1
|
Campaña-Duel E, Ceccato A, Morales-Quinteros L, Camprubí-Rimblas M, Artigas A. Hypercapnia and its relationship with respiratory infections. Expert Rev Respir Med 2024; 18:41-47. [PMID: 38489161 DOI: 10.1080/17476348.2024.2331767] [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: 11/13/2023] [Accepted: 03/13/2024] [Indexed: 03/17/2024]
Abstract
INTRODUCTION Hypercapnia is developed in patients with acute and/or chronic respiratory conditions. Clinical data concerning hypercapnia and respiratory infections interaction is limited. AREAS COVERED Currently, the relationship between hypercapnia and respiratory infections remains unclear. In this review, we summarize studies on the effects of hypercapnia on models of pulmonary infections to clarify the role of elevated CO2 in these pulmonary pathologies. Hypercapnia affects different cell types in the alveoli, leading to changes in the immune response. In vitro studies show that hypercapnia downregulates the NF-κβ pathway, reduces inflammation and impairs epithelial wound healing. While in vivo models show a dual role between short- and long-term effects of hypercapnia on lung infection. However, it is still controversial whether the effects observed under hypercapnia are pH dependent or not. EXPERT OPINION The role of hypercapnia is still a controversial debate. Hypercapnia could play a beneficial role in mechanically ventilated models, by lowering the inflammation produced by the stretch condition. But it could be detrimental in infectious scenarios, causing phagocyte dysfunction and lack of infection control. Further data concerning hypercapnia on respiratory infections is needed to elucidate this interaction.
Collapse
Affiliation(s)
- Elena Campaña-Duel
- Critical care center, Parc Taulí Hospital Universitari. Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA). Universitat Autònoma de Barcelona, Sabadell, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
| | - Adrian Ceccato
- Critical care center, Parc Taulí Hospital Universitari. Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA). Universitat Autònoma de Barcelona, Sabadell, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
- Intensive care unit, Hospital Universitari Sagrat Cor, Grupo Quironsalud, Barcelona, Spain
| | - Luis Morales-Quinteros
- Critical care center, Parc Taulí Hospital Universitari. Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA). Universitat Autònoma de Barcelona, Sabadell, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
- Servei de Medicina Intensiva, Hospital de la Santa Creu y Sant Pau, Barcelona, Spain
| | - Marta Camprubí-Rimblas
- Critical care center, Parc Taulí Hospital Universitari. Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA). Universitat Autònoma de Barcelona, Sabadell, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
| | - Antonio Artigas
- Critical care center, Parc Taulí Hospital Universitari. Institut d'Investigació i Innovació Parc Taulí (I3PT-CERCA). Universitat Autònoma de Barcelona, Sabadell, Spain
- CIBER of Respiratory Diseases (CIBERES), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
2
|
Intermittent Exposure of Hypercapnia Suppresses Allograft Rejection via Induction of Treg Differentiation and Inhibition of Neutrophil Accumulation. Biomedicines 2022; 10:biomedicines10040836. [PMID: 35453586 PMCID: PMC9028437 DOI: 10.3390/biomedicines10040836] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/09/2022] [Accepted: 03/30/2022] [Indexed: 11/17/2022] Open
Abstract
Background: In the management of major burn wounds, allogeneic skin transplantation is a critical procedure to improve wound repair. Our previous works found that intermittent exposure to carbon dioxide leads to permissive hypercapnia (HCA) and prolongs skin allograft survival. However, the modulatory effects of HCA exposure on the immune system are not well understood. Objectives: Our purpose was to investigate how intermittent exposure to HCA can effectively reduce the immune reaction to allogeneic skin graft rejection. Methods: A fully major histocompatibility complex-incompatible skin transplant from BALB/c to C57BL/6 mice model was utilized. Immune cells from splenic and draining lymph nodes were analyzed by flow cytometry. Serum proinflammatory cytokines were analyzed by ELISA. Results: Serum levels of IFN-γ, IL-2, IL-6, and TNF-α were significantly decreased in the HCA group. Additionally, the percentage of CD8+ cells in draining lymph nodes was significantly lower in HCA than in the control group. Moreover, the generation rate of FoxP3+ regulatory T cells (Tregs) from spleen naïve CD4+ T cells was increased by intermittent exposure to carbon dioxide. The infiltrated neutrophils were also eliminated by HCA. Taken together, we concluded that intermittent hypercapnia exposure could effectively suppress skin rejection by stimulating Treg cell generation and suppressing immune reactions.
