1
|
Endo Y, Aoki T, Jafari D, Rolston DM, Hagiwara J, Ito-Hagiwara K, Nakamura E, Kuschner CE, Becker LB, Hayashida K. Acute lung injury and post-cardiac arrest syndrome: a narrative review. J Intensive Care 2024; 12:32. [PMID: 39227997 PMCID: PMC11370287 DOI: 10.1186/s40560-024-00745-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/22/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Post-cardiac arrest syndrome (PCAS) presents a multifaceted challenge in clinical practice, characterized by severe neurological injury and high mortality rates despite advancements in management strategies. One of the important critical aspects of PCAS is post-arrest lung injury (PALI), which significantly contributes to poor outcomes. PALI arises from a complex interplay of pathophysiological mechanisms, including trauma from chest compressions, pulmonary ischemia-reperfusion (IR) injury, aspiration, and systemic inflammation. Despite its clinical significance, the pathophysiology of PALI remains incompletely understood, necessitating further investigation to optimize therapeutic approaches. METHODS This review comprehensively examines the existing literature to elucidate the epidemiology, pathophysiology, and therapeutic strategies for PALI. A comprehensive literature search was conducted to identify preclinical and clinical studies investigating PALI. Data from these studies were synthesized to provide a comprehensive overview of PALI and its management. RESULTS Epidemiological studies have highlighted the substantial prevalence of PALI in post-cardiac arrest patients, with up to 50% of survivors experiencing acute lung injury. Diagnostic imaging modalities, including chest X-rays, computed tomography, and lung ultrasound, play a crucial role in identifying PALI and assessing its severity. Pathophysiologically, PALI encompasses a spectrum of factors, including chest compression-related trauma, pulmonary IR injury, aspiration, and systemic inflammation, which collectively contribute to lung dysfunction and poor outcomes. Therapeutically, lung-protective ventilation strategies, such as low tidal volume ventilation and optimization of positive end-expiratory pressure, have emerged as cornerstone approaches in the management of PALI. Additionally, therapeutic hypothermia and emerging therapies targeting mitochondrial dysfunction hold promise in mitigating PALI-related morbidity and mortality. CONCLUSION PALI represents a significant clinical challenge in post-cardiac arrest care, necessitating prompt diagnosis and targeted interventions to improve outcomes. Mitochondrial-related therapies are among the novel therapeutic strategies for PALI. Further clinical research is warranted to optimize PALI management and enhance post-cardiac arrest care paradigms.
Collapse
Affiliation(s)
- Yusuke Endo
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Tomoaki Aoki
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Daniel Jafari
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Daniel M Rolston
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Jun Hagiwara
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Kanako Ito-Hagiwara
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Eriko Nakamura
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
| | - Cyrus E Kuschner
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Lance B Becker
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Kei Hayashida
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health System, Manhasset, NY, USA.
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
| |
Collapse
|
2
|
Shanholtz CB, Terrin ML, Harrington T, Chan C, Warren W, Walter R, Armstrong F, Marshall J, Scheraga R, Duggal A, Formanek P, Baram M, Afshar M, Marchetti N, Singla S, Reilly J, Knox D, Puri N, Chung K, Brown CH, Hasday JD. Design and rationale of the CHILL phase II trial of hypothermia and neuromuscular blockade for acute respiratory distress syndrome. Contemp Clin Trials Commun 2023; 33:101155. [PMID: 37228902 PMCID: PMC10191700 DOI: 10.1016/j.conctc.2023.101155] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 05/08/2023] [Accepted: 05/14/2023] [Indexed: 05/27/2023] Open
Abstract
The Cooling to Help Injured Lungs (CHILL) trial is an open label, two group, parallel design multicenter, randomized phase IIB clinical trial assessing the efficacy and safety of targeted temperature management with combined external cooling and neuromuscular blockade to block shivering in patients with early moderate-severe acute respiratory distress syndrome (ARDS). This report provides the background and rationale for the clinical trial and outlines the methods using the Consolidated Standards of Reporting Trials guidelines. Key design challenges include: [1] protocolizing important co-interventions; [2] incorporation of patients with COVID-19 as the cause of ARDS; [3] inability to blind the investigators; and [4] ability to obtain timely informed consent from patients or legally authorized representatives early in the disease process. Results of the Reevaluation of Systemic Early Neuromuscular Blockade (ROSE) trial informed the decision to mandate sedation and neuromuscular blockade only in the group assigned to therapeutic hypothermia and proceed without this mandate in the control group assigned to a usual temperature management protocol. Previous trials conducted in National Heart, Lung, and Blood Institute ARDS Clinical Trials (ARDSNet) and Prevention and Early Treatment of Acute Lung Injury (PETAL) Networks informed ventilator management, ventilation liberation and fluid management protocols. Since ARDS due to COVID-19 is a common cause of ARDS during pandemic surges and shares many features with ARDS from other causes, patients with ARDS due to COVID-19 are included. Finally, a stepwise approach to obtaining informed consent prior to documenting critical hypoxemia was adopted to facilitate enrollment and reduce the number of candidates excluded because eligibility time window expiration.
Collapse
Affiliation(s)
- Carl B. Shanholtz
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michael L. Terrin
- Department of Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Thelma Harrington
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Caleb Chan
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Whittney Warren
- Department of Pulmonary and Critical Care Medicine, Brooke Army Medical Center, San Antonio, TX, USA
| | - Robert Walter
- Department of Pulmonary and Critical Care Medicine, Brooke Army Medical Center, San Antonio, TX, USA
| | | | | | | | - Abjihit Duggal
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Perry Formanek
- Department of Medicine, Loyola University Medical Center, Maywood, IL, USA
| | - Michael Baram
- Department of Medicine, Sidney Kimmel College of Medicine USA, Philadelphia, PA, USA
| | - Majid Afshar
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Nathaniel Marchetti
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Sunit Singla
- Division of Pulmonary, Critical Care, Sleep, and Allergy Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - John Reilly
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Dan Knox
- Division of Pulmonary and Critical Care Medicine, Intermountain Medical Center, Murray, UT, USA
| | - Nitin Puri
- Division of Critical Care, Cooper University Health Care, USA
| | - Kevin Chung
- Department of Medicine, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Clayton H. Brown
- Department of Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jeffrey D. Hasday
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
3
|
Akyol O, Demirgan S, Şengelen A, Güneyli HC, Oran DS, Yıldırım F, Haktanır D, Sevdi MS, Erkalp K, Selcan A. Mild Hypothermia via External Cooling Improves Lung Function and Alleviates Pulmonary Inflammatory Response and Damage in Two-Hit Rabbit Model of Acute Lung Injury. J INVEST SURG 2022; 35:1472-1483. [PMID: 35435080 DOI: 10.1080/08941939.2022.2064010] [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: 01/11/2022] [Revised: 03/26/2022] [Accepted: 04/04/2022] [Indexed: 10/18/2022]
Abstract
OBJECTIVES Targeted temperature management (TTM) with therapeutic hypothermia (TH) has an organ-protective effect by mainly reducing inflammatory response. Here, our objective was to determine, for the first time, whether mild TH with external cooling, a simple and inexpensive method, could be safe or even beneficial in two-hit rabbit model of acute lung injury/acute respiratory distress syndrome (ALI/ARDS). METHODS Twenty-two New Zealand rabbits (6-month-old) were randomly divided into healthy control (HC) with conventional ventilation, but without injury, model group (ALI), and hypothermia group with external cooling (ALI-HT). After induction of ALI/ARDS through mild lung-lavages followed by non-protective ventilation, mild hypothermia was started in ALI-HT group (body temperature of 33-34 °C). All rabbits were conventionally ventilated for an additional 6-h by recording respiratory parameters. Finally, lung histopathology and inflammatory response were evaluated. RESULTS Hypothermia was associated with higher oxygen saturation, resulting in partial improvement in the P/F ratio (PaO2/FiO2), oxygenation index, mean airway pressure, and PaCO2, but did not affect lactate levels. The ALI-HT group had lower histopathological injury scores (hyperemia, edema, emphysema, atelectasis, and PMN infiltration). Further, tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6 and -8 levels in lung tissue and serum samples markedly reduced due to hypothermia. CONCLUSION Mild TH with external cooling reduced lung inflammation and damage, whereas it resulted in partial improvement in gas exchanges. Our findings highlight that body temperature control may be a potentially supportive therapeutic option for regulating cytokine production and respiratory parameters in ALI/ARDS.
