M, McCook O, Scheuerle A, Asfar P, Stahl B, Huber-Lang M, Ignatius A, Jung B, Duechs M, Möller P, Georgieff M, Calzia E, Radermacher P, Wagner F. Blunt Chest Trauma in Mice after Cigarette Smoke-Exposure: Effects of Mechanical Ventilation with 100% O2. PLoS One 2015. [PMID: 26225825 PMCID: PMC4520521 DOI: 10.1371/journal.pone.0132810] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cigarette smoking (CS) aggravates post-traumatic acute lung injury and increases ventilator-induced lung injury due to more severe tissue inflammation and apoptosis. Hyper-inflammation after chest trauma is due to the physical damage, the drop in alveolar PO2, and the consecutive hypoxemia and tissue hypoxia. Therefore, we tested the hypotheses that 1) CS exposure prior to blunt chest trauma causes more severe post-traumatic inflammation and thereby aggravates lung injury, and that 2) hyperoxia may attenuate this effect. Immediately after blast wave-induced blunt chest trauma, mice (n=32) with or without 3-4 weeks of CS exposure underwent 4 hours of pressure-controlled, thoraco-pulmonary compliance-titrated, lung-protective mechanical ventilation with air or 100 % O2. Hemodynamics, lung mechanics, gas exchange, and acid-base status were measured together with blood and tissue cytokine and chemokine concentrations, heme oxygenase-1 (HO-1), activated caspase-3, and hypoxia-inducible factor 1-α (HIF-1α) expression, nuclear factor-κB (NF-κB) activation, nitrotyrosine formation, purinergic receptor 2X4 (P2XR4) and 2X7 (P2XR7) expression, and histological scoring. CS exposure prior to chest trauma lead to higher pulmonary compliance and lower PaO2 and Horovitz-index, associated with increased tissue IL-18 and blood MCP-1 concentrations, a 2-4-fold higher inflammatory cell infiltration, and more pronounced alveolar membrane thickening. This effect coincided with increased activated caspase-3, nitrotyrosine, P2XR4, and P2XR7 expression, NF-κB activation, and reduced HIF-1α expression. Hyperoxia did not further affect lung mechanics, gas exchange, pulmonary and systemic cytokine and chemokine concentrations, or histological scoring, except for some patchy alveolar edema in CS exposed mice. However, hyperoxia attenuated tissue HIF-1α, nitrotyrosine, P2XR7, and P2XR4 expression, while it increased HO-1 formation in CS exposed mice. Overall, CS exposure aggravated post-traumatic inflammation, nitrosative stress and thereby organ dysfunction and injury; short-term, lung-protective, hyperoxic mechanical ventilation have no major beneficial effect despite attenuation of nitrosative stress, possibly due to compensation of by regional alveolar hypoxia and/or consecutive hypoxemia, resulting in down-regulation of HIF-1α expression.