Collapse
|
3
|
Zhan Y, Ling Y, Deng Q, Qiu Y, Shen J, Lai H, Chen Z, Huang C, Liang L, Li X, Wu J, Huang W, Wen S. HMGB1-Mediated Neutrophil Extracellular Trap Formation Exacerbates Intestinal Ischemia/Reperfusion-Induced Acute Lung Injury. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:968-978. [PMID: 35063996 DOI: 10.4049/jimmunol.2100593] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022]
Abstract
Influx of activated neutrophils into the lungs is the histopathologic hallmark of acute lung injury (ALI) after intestinal ischemia/reperfusion (I/R). Neutrophils can release DNA and granular proteins to form cytotoxic neutrophil extracellular traps (NETs), which promotes bystander tissue injury. However, whether NETs are responsible for the remote ALI after intestinal I/R and the mechanisms underlying the dissemination of harmful gut-derived mediators to the lungs are unknown. In the C57BL/6J mouse intestinal I/R model, DNase I-mediated degradation and protein arginine deiminase 4 (PAD4) inhibitor-mediated inhibition of NET treatments reduced NET formation, tissue inflammation, and pathological injury in the lung. High-mobility group protein B1 (HMGB1) blocking prevented NET formation and protected against tissue inflammation, as well as reduced cell apoptosis and improved survival rate. Moreover, recombinant human HMGB1 administration further drives NETs and concurrent tissue toxic injury, which in turn can be reversed by neutrophil deletion via anti-Ly6G Ab i.p. injection. Furthermore, global MyD88 deficiency regulated NET formation and alleviated the development of ALI induced by intestinal I/R. Thus, HMGB1 released from necroptotic enterocytes caused ALI after intestinal I/R by inducing NET formation. Targeting NETosis and the HMGB1 pathway might extend effective therapeutic strategies to minimize intestinal I/R-induced ALI.
Collapse
Affiliation(s)
- YaQing Zhan
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - YiHong Ling
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qiwen Deng
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - YuXin Qiu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - JianTong Shen
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - HanJin Lai
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - ZhaoRong Chen
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - ChanYan Huang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - LiQun Liang
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; and
| | - Xiang Li
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - JianFeng Wu
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China; and
| | - WenQi Huang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China;
| | - ShiHong Wen
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China;
| |
Collapse
|
4
|
Tiruvoipati R, Serpa Neto A, Young M, Marhoon N, Wilson J, Gupta S, Pilcher D, Bailey M, Bellomo R. An Exploratory Analysis of the Association between Hypercapnia and Hospital Mortality in Critically Ill Patients with Sepsis. Ann Am Thorac Soc 2022; 19:245-254. [PMID: 34380007 DOI: 10.1513/annalsats.202102-104oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Rationale: Hypercapnia may affect the outcome of sepsis. Very few clinical studies conducted in noncritically ill patients have investigated the effects of hypercapnia and hypercapnic acidemia in the context of sepsis. The effect of hypercapnia in critically ill patients with sepsis remains inadequately studied. Objectives: To investigate the association of hypercapnia with hospital mortality in critically ill patients with sepsis. Methods: This is a retrospective study conducted in three tertiary public hospitals. Critically ill patients with sepsis from three intensive care units between January 2011 and May 2019 were included. Five cohorts (exposure of at least 24, 48, 72, 120, and 168 hours) were created to account for immortal time bias and informative censoring. The association between hypercapnia exposure and hospital mortality was assessed with multivariable models. Subgroup analyses compared ventilated versus nonventilated and pulmonary versus nonpulmonary sepsis patients. Results: We analyzed 84,819 arterial carbon dioxide pressure measurements in 3,153 patients (57.6% male; median age was 62.5 years). After adjustment for key confounders, both in mechanically ventilated and nonventilated patients and in patients with pulmonary or nonpulmonary sepsis, there was no independent association of hypercapnia with hospital mortality. In contrast, in ventilated patients, the presence of prolonged exposure to both hypercapnia and acidemia was associated with increased mortality (highest odds ratio of 16.5 for ⩾120 hours of potential exposure; P = 0.007). Conclusions: After adjustment, isolated hypercapnia was not associated with increased mortality in patients with sepsis, whereas prolonged hypercapnic acidemia was associated with increased risk of mortality. These hypothesis-generating observations suggest that as hypercapnia is not an independent risk factor for mortality, trials of permissive hypercapnia avoiding or minimizing acidemia in sepsis may be safe.