Collapse
Affiliation(s)
- Onat Akyol
- T.C. Health Ministry, University of Health Sciences, Bağcılar Training and Research Hospital, Anesthesiology and Reanimation Clinic, Istanbul, Turkey
| | - Serdar Demirgan
- T.C. Health Ministry, University of Health Sciences, Bağcılar Training and Research Hospital, Anesthesiology and Reanimation Clinic, Istanbul, Turkey
- Department of Molecular Biology and Genetics, Institute of Graduate Studies in Sciences, Istanbul University, Istanbul, Turkey
| | - Aslıhan Şengelen
- Department of Molecular Biology and Genetics, Institute of Graduate Studies in Sciences, Istanbul University, Istanbul, Turkey
| | - Hasan Cem Güneyli
- T.C. Health Ministry, University of Health Sciences, Bağcılar Training and Research Hospital, Anesthesiology and Reanimation Clinic, Istanbul, Turkey
| | - Duygu Sultan Oran
- T.C. Health Ministry, University of Health Sciences, Bağcılar Training and Research Hospital, Experimental Research and Skills Development Center, Istanbul, Turkey
| | - Funda Yıldırım
- Department of Pathology, Faculty of Veterinary Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Damla Haktanır
- Department of Pathology, Faculty of Veterinary Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Mehmet Salih Sevdi
- T.C. Health Ministry, University of Health Sciences, Bağcılar Training and Research Hospital, Anesthesiology and Reanimation Clinic, Istanbul, Turkey
| | - Kerem Erkalp
- Department of Anesthesiology and Reanimation, Istanbul University-Cerrahpaşa, Institute of Cardiology, Istanbul, Turkey
| | - Ayşin Selcan
- T.C. Health Ministry, University of Health Sciences, Bağcılar Training and Research Hospital, Anesthesiology and Reanimation Clinic, Istanbul, Turkey
| |
Collapse
|
4
|
Angus SA, Henderson WR, Banoei MM, Molgat‐Seon Y, Peters CM, Parmar HR, Griesdale DEG, Sekhon M, Sheel AW, Winston BW, Dominelli PB. Therapeutic hypothermia attenuates physiologic, histologic, and metabolomic markers of injury in a porcine model of acute respiratory distress syndrome. Physiol Rep 2022; 10:e15286. [PMID: 35510328 PMCID: PMC9069168 DOI: 10.14814/phy2.15286] [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: 01/12/2022] [Revised: 03/29/2022] [Accepted: 04/02/2022] [Indexed: 06/14/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a lung injury characterized by noncardiogenic pulmonary edema and hypoxic respiratory failure. The purpose of this study was to investigate the effects of therapeutic hypothermia on short-term experimental ARDS. Twenty adult female Yorkshire pigs were divided into four groups (n = 5 each): normothermic control (C), normothermic injured (I), hypothermic control (HC), and hypothermic injured (HI). Acute respiratory distress syndrome was induced experimentally via intrapulmonary injection of oleic acid. Target core temperature was achieved in the HI group within 1 h of injury induction. Cardiorespiratory, histologic, cytokine, and metabolomic data were collected on all animals prior to and following injury/sham. All data were collected for approximately 12 h from the beginning of the study until euthanasia. Therapeutic hypothermia reduced injury in the HI compared to the I group (histological injury score = 0.51 ± 0.18 vs. 0.76 ± 0.06; p = 0.02) with no change in gas exchange. All groups expressed distinct phenotypes, with a reduction in pro-inflammatory metabolites, an increase in anti-inflammatory metabolites, and a reduction in inflammatory cytokines observed in the HI group compared to the I group. Changes to respiratory system mechanics in the injured groups were due to increases in lung elastance (E) and resistance (R) (ΔE from pre-injury = 46 ± 14 cmH2 O L-1 , p < 0.0001; ΔR from pre-injury: 3 ± 2 cmH2 O L-1 s- , p = 0.30) rather than changes to the chest wall (ΔE from pre-injury: 0.7 ± 1.6 cmH2 O L-1 , p = 0.99; ΔR from pre-injury: 0.6 ± 0.1 cmH2 O L-1 s- , p = 0.01). Both control groups had no change in respiratory mechanics. In conclusion, therapeutic hypothermia can reduce markers of injury and inflammation associated with experimentally induced short-term ARDS.
Collapse
Affiliation(s)
- Sarah A. Angus
- Department of KinesiologyUniversity of WaterlooWaterlooOntarioCanada
| | - William R. Henderson
- Division of Critical Care MedicineDepartment of MedicineFaculty of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Mohammad M. Banoei
- Department of Critical Care MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Yannick Molgat‐Seon
- Department Kinesiology and Applied HealthUniversity of WinnipegWinnipegManitobaCanada
| | - Carli M. Peters
- School of KinesiologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Hanna R. Parmar
- School of KinesiologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Donald E. G. Griesdale
- Division of Critical Care MedicineDepartment of MedicineFaculty of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of AnesthesiologyPharmacology & TherapeuticsUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Mypinder Sekhon
- Division of Critical Care MedicineDepartment of MedicineFaculty of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Andrew William Sheel
- School of KinesiologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Brent W. Winston
- Department of Critical Care MedicineUniversity of CalgaryCalgaryAlbertaCanada
- Departments of Medicine and Biochemistry & Molecular BiologyUniversity of CalgaryCalgaryAlbertaCanada
| | | |
Collapse
|
5
|
Cruces P, Cores C, Casanova D, Pizarro F, Díaz F. Successful use of mild therapeutic hypothermia as compassionate treatment for severe refractory hypoxemia in COVID-19. J Crit Care 2021; 63:260-263. [PMID: 33583631 PMCID: PMC7825805 DOI: 10.1016/j.jcrc.2021.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 01/06/2021] [Accepted: 01/18/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND COVID-19 is a disease associated with an intense systemic inflammation that could induce severe acute respiratory distress syndrome (ARDS), with life-threatening hypoxia and hypercapnia. We present a case where mild therapeutic hypothermia was associated with improved gas exchange, facing other therapies' unavailability due to the pandemic. CASE REPORT A healthy 38-year-old male admitted for COVID-19 pneumonia developed extreme hypoxia (PaO2/FiO2 ratio 42 mmHg), respiratory acidosis, and hyperthermia, refractory to usual treatment (mechanical ventilation, neuromuscular blockade, and prone position), and advanced therapies were not available. Mild therapeutic hypothermia management (target 33-34 °C) was maintained for five days, with progressive gas exchange improvement, which allowed his recovery over the following weeks. He was discharged home after 68 days without significant ICU associated morbidity. CONCLUSIONS Mild hypothermia is a widely available therapy, that given some specific characteristics of COVID-19, may be explored as adjunctive therapy for life-threatening ARDS, especially during a shortage of other rescue therapies.