Collapse
MESH Headings
- Acute Lung Injury/etiology
- Acute Lung Injury/physiopathology
- Acute Lung Injury/therapy
- Animals
- Disease Models, Animal
- Female
- Hyperoxia/complications
- Hyperoxia/pathology
- Hyperoxia/physiopathology
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Lung/pathology
- Lung/physiopathology
- Male
- Mice
- Mice, Inbred C57BL
- Oxidative Stress
- Pulmonary Disease, Chronic Obstructive/etiology
- Pulmonary Disease, Chronic Obstructive/physiopathology
- Pulmonary Disease, Chronic Obstructive/therapy
- Reactive Nitrogen Species/metabolism
- Receptors, Purinergic P2X/metabolism
- Respiration, Artificial/adverse effects
- Smoking/adverse effects
- Thoracic Injuries/complications
- Thoracic Injuries/physiopathology
- Thoracic Injuries/therapy
- Wounds, Nonpenetrating/complications
- Wounds, Nonpenetrating/physiopathology
- Wounds, Nonpenetrating/therapy
Collapse
Affiliation(s)
- Katja Wagner
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Ulm, Germany
- Klinik für Anästhesiologie, Universitätsklinikum, Ulm, Germany
| | - Michael Gröger
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Ulm, Germany
| | - Oscar McCook
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Ulm, Germany
| | | | - Pierre Asfar
- Laboratoire HIFIH, UPRES EA 3859, PRES l’UNAM, IFR 132, CNRS UMR 6214, INSERM U1083, Université Angers, Département de Réanimation Médicale et de Médecine Hyperbare, Centre Hospitalier Universitaire, Angers, France
| | - Bettina Stahl
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Ulm, Germany
| | - Markus Huber-Lang
- Klinik für Unfall-, Hand-, Plastische und Wiederherstellungschirurgie, Universitätsklinikum, Ulm, Germany
| | - Anita Ignatius
- Institut für Unfallchirurgische Forschung und Biomechanik, Universitätsklinikum, Ulm, Germany
| | - Birgit Jung
- Abteilung Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany
| | - Matthias Duechs
- Abteilung Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach/Riss, Germany
| | - Peter Möller
- Institut für Pathologie, Universitätsklinikum, Ulm, Germany
| | | | - Enrico Calzia
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Ulm, Germany
| | - Peter Radermacher
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Ulm, Germany
- * E-mail:
| | - Florian Wagner
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Ulm, Germany
- Klinik für Anästhesiologie, Universitätsklinikum, Ulm, Germany
| |
Collapse
|
43
|
Woods SJ, Waite AAC, O'Dea KP, Halford P, Takata M, Wilson MR. Kinetic profiling of in vivo lung cellular inflammatory responses to mechanical ventilation. Am J Physiol Lung Cell Mol Physiol 2015; 308:L912-21. [PMID: 25770178 PMCID: PMC4421782 DOI: 10.1152/ajplung.00048.2015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 03/07/2015] [Indexed: 02/07/2023] Open
Abstract
Mechanical ventilation, through overdistension of the lung, induces substantial inflammation that is thought to increase mortality among critically ill patients. The mechanotransduction processes involved in converting lung distension into inflammation during this ventilator-induced lung injury (VILI) remain unclear, although many cell types have been shown to be involved in its pathogenesis. This study aimed to identify the profile of in vivo lung cellular activation that occurs during the initiation of VILI. This was achieved using a flow cytometry-based method to quantify the phosphorylation of several markers (p38, ERK1/2, MAPK-activated protein kinase 2, and NF-κB) of inflammatory pathway activation within individual cell types. Anesthetized C57BL/6 mice were ventilated with low (7 ml/kg), intermediate (30 ml/kg), or high (40 ml/kg) tidal volumes for 1, 5, or 15 min followed by immediate fixing and processing of the lungs. Surprisingly, the pulmonary endothelium was the cell type most responsive to in vivo high-tidal-volume ventilation, demonstrating activation within just 1 min, followed by the alveolar epithelium. Alveolar macrophages were the slowest to respond, although they still demonstrated activation within 5 min. This order of activation was specific to VILI, since intratracheal lipopolysaccharide induced a very different pattern. These results suggest that alveolar macrophages may become activated via a secondary mechanism that occurs subsequent to activation of the parenchyma and that the lung cellular activation mechanism may be different between VILI and lipopolysaccharide. Our data also demonstrate that even very short periods of high stretch can promote inflammatory activation, and, importantly, this injury may be immediately manifested within the pulmonary vasculature.