Collapse
Affiliation(s)
- Ravindranath Tiruvoipati
- Department of Intensive Care Medicine, Peninsula Health, Melbourne, Victoria, Australia
- Australian and New Zealand Intensive Care Research Centre, Peninsula Clinical School, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Ary Serpa Neto
- Australian and New Zealand Intensive Care Research Centre, Peninsula Clinical School, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Marcus Young
- Department of Intensive Care, Austin Health, Heidelberg, Victoria, Australia
| | - Nada Marhoon
- Department of Intensive Care, Austin Health, Heidelberg, Victoria, Australia
| | - John Wilson
- Peninsula Health Informatics, Frankston Hospital, Melbourne, Victoria, Australia
| | - Sachin Gupta
- Department of Intensive Care Medicine, Peninsula Health, Melbourne, Victoria, Australia
- Australian and New Zealand Intensive Care Research Centre, Peninsula Clinical School, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - David Pilcher
- Australian and New Zealand Intensive Care Research Centre, Peninsula Clinical School, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Intensive Care Medicine, The Alfred Hospital, Melbourne, Victoria, Australia; and
| | - Michael Bailey
- Australian and New Zealand Intensive Care Research Centre, Peninsula Clinical School, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Data Analytics Research and Evaluation, the University of Melbourne and Austin Hospital, Melbourne, Victoria, Australia
| | - Rinaldo Bellomo
- Australian and New Zealand Intensive Care Research Centre, Peninsula Clinical School, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Intensive Care, Austin Health, Heidelberg, Victoria, Australia
- Data Analytics Research and Evaluation, the University of Melbourne and Austin Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
5
|
Effect of acute isooxic hypercapnia on oxidative activity of systemic neutrophils in endotoxemic rabbits. Cent Eur J Immunol 2021; 46:47-53. [PMID: 33897283 PMCID: PMC8056343 DOI: 10.5114/ceji.2021.105245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 08/27/2020] [Indexed: 12/14/2022] Open
Abstract
Introduction Whether carbon dioxide (CO2) affects systemic oxidative phenomena under conditions of endotoxemia is not sufficiently clarified. The study aimed to assess the impact of moderate acute hypercapnia on the respiratory burst of circulating neutrophils in mechanically ventilated endotoxemic rabbits. Material and methods Twenty-four endotoxemic rabbits were mechanically ventilated with standard or CO2-enriched gas mixture in order to obtain isooxic hypercapnia. At a baseline point and following 180 min of hypercapnic ventilation, luminol-dependent chemiluminescence (CL) of circulating neutrophils and serum 2-thiobarbituric acid reactive substance (TBARS) concentrations were measured. Throughout the study, leukocyte and neutrophil counts, pH status, circulatory parameters and body temperature were also assessed. Results Following 180 min of hypercapnic ventilation, opsonized zymosan (OZ)-stimulated neutrophils showed lower CL vs. the control group (p = 0.004). Other parameters studied were not affected. Conclusions Short-term isooxic hypercapnia in endotoxemic rabbits preserves circulating neutrophil count pattern and reactive oxygen species (ROS) generation, but it may reduce phagocytosis.