Collapse
Affiliation(s)
- Pablo Cruces
- Unidad de Paciente Crítico Pediátrico, Hospital El Carmen de Maipú, Chile; Centro de Investigación de Medicina Veterinaria, Escuela de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andres Bello, Chile; Red Colaborativa Pediátrica de Latinoamérica (LARed Network), Chile..
| | - Camila Cores
- Unidad de Paciente Crítico Pediátrico, Hospital El Carmen de Maipú, Chile.
| | - Daniel Casanova
- Departamento de Post-Grado Pediatría, Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Chile.
| | - Federico Pizarro
- Departamento de Post-Grado Pediatría, Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Chile.
| | - Franco Díaz
- Unidad de Paciente Crítico Pediátrico, Hospital El Carmen de Maipú, Chile; Red Colaborativa Pediátrica de Latinoamérica (LARed Network), Chile.; Instituto de Ciencias e Innovación en Medicina (ICIM), Universidad del Desarrollo, Santiago, Chile.
| |
Collapse
|
6
|
Partial liquid ventilation–induced mild hypothermia improves the lung function and alleviates the inflammatory response during acute respiratory distress syndrome in canines. Biomed Pharmacother 2019; 118:109344. [PMID: 31545246 PMCID: PMC9386951 DOI: 10.1016/j.biopha.2019.109344] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/02/2019] [Accepted: 08/07/2019] [Indexed: 12/29/2022] Open
|
7
|
Pilot Feasibility Study of Therapeutic Hypothermia for Moderate to Severe Acute Respiratory Distress Syndrome. Crit Care Med 2017; 45:1152-1159. [PMID: 28406814 DOI: 10.1097/ccm.0000000000002338] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVES Prior studies suggest hypothermia may be beneficial in acute respiratory distress syndrome, but cooling causes shivering and increases metabolism. The objective of this study was to assess the feasibility of performing a randomized clinical trial of hypothermia in patients with acute respiratory distress syndrome receiving treatment with neuromuscular blockade because they cannot shiver. DESIGN Retrospective study and pilot, prospective, open-label, feasibility study. SETTING Medical ICU. PATIENTS Retrospective review of 58 patients with acute respiratory distress syndrome based on Berlin criteria and PaO2/FIO2 less than 150 who received neuromuscular blockade. Prospective hypothermia treatment in eight acute respiratory distress syndrome patients with PaO2/FIO2 less than 150 receiving neuromuscular blockade. INTERVENTION Cooling to 34-36°C for 48 hours. MEASUREMENTS AND MAIN RESULTS Core temperature, hemodynamics, serum glucose and electrolytes, and P/F were sequentially measured, and medians (interquartile ranges) presented, 28-day ventilator-free days, and hospital mortality were calculated in historical controls and eight cooled patients. Average patient core temperature was 36.7°C (36-37.3°C), and fever occurred during neuromuscular blockade in 30 of 58 retrospective patients. In the prospectively cooled patients, core temperature reached target range less than or equal to 4 hours of initiating cooling, remained less than 36°C for 92% of the 48 hours cooling period without adverse events, and was lower than the controls (34.35°C [34-34.8°C]; p < 0.0001). Compared with historical controls, the cooled patients tended to have lower hospital mortality (75% vs 53.4%; p = 0.26), more ventilator-free days (9 [0-21.5] vs 0 [0-12]; p = 0.16), and higher day 3 P/F (255 [160-270] vs 171 [120-214]; p = 0.024). CONCLUSIONS Neuromuscular blockade alone does not cause hypothermia but allowed acute respiratory distress syndrome patients to be effectively cooled. Results support conducting a randomized clinical trial of hypothermia in acute respiratory distress syndrome and the feasibility of studying acute respiratory distress syndrome patients receiving neuromuscular blockade.
Collapse
|
8
|
Xia J, Li R, Yang R, Zhang L, Sun B, Feng Y, Jin J, Huang L, Zhan Q. Mild hypothermia attenuate kidney injury in canines with oleic acid-induced acute respiratory distress syndrome. Injury 2016; 47:1445-51. [PMID: 27180146 DOI: 10.1016/j.injury.2016.04.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/18/2016] [Indexed: 02/02/2023]
Abstract
BACKGROUND Hypothermia may attenuate ventilator induced-lung injury in acute respiratory distress syndrome (ARDS). However, the impact of hypothermia on extra-pulmonary organ injury in ARDS remains unclear. The purpose of this study was to investigate whether hypothermia affects extra-pulmonary organ injury in a canine ARDS model induced by oleic acid. OBJECTIVES Twelve anesthetized canines with oleic acid-induced ARDS were randomly divided (n=6 per group) into a hypothermia group (core temperature of 33±1°C, HT group) and a normothermia group (core temperature of 38±1°C, NT group) and treated for four hours. The liver, small intestine and kidney were assessed by evaluating biochemical parameters, plasma and tissue cytokine levels, and tissue histopathological injury scores. RESULTS The HT group showed a lower plateau pressure, lung elastance and pulmonary vascular resistance. Hypothermia was associated with lower oxygen consumption (138.4±55.0mlmin(-1)vs. 72.0±11.2mlmin(-1), P<0.05) and higher oxygen saturation of mixed venous blood (62.8%±8.0% vs. 77.5%±10.1%, P<0.05). Both groups had similar levels of tumour necrosis factor-α in the plasma and extra-pulmonary organ, however, plasma interleukin-10 (97.1±25.0pgml(-1)vs. 131.4±27.0pgml(-1), P<0.05) was higher in the HT group. Further, the animals in the HT group had a lower levels of plasma creatinine (54.6±19.1UL(-1)vs. 29.1±8.0UL(-1), P<0.05), and lower renal histopathological injury scores [4.0(3.5;7.0) vs. 1.5(0.8;3.0), P<0.05]. Hypothermia did not affect the histopathological injury of the liver and small intestine. CONCLUSIONS Short-term mild hypothermia can reduce lung elastance and pulmonary vascular resistance, increase the systemic anti-inflammatory response and attenuate kidney histopathological injury in a canine ARDS model induced by oleic acid.