Collapse
Affiliation(s)
- Samantha J. Woods
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Alicia A. C. Waite
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Kieran P. O'Dea
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Paul Halford
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Masao Takata
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Michael R. Wilson
- Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| |
Collapse
|
44
|
Yehya N, Xin Y, Oquendo Y, Cereda M, Rizi RR, Margulies SS. Cecal ligation and puncture accelerates development of ventilator-induced lung injury. Am J Physiol Lung Cell Mol Physiol 2014; 308:L443-51. [PMID: 25550313 DOI: 10.1152/ajplung.00312.2014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Sepsis is a leading cause of respiratory failure requiring mechanical ventilation, but the interaction between sepsis and ventilation is unclear. While prior studies demonstrated a priming role with endotoxin, actual septic animal models have yielded conflicting results regarding the role of preceding sepsis on development of subsequent ventilator-induced lung injury (VILI). Using a rat cecal ligation and puncture (CLP) model of sepsis and subsequent injurious ventilation, we sought to determine if sepsis affects development of VILI. Adult male Sprague-Dawley rats were subject to CLP or sham operation and, after 12 h, underwent injurious mechanical ventilation (tidal volume 30 ml/kg, positive end-expiratory pressure 0 cmH2O) for either 0, 60, or 120 min. Biochemical and physiological measurements, as well as computed tomography, were used to assess injury at 0, 60, and 120 min of ventilation. Before ventilation, CLP rats had higher levels of alveolar neutrophils and interleukin-1β. After 60 min of ventilation, CLP rats had worse injury as evidenced by increased alveolar inflammation, permeability, respiratory static compliance, edema, oxygenation, and computed tomography. By 120 min, CLP and sham rats had comparable levels of lung injury as assessed by many, but not all, of these metrics. CLP rats had an accelerated and worse loss of end-expiratory lung volume relative to sham, and consistently higher levels of alveolar interleukin-1β. Loss of aeration and progression of edema was more pronounced in dependent lung regions. We conclude that CLP initiated pulmonary inflammation in rats, and accelerated the development of subsequent VILI.
Collapse
Affiliation(s)
- Nadir Yehya
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania;
| | - Yi Xin
- Department of Radiology, Hospital of the University of Pennsylvania, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Yousi Oquendo
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Maurizio Cereda
- Department of Radiology, Hospital of the University of Pennsylvania, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and Department of Anesthesiology and Critical Care Medicine, Hospital of the University of Pennsylvania, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rahim R Rizi
- Department of Radiology, Hospital of the University of Pennsylvania, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Susan S Margulies
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
45
|
Cereda M, Xin Y, Kadlecek S, Hamedani H, Rajaei J, Clapp J, Rizi RR. Hyperpolarized gas diffusion MRI for the study of atelectasis and acute respiratory distress syndrome. NMR IN BIOMEDICINE 2014; 27:1468-78. [PMID: 24920074 PMCID: PMC4232982 DOI: 10.1002/nbm.3136] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 04/03/2014] [Accepted: 04/21/2014] [Indexed: 06/03/2023]
Abstract
Considerable uncertainty remains about the best ventilator strategies for the mitigation of atelectasis and associated airspace stretch in patients with acute respiratory distress syndrome (ARDS). In addition to several immediate physiological effects, atelectasis increases the risk of ventilator-associated lung injury, which has been shown to significantly worsen ARDS outcomes. A number of lung imaging techniques have made substantial headway in clarifying the mechanisms of atelectasis. This paper reviews the contributions of computed tomography, positron emission tomography, and conventional MRI to understanding this phenomenon. In doing so, it also reveals several important shortcomings inherent to each of these approaches. Once these shortcomings have been made apparent, we describe how hyperpolarized (HP) gas MRI--a technique that is uniquely able to assess responses to mechanical ventilation and lung injury in peripheral airspaces--is poised to fill several of these knowledge gaps. The HP-MRI-derived apparent diffusion coefficient (ADC) quantifies the restriction of (3) He diffusion by peripheral airspaces, thereby obtaining pulmonary structural information at an extremely small scale. Lastly, this paper reports the results of a series of experiments that measured ADC in mechanically ventilated rats in order to investigate (i) the effect of atelectasis on ventilated airspaces, (ii) the relationship between positive end-expiratory pressure (PEEP), hysteresis, and the dimensions of peripheral airspaces, and (iii) the ability of PEEP and surfactant to reduce airspace dimensions after lung injury. An increase in ADC was found to be a marker of atelectasis-induced overdistension. With recruitment, higher airway pressures were shown to reduce stretch rather than worsen it. Moving forward, HP MRI has significant potential to shed further light on the atelectatic processes that occur during mechanical ventilation.