Collapse
|
6
|
Masterson C, Horie S, McCarthy SD, Gonzalez H, Byrnes D, Brady J, Fandiño J, Laffey JG, O'Toole D. Hypercapnia in the critically ill: insights from the bench to the bedside. Interface Focus 2021; 11:20200032. [PMID: 33628425 PMCID: PMC7898152 DOI: 10.1098/rsfs.2020.0032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2020] [Indexed: 01/16/2023] Open
Abstract
Carbon dioxide (CO2) has long been considered, at best, a waste by-product of metabolism, and at worst, a toxic molecule with serious health consequences if physiological concentration is dysregulated. However, clinical observations have revealed that 'permissive' hypercapnia, the deliberate allowance of respiratory produced CO2 to remain in the patient, can have anti-inflammatory effects that may be beneficial in certain circumstances. In parallel, studies at the cell level have demonstrated the profound effect of CO2 on multiple diverse signalling pathways, be it the effect from CO2 itself specifically or from the associated acidosis it generates. At the whole organism level, it now appears likely that there are many biological sensing systems designed to respond to CO2 concentration and tailor respiratory and other responses to atmospheric or local levels. Animal models have been widely employed to study the changes in CO2 levels in various disease states and also to what extent permissive or even directly delivered CO2 can affect patient outcome. These findings have been advanced to the bedside at the same time that further clinical observations have been elucidated at the cell and animal level. Here we present a synopsis of the current understanding of how CO2 affects mammalian biological systems, with a particular emphasis on inflammatory pathways and diseases such as lung specific or systemic sepsis. We also explore some future directions and possibilities, such as direct control of blood CO2 levels, that could lead to improved clinical care in the future.
Collapse
|
7
|
Chand R, Swenson ER, Goldfarb DS. Sodium bicarbonate therapy for acute respiratory acidosis. Curr Opin Nephrol Hypertens 2021; 30:223-230. [PMID: 33395037 DOI: 10.1097/mnh.0000000000000687] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Respiratory acidosis is commonly present in patients with respiratory failure. The usual treatment of hypercapnia is to increase ventilation. During the recent surge of COVID-19, respiratory acidosis unresponsive to increased mechanical ventilatory support was common. Increasing mechanical ventilation comes at the expense of barotrauma and hemodynamic compromise from increasing positive end-expiratory pressures or minute ventilation. Treating acute respiratory acidemia with sodium bicarbonate remains controversial. RECENT FINDINGS There are no randomized controlled trials of administration of sodium bicarbonate for respiratory acidemia. A recent review concluded that alkali therapy for mixed respiratory and metabolic acidosis might be useful but was based on the conflicting and not conclusive literature regarding metabolic acidosis. This strategy should not be extrapolated to treatment of respiratory acidemia. Low tidal volume ventilation in acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) has beneficial effects associated with permissive hypercapnia. Whether the putative benefits will be negated by administration of alkali is not known. Hypercapnic acidosis is well tolerated, with few adverse effects as long as tissue perfusion and oxygenation are maintained. SUMMARY There is a lack of clinical evidence that administration of sodium bicarbonate for respiratory acidosis has a net benefit; in fact, there are potential risks associated with it.
Collapse
Affiliation(s)
- Ranjeeta Chand
- Nephrology Division, New York University Langone Health and Nephrology Section, NY Harbor VA Healthcare System
| | - Erik R Swenson
- Pulmonary, Critical Care and Sleep Medicine Division, University of Washington, and VA Puget Sound Healthcare System, Seattle, Washington, USA
| | - David S Goldfarb
- Nephrology Division, New York University Langone Health and Nephrology Section, NY Harbor VA Healthcare System
| |
Collapse
|
8
|
Abstract
Central nervous system injuries are a leading cause of death and disability worldwide. Although the exact pathophysiological mechanisms of various brain injuries vary, central nervous system injuries often result in an inflammatory response, and subsequently lead to brain damage. This suggests that neuroprotection may be necessany in the treatment of multiple disease models. The use of medical gases as neuroprotective agents has gained great attention in the medical field. Medical gases include common gases, such as oxygen, hydrogen and carbon dioxide; hydrogen sulphide and nitric oxide that have been considered toxic; volatile anesthetic gases, such as isoflurane and sevoflurane; and inert gases like helium, argon, and xenon. The neuroprotection from these medical gases has been investigated in experimental animal models of various types of brain injuries, such as traumatic brain injury, stroke, subarachnoid hemorrhage, cerebral ischemic/reperfusion injury, and neurodegenerative diseases. Nevertheless, the transition into the clinical practice is still lagging. This delay could be attributed to the contradictory paradigms and the conflicting results that have been obtained from experimental models, as well as the presence of inconsistent reports regarding their safety. In this review, we summarize the potential mechanisms underlying the neuroprotective effects of medical gases and discuss possible candidates that could improve the outcomes of brain injury.