Collapse
Affiliation(s)
- Jingen Xia
- Department of Respiratory and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, PR China.
| | - Ran Li
- Beijing Institute of Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, PR China.
| | - Rui Yang
- Beijing Institute of Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, PR China.
| | - Li Zhang
- Beijing Institute of Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, PR China.
| | - Bing Sun
- Beijing Institute of Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, PR China.
| | - Yingying Feng
- Department of Respiratory and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, PR China.
| | - Jingjing Jin
- Beijing Institute of Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, PR China.
| | - Linna Huang
- Beijing Institute of Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, PR China.
| | - Qingyuan Zhan
- Department of Respiratory and Critical Care Medicine, China-Japan Friendship Hospital, Beijing, PR China.
| |
Collapse
|
9
|
Hartmann EK, Thomas R, Liu T, Stefaniak J, Ziebart A, Duenges B, Eckle D, Markstaller K, David M. TIP peptide inhalation in experimental acute lung injury: effect of repetitive dosage and different synthetic variants. BMC Anesthesiol 2014; 14:42. [PMID: 24904234 PMCID: PMC4046002 DOI: 10.1186/1471-2253-14-42] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 05/21/2014] [Indexed: 12/17/2022] Open
Abstract
Background Inhalation of TIP peptides that mimic the lectin-like domain of TNF-α is a novel approach to attenuate pulmonary oedema on the threshold to clinical application. A placebo-controlled porcine model of acute respiratory distress syndrome (ARDS) demonstrated a reduced thermodilution-derived extravascular lung water index (EVLWI) and improved gas exchange through TIP peptide inhalation within three hours. Based on these findings, the present study compares a single versus a repetitive inhalation of a TIP peptide (TIP-A) and two alternate peptide versions (TIP-A, TIP-B). Methods Following animal care committee approval ARDS was induced by bronchoalveolar lavage followed by injurious ventilation in 21 anaesthetized pigs. A randomised-blinded three-group setting compared the single-dosed peptide variants TIP-A and TIP-B as well as single versus repetitive inhalation of TIP-A (n = 7 per group). Over two three-hour intervals parameters of gas exchange, transpulmonary thermodilution, calculated alveolar fluid clearance, and ventilation/perfusion-distribution were assessed. Post-mortem measurements included pulmonary wet/dry ratio and haemorrhage/congestion scoring. Results The repetitive TIP-A inhalation led to a significantly lower wet/dry ratio than a single dose and a small but significantly lower EVLWI. However, EVLWI changes over time and the derived alveolar fluid clearance did not differ significantly. The comparison of TIP-A and B showed no relevant differences. Gas exchange and ventilation/perfusion-distribution significantly improved in all groups without intergroup differences. No differences were found in haemorrhage/congestion scoring. Conclusions In comparison to a single application the repetitive inhalation of a TIP peptide in three-hour intervals may lead to a small additional reduction the lung water content. Two alternate TIP peptide versions showed interchangeable characteristics.
Collapse
Affiliation(s)
- Erik K Hartmann
- Department of Anaesthesiology, Medical Centre of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Rainer Thomas
- Department of Anaesthesiology, Medical Centre of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Tanghua Liu
- Department of Anaesthesiology, Medical Centre of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Joanna Stefaniak
- Department of Anaesthesiology, General Critical Care Medicine and Pain Therapy, Medical University Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Alexander Ziebart
- Department of Anaesthesiology, Medical Centre of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Bastian Duenges
- Department of Anaesthesiology, Medical Centre of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Daniel Eckle
- Department of Anaesthesiology, Medical Centre of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Klaus Markstaller
- Department of Anaesthesiology, Medical Centre of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany ; Department of Anaesthesiology, General Critical Care Medicine and Pain Therapy, Medical University Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Matthias David
- Department of Anaesthesiology, Medical Centre of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| |
Collapse
|
10
|
Karnatovskaia LV, Festic E, Freeman WD, Lee AS. Effect of therapeutic hypothermia on gas exchange and respiratory mechanics: a retrospective cohort study. Ther Hypothermia Temp Manag 2014; 4:88-95. [PMID: 24840620 DOI: 10.1089/ther.2014.0004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Targeted temperature management (TTM) may improve respiratory mechanics and lung inflammation in acute respiratory distress syndrome (ARDS) based on animal and limited human studies. We aimed to assess the pulmonary effects of TTM in patients with respiratory failure following cardiac arrest. Retrospective review of consecutive cardiac arrest cases occurring out of hospital or within 24 hours of hospital admission (2002-2012). Those receiving TTM (n=44) were compared with those who did not (n=42), but required mechanical ventilation (MV) for at least 4 days following the arrest. There were no between-group differences in age, gender, body mass index, APACHE II, or fluid balance during the study period. The TTM group had lower ejection fraction, Glasgow Coma Score, and more frequent use of paralytics. Matched data analyses (change at day 4 compared with baseline of the individual subject) showed favorable, but not statistically significant trends in respiratory mechanics endpoints (airway pressure, compliance, tidal volume, and PaO2/FiO2) in the TTM group. The PaCO2 decreased significantly more in the TTM group, as compared with controls (-12 vs. -5 mmHg, p=0.02). For clinical outcomes, the TTM group consistently, although not significantly, did better in survival (59% vs. 43%) and hospital length of stay (12 vs. 15 days). The MV duration and Cerebral Performance Category score on discharge were significantly lower in the TTM group (7.3 vs. 10.7 days, p=0.04 and 3.2 vs. 4, p=0.01). This small retrospective cohort suggests that the effect of TTM ranges from equivalent to favorable, compared with controls, for the specific respiratory and clinical outcomes in patients with respiratory failure following cardiac arrest.
Collapse
|
11
|
Cruces P, Erranz B, Donoso A, Carvajal C, Salomón T, Torres MF, Díaz F. Mild hypothermia increases pulmonary anti-inflammatory response during protective mechanical ventilation in a piglet model of acute lung injury. Paediatr Anaesth 2013; 23:1069-77. [PMID: 23731357 DOI: 10.1111/pan.12209] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/08/2013] [Indexed: 12/15/2022]
Abstract
BACKGROUND The effects of mild hypothermia (HT) on acute lung injury (ALI) are unknown in species with metabolic rate similar to that of humans, receiving protective mechanical ventilation (MV). We hypothesized that mild hypothermia would attenuate pulmonary and systemic inflammatory responses in piglets with ALI managed with a protective MV. METHODS Acute lung injury (ALI) was induced with surfactant deactivation in 38 piglets. The animals were then ventilated with low tidal volume, moderate positive end-expiratory pressure (PEEP), and permissive hypercapnia throughout the experiment. Subjects were randomized to HT (33.5°C) or normothermia (37°C) groups over 4 h. Plasma and tissue cytokines, tissue apoptosis, lung mechanics, pulmonary vascular permeability, hemodynamic, and coagulation were evaluated. RESULTS Lung interleukin-10 concentrations were higher in subjects that underwent HT after ALI induction than in those that maintained normothermia. No difference was found in other systemic and tissue cytokines. HT did not induce lung or kidney tissue apoptosis or influence lung mechanics or markers of pulmonary vascular permeability. Heart rate, cardiac output, oxygen uptake, and delivery were significantly lower in subjects that underwent HT, but no difference in arterial lactate, central venous oxygen saturation, and coagulation test was observed. CONCLUSIONS Mild hypothermia induced a local anti-inflammatory response in the lungs, without affecting lung function or coagulation, in this piglet model of ALI. The HT group had lower cardiac output without signs of global dysoxia, suggesting an adaptation to the decrease in oxygen uptake and delivery. Studies are needed to determine the therapeutic role of HT in ALI.