Collapse
Affiliation(s)
- Maurizio Cereda
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, USA
| | - Yi Xin
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen Kadlecek
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Hooman Hamedani
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennia Rajaei
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Justin Clapp
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Rahim R. Rizi
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
46
|
Anderson SL, Duke-Novakovski T, Singh B. The immune response to anesthesia: part 1. Vet Anaesth Analg 2014; 41:113-26. [PMID: 24588928 DOI: 10.1111/vaa.12125] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 11/07/2013] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To review the immune response to anesthesia including mechanical ventilation, inhaled anesthetic gases, and injectable anesthetics and sedatives. STUDY DESIGN Review. METHODS AND DATABASES Multiple literature searches were performed using PubMed and Google Scholar from spring 2012 through fall 2013. Relevant anesthetic and immune terms were used to search databases without year published or species constraints. The online database for Veterinary Anaesthesia and Analgesia and the Journal of Veterinary Emergency and Critical Care were searched by issue starting in 2000 for relevant articles. CONCLUSION Recent research data indicate that commonly used volatile anesthetic agents, such as isoflurane and sevoflurane, may have a protective effect on vital organs. With the lung as the target organ, protection using an appropriate anesthetic protocol may be possible during direct pulmonary insults, including mechanical ventilation, and during systemic disease processes, such as endotoxemia, generalized sepsis, and ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Stacy L Anderson
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | | | | |
Collapse
|
47
|
Kaniaris E, Vaporidi K, Vergadi E, Theodorakis EE, Kondili E, Lagoudaki E, Tsatsanis C, Georgopoulos D. Genetic and pharmacologic inhibition of Tpl2 kinase is protective in a mouse model of ventilator-induced lung injury. Intensive Care Med Exp 2014; 2:15. [PMID: 26266915 PMCID: PMC4513004 DOI: 10.1186/2197-425x-2-15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 03/26/2014] [Indexed: 12/17/2022] Open
Abstract
Background Mechanical stress induced by injurious ventilation leads to pro-inflammatory cytokine production and lung injury. The extracellular-signal-regulated-kinase, ERK1/2, participates in the signaling pathways activated upon mechanical stress in the lungs to promote the inflammatory response. Tumor progression locus 2 (Tpl2) is a MAP3kinase that activates ERK1/2 upon cytokine or TLR signaling, to induce pro-inflammatory cytokine production. The role of Tpl2 in lung inflammation, and specifically in the one caused by mechanical stress has not been investigated. The aim of the study was to examine if genetic or pharmacologic inhibition of Tpl2 could ameliorate ventilator-induced lung injury. Methods Adult male wild-type and Tpl2-deficient mice were ventilated with normal or high tidal volume for 4 h. Additional wild-type mice were treated with a Tpl2 inhibitor either before or 30 min after initiation of high tidal ventilation. Non-ventilated mice of both genotypes served as controls. The development of lung injury was evaluated by measuring lung mechanics, arterial blood gases, concentrations of proteins, IL-6, and MIP-2 in bronchoalveolar lavage fluid (BALF) and by lung histology. Data were compared by Kruskal-Wallis non-parametric test and significance was defined as p < 0.05. Results Mechanical ventilation with normal tidal volume induced a mild increase of IL-6 in BALF in both strains. High tidal volume ventilation induced lung injury in wild-type mice, characterized by decreased lung compliance, increased concentrations of proteins, IL-6 and MIP-2 in BALF, and inflammatory cell infiltration on histology. All indices of lung injury were ameliorated in Tpl2-deficient mice. Wild-type mice treated with the Tpl2 inhibitor, either prior of after the initiation of high tidal volume ventilation were protected from the development of lung injury, as indicated by preserved lung compliance and lower BALF concentrations of proteins and IL-6, than similarly ventilated, untreated wild-type mice. Conclusions Genetic and pharmacologic inhibition of Tpl2 is protective in a mouse model of ventilator-induced lung injury, ameliorating both high-permeability pulmonary edema and lung inflammation.