Collapse
Affiliation(s)
- Yue-Zhen Wang
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Ting-Ting Li
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Hong-Ling Cao
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wan-Chao Yang
- Department of Anesthesiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
9
|
Pre-Treatment with Ten-Minute Carbon Dioxide Inhalation Prevents Lipopolysaccharide-Induced Lung Injury in Mice via Down-Regulation of Toll-Like Receptor 4 Expression. Int J Mol Sci 2019; 20:ijms20246293. [PMID: 31847115 PMCID: PMC6940754 DOI: 10.3390/ijms20246293] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/07/2019] [Accepted: 12/11/2019] [Indexed: 12/29/2022] Open
Abstract
Various animal studies have shown beneficial effects of hypercapnia in lung injury. However, in patients with acute respiratory distress syndrome (ARDS), there is controversial information regarding the effect of hypercapnia on outcomes. The duration of carbon dioxide inhalation may be the key to the protective effect of hypercapnia. We investigated the effect of pre-treatment with inhaled carbon dioxide on lipopolysaccharide (LPS)-induced lung injury in mice. C57BL/6 mice were randomly divided into a control group or an LPS group. Each LPS group received intratracheal LPS (2 mg/kg); the LPS groups were exposed to hypercapnia (5% carbon dioxide) for 10 min or 60 min before LPS. Bronchoalveolar lavage fluid (BALF) and lung tissues were collected to evaluate the degree of lung injury. LPS significantly increased the ratio of lung weight to body weight; concentrations of BALF protein, tumor necrosis factor-α, and CXCL2; protein carbonyls; neutrophil infiltration; and lung injury score. LPS induced the degradation of the inhibitor of nuclear factor-κB-α (IκB-α) and nuclear translocation of NF-κB. LPS increased the surface protein expression of toll-like receptor 4 (TLR4). Pre-treatment with inhaled carbon dioxide for 10 min, but not for 60 min, inhibited LPS-induced pulmonary edema, inflammation, oxidative stress, lung injury, and TLR4 surface expression, and, accordingly, reduced NF-κB signaling. In summary, our data demonstrated that pre-treatment with 10-min carbon dioxide inhalation can ameliorate LPS-induced lung injury. The protective effect may be associated with down-regulation of the surface expression of TLR4 in the lungs.
Collapse
|
10
|
Zhang C, Dong H, Chen F, Wang Y, Ma J, Wang G. The HMGB1-RAGE/TLR-TNF-α signaling pathway may contribute to kidney injury induced by hypoxia. Exp Ther Med 2018; 17:17-26. [PMID: 30651760 PMCID: PMC6307518 DOI: 10.3892/etm.2018.6932] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 09/06/2018] [Indexed: 01/21/2023] Open
Abstract
The hypoxia-reoxygenation process of obstructive sleep apnea (OSA) may cause oxidative stress injury of the kidney, but the molecular mechanisms are not clear. The present study aimed to investigate whether high mobility group box 1 protein (HMGB1) and its subsequent inflammatory pathway served a role in kidney injury. Adult Sprague Dawley rats were used to establish hypoxia models: Continuous hypoxia, intermittent hypoxia and intermittent hypoxia with hypercapnia. Rat kidney tissues and peripheral blood samples were obtained. Histopathological and ultrastructural changes were observed by light and electron microscopy. Immunohistochemical (IHC) staining was used to detect the distribution of HMGB1. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to detect the expression of HMGB1, receptor for advanced glycosylation end products (RAGE), toll-like receptor 4 (TLR4), nuclear factor kappa-light-chain-enhancer of active B cells (NF-κB) p65 subunit, tumor necrosis factor-α (TNF-α), interleukin (IL)-6, NAD-dependent protein deacetylase sirtuin-1 (SIRT1), peroxisome proliferator-activated receptor (PPAR) mRNA in renal tissues. An ELISA was used to detect the expression of soluble TLR2, TLR4, PPAR-γ, TNF-α, IL-6 in peripheral blood. Hematoxylin & eosin staining demonstrated that there was no serious injury to the kidneys due to hypoxia, with the exception of a certain degree of renal