Collapse
Affiliation(s)
- Pablo Cruces
- Área de Cuidados Críticos, Hospital Padre Hurtado, Santiago, Chile; Centro de Investigación de Medicina Veterinaria, Escuela de Medicina Veterinaria, Facultad de Ecología y Recursos Naturales, Universidad Andres Bello, Santiago, Chile
| | | | | | | | | | | | | |
Collapse
|
12
|
Altınsoy C, Tuzun F, Duman N, Sever AH, Dilek M, Ozbal S, Ergur BU, Yesilirmak DC, Yılmaz O, Kumral A, Ozkan H. Effect of induced hypothermia on lipopolysaccharide-induced lung injury in neonatal rats. J Matern Fetal Neonatal Med 2013; 27:421-9. [DOI: 10.3109/14767058.2013.818115] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
13
|
Chiou SY, Lee YS, Jeng MJ, Tsao PC, Soong WJ. Moderate hypothermia attenuates oxidative stress injuries in alveolar epithelial A549 cells. Exp Lung Res 2013; 39:217-28. [PMID: 23647088 DOI: 10.3109/01902148.2013.792881] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Reactive oxygen species (ROS) are generally involved in lung inflammation and acute lung injury. We investigated the effects of hypothermia on ROS-induced cell damage in human alveolar type II cells. A549 cells were exposed to H2O2 and cultured at different temperatures, namely, normthermia (37°C), mild hypothermia (34°C), or moderate hypothermia (32°C). Cell damage was measured using various assays. The biochemical studies demonstrated a significant increase in apoptosis and intracellular ROS at 32°C in uninjured A549 cells. After exposure to H2O2, a marked decrease in cell viability (<50%) was demonstrated, and this was significantly ameliorated upon culture at 32°C. Significantly intracellular damage was found to affect the 24-hour H2O2-exposed cells in 37°C (P < .05), including an increase in apoptosis and necrosis, intracellular ROS, caspase-3 activity, HMGB1 protein expression, and some alterations to the cell cycle. On hypothermic treatment, the 24-hour H2O2-induced caspase-3 activation was significantly suppressed in cells cultured at both 32°C and 34°C (P < .05 versus 37°C). The cell cycle changes in 24-hour H2O2-exposed cells were significantly diminished when the cells were cultured in 32°C (P < .05 versus 37°C). However, these intracellular alterations were not seen in 6-hour H2O2-exposed cells. We concluded that moderate hypothermia (32°C) of alveolar epithelial A549 cells seems to provide protection against H2O2-induced 24-hour oxidative stress by attenuating cell death and intracellular damage. However, moderate hypothermia might cause minor damage to uninjured cells, so the use of hypothermic treatment needs to be judiciously applied.
Collapse
Affiliation(s)
- Shr-Yun Chiou
- Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan, ROC
| | | | | | | | | |
Collapse
|
14
|
Aslami H, Kuipers MT, Beurskens CJP, Roelofs JJTH, Schultz MJ, Juffermans NP. Mild hypothermia reduces ventilator-induced lung injury, irrespective of reducing respiratory rate. Transl Res 2012; 159:110-7. [PMID: 22243795 DOI: 10.1016/j.trsl.2011.10.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 10/13/2011] [Accepted: 10/14/2011] [Indexed: 10/15/2022]
Abstract
In the era of lung-protective mechanical ventilation using limited tidal volumes, higher respiratory rates are applied to maintain adequate minute volume ventilation. However, higher respiratory rates may contribute to ventilator-induced lung injury (VILI). Induced hypothermia reduces carbon dioxide production and might allow for lower respiratory rates during mechanical ventilation. We hypothesized that hypothermia protects from VILI and investigated whether reducing respiratory rates enhance lung protection in an in vivo model of VILI. During 4 h of mechanical ventilation, VILI was induced by tidal volumes of 18 mL/kg in rats, with respiratory rates set at 15 or 10 breaths/min in combination with hypothermia (32°C) or normothermia (37°C). Hypothermia was induced by external cooling. A physiologic model was established. VILI was characterized by increased pulmonary neutrophil influx, protein leak, wet weights, histopathology score, and cytokine levels compared with lung protective mechanical ventilation. Hypothermia decreased neutrophil influx, pulmonary levels, systemic interleukin-6 levels, and histopathology score, and it tended to decrease the pulmonary protein leak. Reducing the respiratory rate in combination with hypothermia did not reduce the parameters of the lung injury. In conclusion, hypothermia protected from lung injury in a physiologic VILI model by reducing inflammation. Decreasing the respiratory rate mildly did not enhance protection.
Collapse
Affiliation(s)
- Hamid Aslami
- Laboratory of Experimental Intensive Care and Anesthesiology, Academic Medical Center, Meibergdreef 9, Amsterdam, the Netherlands.
| | | | | | | | | | | |
Collapse
|
15
|
Cruces P, Ronco R, Erranz B, Conget P, Carvajal C, Donoso A, Díaz F. Mild hypothermia attenuates lung edema and plasma interleukin-1β in a rat mechanical ventilation-induced lung injury model. Exp Lung Res 2011; 37:549-54. [DOI: 10.3109/01902148.2011.616983] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
16
|
Hoegl S, Bachmann M, Scheiermann P, Goren I, Hofstetter C, Pfeilschifter J, Zwissler B, Muhl H. Protective properties of inhaled IL-22 in a model of ventilator-induced lung injury. Am J Respir Cell Mol Biol 2011; 44:369-76. [PMID: 20463292 DOI: 10.1165/rcmb.2009-0440oc] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
High-pressure ventilation induces barotrauma and pulmonary inflammation, thus leading to ventilator-induced lung injury (VILI). IL-22 has both immunoregulatory and tissue-protective properties. Functional IL-22 receptor expression is restricted to nonleukocytic cells, such as alveolar epithelial cells. When applied via inhalation, IL-22 reaches the pulmonary system directly and in high concentrations, and may protect alveolar epithelial cells against cellular stress and biotrauma associated with VILI. In A549 lung epithelial cells, IL-22 was able to induce rapid signal transducer and activator of transcription (STAT)-3 phosphorylation/activation, and hereon mediated stable suppressor of cytokine signaling (SOCS) 3 expression detectable even 24 hours after onset of stimulation. In a rat model of VILI, the prophylactic inhalation of IL-22 before induction of VILI (peak airway pressure = 45 cm H(2)O) protected the lung against pulmonary disintegration and edema. IL-22 reduced VILI-associated biotrauma (i.e., pulmonary concentrations of macrophage inflammatory protein-2, IL-6, and matrix metalloproteinase 9) and mediated pulmonary STAT3/SOCS3 activation. In addition, despite a short observation period of 4 hours, inhaled IL-22 resulted in an improved survival of the rats. These data support the hypothesis that IL-22, likely via activation of STAT3 and downstream genes (e.g., SOCS3), is able to protect against cell stretch and pulmonary baro-/biotrauma by enhancing epithelial cell resistibility.