Collapse
Affiliation(s)
- Evangelos Kaniaris
- Department of Intensive Care Medicine, Experimental Intensive Care Medicine Laboratory, University of Crete, School of Medicine, Heraklio, Crete, 71003, Greece,
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Wakabayashi K, Wilson MR, Tatham KC, O'Dea KP, Takata M. Volutrauma, but not atelectrauma, induces systemic cytokine production by lung-marginated monocytes. Crit Care Med 2014; 42:e49-57. [PMID: 23963135 DOI: 10.1097/ccm.0b013e31829a822a] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVES Ventilator-induced lung injury has substantive impact on mortality of patients with acute respiratory distress syndrome. Although low tidal volume ventilation has been shown to reduce mortality, clinical benefits of open-lung strategy are controversial. In this study, we investigated the impact of two distinct forms of ventilator-induced lung injury, i.e., volutrauma and atelectrauma, on the progression of lung injury and inflammation, in particular alveolar and systemic cytokine production. DESIGN Ex vivo study. SETTING University research laboratory. SUBJECTS C57BL/6 mice. INTERVENTIONS Isolated, buffer-perfused lungs were allocated to one of three ventilatory protocols for 3 hours: control group received low tidal volume (7 mL/kg) with positive end-expiratory pressure (5 cm H2O) and regular sustained inflation; high-stretch group received high tidal volume (30-32 mL/kg) with positive end-expiratory pressure (3 cm H2O) and sustained inflation; and atelectasis group received the same tidal volume as control but neither positive end-expiratory pressure nor sustained inflation. MEASUREMENTS AND MAIN RESULTS Both injurious ventilatory protocols developed comparable levels of physiological injury and pulmonary edema, measured by respiratory system mechanics and lavage fluid protein. High-stretch induced marked increases in proinflammatory cytokines in perfusate and lung lavage fluid, compared to control. In contrast, atelectasis had no effect on perfusate cytokines compared to control but did induce some up-regulation of lavage cytokines. Depletion of monocytes marginated within the lung microvasculature, achieved by pretreating mice with i.v. liposome-encapsulated clodronate, significantly attenuated perfusate cytokine levels, especially tumor necrosis factor, in the high-stretch, but not atelectasis group. CONCLUSIONS Volutrauma (high-stretch), but not atelectrauma (atelectasis), directly activates monocytes within the pulmonary vasculature, leading to cytokine release into systemic circulation. We postulate this as a potential explanation why open-lung strategy has limited mortality benefits in ventilated critically ill patients.
Collapse
Affiliation(s)
- Kenji Wakabayashi
- All authors: Section of Anaesthetics, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | | | | | | | | |
Collapse
|
49
|
Leong AFT, Fouras A, Islam MS, Wallace MJ, Hooper SB, Kitchen MJ. High spatiotemporal resolution measurement of regional lung air volumes from 2D phase contrast x-ray images. Med Phys 2013; 40:041909. [PMID: 23556903 DOI: 10.1118/1.4794926] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
PURPOSE Described herein is a new technique for measuring regional lung air volumes from two-dimensional propagation-based phase contrast x-ray (PBI) images at very high spatial and temporal resolution. Phase contrast dramatically increases lung visibility and the outlined volumetric reconstruction technique quantifies dynamic changes in respiratory function. These methods can be used for assessing pulmonary disease and injury and for optimizing mechanical ventilation techniques for preterm infants using animal models. METHODS The volumetric reconstruction combines the algorithms of temporal subtraction and single image phase retrieval (SIPR) to isolate the image of the lungs from the thoracic cage in order to measure regional lung air volumes. The SIPR algorithm was used to recover the change in projected thickness of the lungs on a pixel-by-pixel basis (pixel dimensions ≈ 16.2 μm). The technique has been validated using numerical simulation and compared results of measuring regional lung air volumes with and without the use of temporal subtraction for removing the thoracic cage. To test this approach, a series of PBI images of newborn rabbit pups mechanically ventilated at different frequencies was employed. RESULTS Regional lung air volumes measured from PBI images of newborn rabbit pups showed on average an improvement of at least 20% in 16% of pixels within the lungs in comparison to that measured without the use of temporal subtraction. The majority of pixels that showed an improvement was found to be in regions occupied by bone. Applying the volumetric technique to sequences of PBI images of newborn rabbit pups, it is shown that lung aeration at birth can be highly heterogeneous. CONCLUSIONS This paper presents an image segmentation technique based on temporal subtraction that has successfully been used to isolate the lungs from PBI chest images, allowing the change in lung air volume to be measured over regions as small as the pixel size. Using this technique, it is possible to measure changes in regional lung volume at high spatial and temporal resolution during breathing at much lower x-ray dose than would be required using computed tomography.