tubular epithelial cell vacuolation. By contrast, ultrastructural changes by electron microscopy were more significant in the hypoxia groups compared with the control, including foot process fusion in the glomerulus and degeneration of mitochondria in the proximal convoluted tubules. IHC also indicated increased expression of HMGB1 and nuclear translocation in the hypoxia groups. The results of the RT-qPCR demonstrated that hypoxia stimulation increased the expression of HMGB1, PPAR, RAGE and TNF-α mRNA, and decreased the expression of SIRT1 mRNA in kidney tissues (P<0.05). The results of the ELISA suggested that hypoxia stimulation increased the expression of soluble TLR4, TNF-α and IL-6 in the peripheral blood, and decreased the expression of soluble TLR2 and PPAR-γ. In summary, hypoxia stimulation may cause early renal injury at the subcellular level and increase the expression and translocation of HMGB1. Hypoxia also upregulated the mRNA expression of the HMGB1-RAGE-TNF-α pathway in kidney tissue and increased the expression of soluble TLR4, TNF-α and IL-6 in the peripheral blood. This suggested that the HMGB1-RAGE/TLR-TNF-α pathway may contribute to the molecular mechanisms of early renal injury induced by hypoxia. The pathway may contain potential markers for OSA-associated early renal injury and drug intervention targets in the future.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Respiratory and Critical Medicine, Peking University First Hospital, Beijing 100034, P.R. China
| | - Hui Dong
- Department of Respiratory and Critical Medicine, Peking University First Hospital, Beijing 100034, P.R. China
| | - Fengwei Chen
- Department of Respiratory and Critical Medicine, Peking University First Hospital, Beijing 100034, P.R. China
| | - Yunxia Wang
- Department of Respiratory and Critical Medicine, Peking University First Hospital, Beijing 100034, P.R. China
| | - Jing Ma
- Department of Respiratory and Critical Medicine, Peking University First Hospital, Beijing 100034, P.R. China
| | - Guangfa Wang
- Department of Respiratory and Critical Medicine, Peking University First Hospital, Beijing 100034, P.R. China
| |
Collapse
|
11
|
Tiruvoipati R, Gupta S, Pilcher D, Bailey M. Hypercapnia and hypercapnic acidosis in sepsis: harmful, beneficial or unclear? CRIT CARE RESUSC 2018; 20:94-100. [PMID: 29852847 DOI: 10.1016/s1441-2772(23)00763-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Mortality related to sepsis among critically ill patients remains high. Recent literature suggests that hypercapnia may affect the pathophysiology of sepsis. The effects of hypercapnia on sepsis are largely related to the direct effect of hypercapnic acidosis on immune function and, as a consequence, of increased cardiac output that subsequently leads to improved tissue oxygenation. Appropriate management of hypercapnia may aid in improving the outcomes of sepsis. Our aim was to review the effects of compensated hypercapnia and hypercapnic acidosis on sepsis, with a specific focus on critically ill patients. Hypercapnic acidosis has been extensively studied in various in vivo animal models of sepsis and ex vivo studies. Published data from animal experimental studies suggest that the effects of hypercapnic acidosis are variable, with benefit shown in some settings of sepsis and harm in others. The effects may also vary at different time points during the course of sepsis. There are very few clinical studies investigating the effects of hypercapnia in prevention of sepsis and in established sepsis. It appears from these very limited clinical data that hypercapnia may be associated with adverse outcomes. There are no clinical studies investigating clinical outcomes of hypercapnic acidosis or compensated hypercapnia in sepsis and septic shock in critical care settings, thus extrapolation of the experimental results to guide critical care practice is difficult. Clinical studies are needed, especially in critically ill patients, to define the effects of compensated hypercapnia and hypercapnic acidosis that may aid clinicians to improve the outcomes in sepsis.