Collapse
Affiliation(s)
- Sandra Hoegl
- Clinic for Anesthesiology, University Hospital of Ludwig-Maximilians-University, Munich, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
BACKGROUND In previous animal studies, induction of therapeutic hypothermia (HT) in hemorrhagic shock (HS) had beneficial effects on the hemodynamic and metabolic parameters and on the survival. However, the effect of induced HT on acute lung injury (ALI) in HS has not been investigated. We sought to determine the effects of HT on ALI in HS. METHODS Male Sprague-Dawley rats (350-390 g; n = 8 per group) were randomized to the normothermia (NT; 36-37 degrees C) group or the moderate HT (27-30 degrees C) group and were subjected to volume-controlled (2 mL/100 g weight) HS (90 minutes) followed by 90 minutes of resuscitation. ALI score, lung malondialdehyde content, and myeloperoxidase activity were measured. The expression of glycogen synthase kinase 3beta (GSK-3beta), phosphorylated GSK-3beta, inducible nitric oxide synthase (iNOS), heat shock protein (HSP) 72, and nuclear factor-kappaB (NF-kappaB) in the lung were compared. RESULTS ALI score, lung malondialdehyde content, and myeloperoxidase were lower in the HT group. GSK-3beta and iNOS gene expressions in lung tissue were significantly decreased in the HT group (p < 0.05). On the contrary, the expression of phosphorylated GSK-3beta was increased in the HT group (p < 0.001). HSP 72 was expressed in the HT group but not in the NT group. The activated p65 NF-kappaB levels in lung nuclear extract were significantly lower in the NT group (p = 0.03). CONCLUSIONS HT attenuates HS-induced ALI in rats by the modulation of GSK, HSP 72, iNOS, and NF-kappaB.
Collapse
|
18
|
Yang HH, Chang CP, Cheng RT, Lin MT. Attenuation of acute lung inflammation and injury by whole body cooling in a rat heatstroke model. J Biomed Biotechnol 2009; 2009:768086. [PMID: 20037732 PMCID: PMC2796336 DOI: 10.1155/2009/768086] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2009] [Revised: 09/25/2009] [Accepted: 10/01/2009] [Indexed: 12/21/2022] Open
Abstract
Whole body cooling is the current therapy of choice for heatstroke because the therapeutic agents are not available. In this study, we assessed the effects of whole body cooling on several indices of acute lung inflammation and injury which might occur during heatstroke. Anesthetized rats were randomized into the following groups and given (a) no treatment or (b) whole body cooling immediately after onset of heatstroke. As compared with the normothermic controls, the untreated heatstroke rats had higher levels of pleural exudates volume and polymorphonuclear cell numbers, lung myloperoxidase activity and inducible nitric oxide synthase expression, histologic lung injury score, and bronchoalveolar proinflammatory cytokines and glutamate, and PaCO2. In contrast, the values of mean arterial pressure, heart rate, PaO2, pH, and blood HCO3(-) were all significantly lower during heatstroke. The acute lung inflammation and injury and electrolyte imbalance that occurred during heatstroke were significantly reduced by whole body cooling. In conclusion, we identified heat-induced acute lung inflammation and injury and electrolyte imbalance could be ameliorated by whole body cooling.
Collapse
Affiliation(s)
- Hsi-Hsing Yang
- Institute of Pharmacology, National Cheng Kung University Medical School, Tainan 701, Taiwan
- Department of Internal Medicine, Chi Mei Medical Center, Tainan 710, Taiwan
| | - Ching-Ping Chang
- Department of Biotechnology, Southern Taiwan University, Tainan 710, Taiwan
| | - Ruei-Tang Cheng
- Department of Medical Research, Chi Mei Medical Center, Tainan 710, Taiwan
| | - Mao-Tsun Lin
- Department of Medical Research, Chi Mei Medical Center, Tainan 710, Taiwan
| |
Collapse
|
19
|
Inhaled IL-10 reduces biotrauma and mortality in a model of ventilator-induced lung injury. Respir Med 2008; 103:463-70. [PMID: 19006658 DOI: 10.1016/j.rmed.2008.09.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Revised: 09/08/2008] [Accepted: 09/15/2008] [Indexed: 11/22/2022]
Abstract
BACKGROUND High-pressure ventilation induces barotrauma and pulmonary inflammation, thus leading to ventilator-induced lung injury (VILI). By limiting the pulmonal inflammation cascade the anti-inflammatory cytokine interleukin (IL)-10 may have protective effects. Via inhalation, IL-10 reaches the pulmonary system directly and in high concentrations. METHODS Thirty six male, anesthetized and mechanically ventilated Sprague-Dawley rats were randomly assigned to the following groups (n=9, each): SHAM: pressure controlled ventilation with p(max)=20cmH(2)O, PEEP=4; VILI: ventilator settings were changed for 20min to p(max)=45cmH(2)O, PEEP=0; IL-10(high): inhalation of 10microg/kg IL-10 prior to induction of VILI; and IL-10(low): inhalation of 1microg/kg IL-10 prior to induction of VILI. All groups were ventilated and observed for 4h. RESULTS High-pressure ventilation increased the concentrations of macrophage inflammatory protein (MIP)-2 and IL-1beta in bronchoalveolar lavage fluid (BALF) and plasma. This effect was reduced by the inhalation of IL-10 (10microg/kg). Additionally, IL-10 increased the animal survival time (78% vs. 22% 4-h mortality rate) and reduced NO-release from ex vivo cultured alveolar macrophages. Moreover, VILI-induced pulmonary heat shock protein-70 expression was reduced by IL-10 aerosol in a dose-dependent manner. Similarly, the activation of matrix metalloproteinase (MMP)-9 in BALF was reduced dose-dependently by IL-10. IL-10-treated animals showed a lower macroscopic lung injury score and less impairment of lung integrity and gas exchange. CONCLUSIONS Prophylactic inhalation of IL-10 improved survival and reduced lung injury in experimental VILI. Results indicate that this effect may be mediated by the inhibition of stress-induced inflammation and pulmonary biotrauma.
Collapse
|
20
|
Kumral A, Yesilirmak D, Tuzun F, Duman N, Ozkan H. Induced hypothermia as a new approach to bronchopulmonary dysplasia. Med Hypotheses 2008; 71:617-8. [DOI: 10.1016/j.mehy.2008.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Revised: 05/02/2008] [Accepted: 05/04/2008] [Indexed: 10/21/2022]
|
21
|
Boost KA, Hoegl S, Dolfen A, Czerwonka H, Scheiermann P, Zwissler B, Hofstetter C. Inhaled levosimendan reduces mortality and release of proinflammatory mediators in a rat model of experimental ventilator-induced lung injury*. Crit Care Med 2008; 36:1873-9. [DOI: 10.1097/ccm.0b013e3181743e63] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
22
|
Hamanaka K, Jian MY, Weber DS, Alvarez DF, Townsley MI, Al-Mehdi AB, King JA, Liedtke W, Parker JC. TRPV4 initiates the acute calcium-dependent permeability increase during ventilator-induced lung injury in isolated mouse lungs. Am J Physiol Lung Cell Mol Physiol 2007; 293:L923-32. [PMID: 17660328 DOI: 10.1152/ajplung.00221.2007] [Citation(s) in RCA: 148] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We have previously implicated calcium entry through stretch-activated cation channels in initiating the acute pulmonary vascular permeability increase in response to high peak inflation pressure (PIP) ventilation. However, the molecular identity of the channel is not known. We hypothesized that the transient receptor potential vanilloid-4 (TRPV4) channel may initiate this acute permeability increase because endothelial calcium entry through TRPV4 channels occurs in response to hypotonic mechanical stress, heat, and P-450 epoxygenase metabolites of arachidonic acid. Therefore, permeability was assessed by measuring the filtration coefficient (K(f)) in isolated perfused lungs of C57BL/6 mice after 30-min ventilation periods of 9, 25, and 35 cmH(2)O PIP at both 35 degrees C and 40 degrees C. Ventilation with 35 cmH(2)O PIP increased K(f) by 2.2-fold at 35 degrees C and 3.3-fold at 40 degrees C compared with baseline, but K(f) increased significantly with time at 40 degrees C with 9 cmH(2)O PIP. Pretreatment with inhibitors of TRPV4 (ruthenium red), arachidonic acid production (methanandamide), or P-450 epoxygenases (miconazole) prevented the increases in K(f). In TRPV4(-/-) knockout mice, the high PIP ventilation protocol did not increase K(f) at either temperature. We have also found that lung distention caused Ca(2+) entry in isolated mouse lungs, as measured by ratiometric fluorescence microscopy, which was absent in TRPV4(-/-) and ruthenium red-treated lungs. Alveolar and perivascular edema was significantly reduced in TRPV4(-/-) lungs. We conclude that rapid calcium entry through TRPV4 channels is a major determinant of the acute vascular permeability increase in lungs following high PIP ventilation.