Collapse
|
50
|
Li LF, Chu PH, Hung CY, Kao WWY, Lin MC, Liu YY, Yang CT. Lumican regulates ventilation-induced epithelial-mesenchymal transition through extracelluar signal-regulated kinase pathway. Chest 2013; 143:1252-1260. [PMID: 23154825 DOI: 10.1378/chest.12-2058] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND Mechanical ventilation used in patients with acute lung injury can damage pulmonary epithelial cells through production of inflammatory cytokines and excess deposition of the extracellular matrix protein lumican. Lumican participates in macrophage inflammatory protein (MIP)-2 and transforming growth factor-β₁ (TGF-β₁) signaling during the fibroproliferative phase of acute lung injury, which involves a process of epithelial-mesenchymal transition (EMT). The mechanisms regulating interactions between mechanical ventilation and lung injury are unclear. We hypothesized that lung damage and EMT by high tidal volume (Vt) mechanical stretch causes upregulation of lumican that modulates MIP-2 and TGF-β₁ through the extracellular signal-regulated kinase (ERK) 1/2 pathway. METHODS Male C57BL/6 mice (either wild type or lumican null) aged 3 months and weighing between 25 and 30 g were exposed to low Vt (6 mL/kg) or high Vt (30 mL/kg) mechanical ventilation with room air for 2 to 8 h. Nonventilated mice were used as control subjects. RESULTS We found that high Vt mechanical ventilation increased microvascular permeability, neutrophil influx, production of free radicals, MIP-2 and TGF-β₁ proteins, positive staining of α-smooth muscle actin and S100A4/fibroblast-specific protein-1, Masson trichrome staining and extracellular collagen, and activation of lumican and ERK1/2 in wild-type mice. Decreased staining of the epithelial marker E-cadherin was also observed. Mechanical stretch-augmented EMT was attenuated with lumican-deficient mice and pharmacologic inhibition of ERK1/2 activity by PD98059. CONCLUSIONS The data suggest that lumican promotes high Vt mechanical ventilation-induced lung injury and EMT through the activation of the ERK1/2 pathway.
Collapse
Affiliation(s)
- Li-Fu Li
- Division of Pulmonary and Critical Care Medicine, Kweishan, Taoyuan, Taiwan; Department of Respiratory Therapy, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan; Department of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Pao-Hsien Chu
- Department of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan; First Cardiovascular Division, Department of Internal Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Cheng-Yiu Hung
- Division of Pulmonary and Critical Care Medicine, Kweishan, Taoyuan, Taiwan; Department of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Winston W-Y Kao
- Edith J. Crawley Vision Science Research Laboratory, Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, OH
| | - Meng-Chih Lin
- Department of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan; Division of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yung-Yang Liu
- Department of Respiratory Therapy, Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan; Department of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan; Chest Department, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Cheng-Ta Yang
- Division of Pulmonary and Critical Care Medicine, Kweishan, Taoyuan, Taiwan.
| |
Collapse
|