Collapse
Affiliation(s)
- Kazutoshi Hamanaka
- Department of Physiology, College of Medicine, MSB 3074, University of South Alabama, 307 Univ. Blvd., Mobile, AL 36688, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Hotchkiss JR, Broccard AF. Modulating cofactors of acute lung injury 2005–2006: any closer to ‘prime time’? Curr Opin Crit Care 2007; 13:39-44. [PMID: 17198047 DOI: 10.1097/mcc.0b013e328012c599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Considerable progress has recently been made in understanding the modulation of acute lung injury by cofactors that are not traditionally considered 'pulmonary' in nature. We will review findings regarding some of these extrapulmonary cofactors, focusing on those most readily manipulated in the current clinical setting. RECENT FINDINGS Recent studies have demonstrated that limiting fluid administration in the setting of acute lung injury might improve surrogate outcomes; that hypercapnea and induced hypothermia might protect against or attenuate acute lung injury; that corticosteroids can improve mechanics but not mortality in acute respiratory distress syndrome; a potential role for concomitant administration of colloid and diuretic in acute lung injury; and the potential benefits of inhaled beta agonists in acute lung injury. SUMMARY There are a number of simple, low-cost, and rapidly deployable approaches to reducing the severity of acute lung injury that are not directly pulmonary in origin. These interventions could be rapidly implemented in any intensive care unit, once evidence for their efficacy and safety is adequate.
Collapse
Affiliation(s)
- John R Hotchkiss
- University of Pittsburgh, Pittsburgh, Pennsylvania, PA 15261, USA.
| | | |
Collapse
|
24
|
López-Aguilar J, Piacentini E, Villagrá A, Murias G, Pascotto S, Saenz-Valiente A, Fernández-Segoviano P, Hotchkiss JR, Blanch L. Contributions of vascular flow and pulmonary capillary pressure to ventilator-induced lung injury. Crit Care Med 2006; 34:1106-12. [PMID: 16484897 DOI: 10.1097/01.ccm.0000205757.66971.da] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To evaluate the influence of vascular flow on ventilator-induced lung injury independent of vascular pressures. DESIGN Laboratory study. SETTING Hospital laboratory. SUBJECTS Thirty-two New Zealand White rabbits. INTERVENTIONS Thirty-two isolated perfused rabbit lungs were allocated into four groups: low flow/low pulmonary capillary pressure; high flow/high pulmonary capillary pressure; low flow/high pulmonary capillary pressure, and high flow/low pulmonary capillary pressure. All lungs were ventilated with peak airway pressure 30 cm H2O and positive end-expiratory pressure 5 cm H2O for 30 mins. MEASUREMENTS AND MAIN RESULTS Outcome measures included frequency of gross structural failure (pulmonary rupture), pulmonary hemorrhage, edema formation, changes in lung compliance, pulmonary vascular resistance, and pulmonary ultrafiltration coefficient. Lungs exposed to high pulmonary vascular flow ruptured more frequently, displayed more hemorrhage, developed more edema, suffered larger decreases in compliance, and had larger increases in vascular resistance than lungs exposed to low vascular flows (p < .05 for each pairwise comparison between groups). CONCLUSIONS These findings suggest that high pulmonary vascular flows might exacerbate ventilator-induced lung injury independent of their effects on pulmonary vascular pressures.
Collapse
Affiliation(s)
- Josefina López-Aguilar
- Critical Care Center, Hospital de Sabadell, Institut Universitari Parc Taulí, Universitat Autónoma de Barcelona, Esfera UAB, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Kira S, Mori M, Takatani J, Uchino T, Yasuda N, Miyakawa H, Noguchi T. Effects of high peak airway pressure on the expression of heat shock protein 70 in rat lungs: a preliminary study. Acta Anaesthesiol Scand 2006; 50:469-74. [PMID: 16548859 DOI: 10.1111/j.1399-6576.2005.00942.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Heat shock protein 70 (HSP70) is induced by a wide variety of stresses in addition to hyperthermia. Recent studies have clarified that mechanical stretching and pressure overload can induce HSP70 in some tissues and cells. However, it remains unclear whether HSP70 is induced in stretch-subjected lungs, such as those under mechanical ventilation. This study was designed to investigate the effects of high peak airway pressure (PAP) ventilation on HSP70 expression in intact rat lungs. METHODS Male Sprague-Dawley rats were randomly allocated to one of three groups: non-ventilated (anesthesia alone) control group; PAP 15 cm H(2)O group (P15); and PAP 30 cm H(2)O group (P30). The rats in the PAP groups were subjected to pressure-controlled assisted ventilation at the appropriate PAP for 30 min. Rats were killed at 12, 24 and 48 h after ventilation or anesthesia alone, and the lungs were removed. The lung tissues were processed for immunohistochemical and Western blotting analyses of HSP70. RESULTS Following 30 min of pressure-controlled assisted ventilation, HSP70 expression in the P30 group was significantly up-regulated in bronchiolar cells and subepithelial tissues at 12 h, and this up-regulation continued throughout the observation period. In contrast, there were no significant differences between the control and P15 groups, although the expression of HSP70 was higher in the P15 group than in the control group at all time points. CONCLUSIONS HSP70 was induced by high PAP ventilation, but its specific role and induction mechanism remain unclear. Therefore, further investigations should be encouraged.
Collapse
Affiliation(s)
- S Kira
- Department of Anesthesiology, Oita University Faculty of Medicine, Hasama-machi, Oita, Japan.
| | | | | | | | | | | | | |
Collapse
|
26
|
Huang PS, Tang GJ, Chen CH, Kou YR. Whole-body moderate hypothermia confers protection from wood smoke-induced acute lung injury in rats: The therapeutic window*. Crit Care Med 2006; 34:1160-7. [PMID: 16484924 DOI: 10.1097/01.ccm.0000207342.50559.0f] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVE Toxic smoke inhalation causes acute lung injury. We studied the efficacy and therapeutic window of whole-body hypothermia in rats with wood smoke-induced acute lung injury. DESIGN Randomized, controlled study. SETTING Research laboratory. SUBJECTS Anesthetized, paralyzed, and artificially ventilated rats (n = 100) were used. INTERVENTIONS Air or wood smoke (30 breaths) was delivered into the lung using a respirator. Immediately after challenge, the rat's colonic temperature was kept a) 37 degrees C (normothermia, NT) for 1 (NT-1-Air and NT-1-Smoke), 2.5 (NT-2.5-Air and NT-2.5-Smoke), or 5 hrs (NT-5-Air and NT-5-Smoke) in six groups; b) 30 degrees C (hypothermia, HT) for 2.5 (HT-2.5-Smoke) or 5 hrs (HT-5-Air and HT-5-Smoke) in three groups; c) 30 degrees C for the first 2.5 hrs followed by 37 degrees C for another 2.5 hrs (HT-NT-5-Smoke) in one group; or d) 37 degrees C for the first 2.5 hrs followed by 30 degrees C for another 2.5 hrs (NT-HT-5-Smoke) in on group. MEASUREMENTS AND MAIN RESULTS Various acute lung injury indexes were assessed at 1, 2.5, or 5 hrs after challenge. In the air group, whole-body hypothermia did not affect the level of lung lipid peroxidation and the amount of proteins, total and differential cell counts, and concentrations of tumor necrosis factor-alpha and interleukin-1beta in bronchoalveolar lavage fluid. In the smoke groups, these acute lung injury indexes were increased showing that NT-5-Smoke > NT-2.5-Smoke > NT-1-Smoke. Whole-body hypothermia prevented increases in these acute lung injury indexes in the HT-2.5-Smoke and HT-5-Smoke groups. The efficacy of whole-body hypothermia in the HT-NT-5-Smoke group was superior to that in the NT-HT-5-Smoke group and similar to that in the HT-5-Smoke group. Whole-body hypothermia also alleviated smoke-induced poor gas exchange, pulmonary edema, and pathohistologic injurious signs. CONCLUSIONS Whole-body hypothermia confers protection from wood smoke-induced acute lung injury in rats by suppressing oxidant bronchoalveolar damage and pulmonary inflammation. Early and short-period (2 hrs) application of whole-body hypothermia provides favorable therapeutic effects.
Collapse
Affiliation(s)
- Pin-Shiun Huang
- Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | | | | | | |
Collapse
|
27
|
Hong SB, Koh Y, Lee IC, Kim MJ, Kim WS, Kim DS, Kim WD, Lim CM. Induced hypothermia as a new approach to lung rest for the acutely injured lung*. Crit Care Med 2005; 33:2049-55. [PMID: 16148479 DOI: 10.1097/01.ccm.0000178186.37167.53] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To investigate whether low-frequency ventilation during hypothermia could attenuate lung injury associated with endotoxin and mechanical ventilation. DESIGN : Experimental animal study. SETTING University-affiliated animal laboratory. SUBJECTS Forty-eight Sprague-Dawley rats. INTERVENTIONS : Lipopolysaccharide was administered to rats intratracheally to induce acute lung injury. After 1 hr of this treatment, animals were assigned to normothermia-only (NO, rectal temperature 37 degrees C, ventilatory frequency 90/min), normothermia-lung rest (NR, 37 degrees C, 45/min), hypothermia-only (HO, 27 degrees C, 90/min), or hypothermia-lung rest (HR, 27 degrees C, 45/min). After 1 hr of injurious ventilation, the lungs of the rats were removed for bronchoalveolar lavage and histologic examination. MEASUREMENTS AND MAIN RESULTS Compared with the normothermia groups (NO, NR), the neutrophil counts (per milliliter) (NO, 7708 +/- 5704; NR, 10,479 +/- 11,152; HO, 1638 +/- 955; HR, 805 +/- 591) and interleukin-1beta levels (pg/mL) (1180 +/- 439, 1081 +/- 652, 620 +/- 426, 420 +/- 182, respectively) in the bronchoalveolar lavage fluid, the wet-to-dry lung weight ratios (6.0 +/- 0.4, 5.7 +/- 0.4, 5.6 +/- 0.2, 5.2 +/- 0.2, respectively), and histologic acute lung injury scores (8.3 +/- 2.7, 10.4 +/- 3.1, 3.5 +/- 2.1, 3.1 +/- 2.2, respectively) of the hypothermia groups (HO, HR) were lower (all p < .001). Compared with the HO group, the neutrophil counts and protein content (HO, 1367 +/- 490 mug/mL vs. HR, 831 +/- 369 mug/mL) in the bronchoalveolar lavage fluid, the serum lactate dehydrogenase levels (units/mL) (9.1 +/- 3.6 vs. 5.3 +/- 1.5), and the wet-to-dry lung weight ratios of the HR group were lower (all p < .05). CONCLUSIONS Reduction of ventilatory frequency in conjunction with hypothermia attenuated many variables of acute lung injury in rats. Use of hypothermia could be exploited as a new approach to lung rest for the ventilatory management of the acutely injured lung.
Collapse
Affiliation(s)
- Sang-Bum Hong
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Hotchkiss JR, Gunn SR. Simple approaches to a complex problem: Combining interventions to limit ventilator-induced lung injury*. Crit Care Med 2005; 33:2138-40. [PMID: 16148503 DOI: 10.1097/01.ccm.0000178343.79275.a8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
29
|
Akinci OI, Celik M, Mutlu GM, Martino JM, Tugrul S, Ozcan PE, Yilmazbayhan D, Yeldandi AV, Turkoz KH, Kiran B, Telci L, Cakar N. Effects of body temperature on ventilator-induced lung injury. J Crit Care 2005; 20:66-73. [PMID: 16015518 DOI: 10.1016/j.jcrc.2004.11.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE To evaluate the effects of body temperature on ventilator-induced lung injury. MATERIAL AND METHODS Thirty-four male Sprague-Dawley rats were randomized into 6 groups based on their body temperature (normothermia, 37 +/- 1 degrees C; hypothermia, 31 +/- 1 degrees C; hyperthermia, 41 +/- 1 degrees C). Ventilator-induced lung injury was achieved by ventilating for 1 hour with pressure-controlled ventilation mode set at peak inspiratory pressure (PIP) of 30 cmH2O (high pressure, or HP) and positive end-expiratory pressure (PEEP) of 0 cmH2O. In control subjects, PIP was set at 14 cmH2O (low pressure, or LP) and PEEP set at 0 cmH2O. Systemic chemokine and cytokine (tumor necrosis factor alpha , interleukin 1 beta , interleukin 6, and monocyte chemoattractant protein 1) levels were measured. The lungs were assessed for histological changes. RESULTS Serum chemokines and cytokines were significantly elevated in the hyperthermia HP group compared with all 3 groups, LP (control), normothermia HP, and hypothermia HP. Oxygenation was better but not statistically significant in hypothermia HP compared with other HP groups. Cumulative mean histology scores were higher in hyperthermia HP and normothermia HP groups compared with control and normothermia HP groups. CONCLUSIONS Concomitant hyperthermia increased systemic inflammatory response during HP ventilation. Although hypothermia decreased local inflammation in the lung, it did not completely attenuate systemic inflammatory response associated with HP ventilation.
Collapse
Affiliation(s)
- Ozkan I Akinci
- Department of Anesthesiology and Intensive Care, Istanbul Medical Faculty, Capa, Turkey.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Chu SJ, Perng WC, Hung CM, Chang DM, Lin SH, Huang KL. Effects of Various Body Temperatures After Lipopolysaccharide-Induced Lung Injury in Rats. Chest 2005. [DOI: 10.1016/s0012-3692(15)37965-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|