Collapse
Affiliation(s)
| | - Sachin Gupta
- Department of Intensive Care medicine, Frankston Hospital, Frankston, Vic, Australia
| | - David Pilcher
- Australian and New Zealand Intensive Care Research Centre, Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, Vic, Australia
| | - Michael Bailey
- Australian and New Zealand Intensive Care Research Centre, Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne, Vic, Australia
| |
Collapse
|
12
|
Liu J, Wang W, Liu F, Li Z. Pediatric acute respiratory distress syndrome - current views. Exp Ther Med 2018; 15:1775-1780. [PMID: 29434764 PMCID: PMC5776650 DOI: 10.3892/etm.2017.5628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 11/29/2017] [Indexed: 12/18/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) mainly involves acute respiratory failure. In addition to this affected patients feel progressive arterial hypoxemia, dyspnea, and a marked increase in the work of breathing. The only clinical solution for the above pathological state is ventilation. Mechanical ventilation is necessary to support life in ARDs but it itself worsen lung injury and the term is known clinically as ‘ventilation induced lung injury’ (VILI). At the cellular level, respiratory epithelial cells are subjected to cyclic stretch, i.e. repeated cycles of positive and negative strain, during normal tidal ventilation. In aerated areas of diseased lungs, or even normal lungs subjected to injurious positive pressure mechanical ventilation, the cells are at risk of being over distended, and worsening injury by disrupting the alveolar epithelial barrier. Further, hypercapnic acidosis (HCA) in itself confers protection from stretch injury, potentially via a mechanisms involving inhibition of nuclear factor κB (NF-κB), a transcription factor central to inflammation, injury and repair. Mesenchymal stem cells are the latest in the field and are being investigated as a possible therapy for ARDS.
Collapse
Affiliation(s)
- Jinfeng Liu
- Department of Neonatology, Xuzhou Chlidren's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Wei Wang
- Department of Neonatology, Xuzhou Chlidren's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Fengli Liu
- Department of Neonatology, Xuzhou Chlidren's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Zhenguang Li
- Department of Neonatology, Xuzhou Chlidren's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
13
|
Abstract
Sepsis is the main cause of close to 70% of all cases of acute respiratory distress syndromes (ARDS). In addition, sepsis increases susceptibility to ventilator-induced lung injury. Therefore, the development of a ventilatory strategy that can achieve adequate oxygenation without injuring the lungs is highly sought after for patients with acute infection and represents an important therapeutic window to improve patient care. Suboptimal ventilatory settings cannot only harm the lung, but may also contribute to the cascade of organ failure in sepsis due to organ crosstalk.Despite the prominent role of sepsis as a cause for lung injury, most of the studies that addressed mechanical ventilation strategies in ARDS did not specifically assess sepsis-related ARDS patients. Consequently, most of the recommendations regarding mechanical ventilation in sepsis patients are derived from ARDS trials that included multiple clinical diagnoses. While there have been important improvements in general ventilatory management that should apply to all critically ill patients, sepsis-related lung injury might still have particularities that could influence bedside management.After revisiting the interplay between sepsis and ventilation-induced lung injury, this review will reappraise the evidence for the major components of the lung protective ventilation strategy, emphasizing the particularities of sepsis-related acute lung injury.
Collapse
|
14
|
Barnes T, Zochios V, Parhar K. Re-examining Permissive Hypercapnia in ARDS: A Narrative Review. Chest 2017; 154:185-195. [PMID: 29175086 DOI: 10.1016/j.chest.2017.11.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 10/20/2017] [Accepted: 11/13/2017] [Indexed: 12/16/2022] Open
Abstract
Lung-protective ventilation (LPV) has become the cornerstone of management in patients with ARDS. A subset of patients is unable to tolerate LPV without significant CO2 elevation. In these patients, permissive hypercapnia is used. Although thought to be benign, it is becoming increasingly evident that elevated CO2 levels have significant physiological effects. In this narrative review, we highlight clinically relevant end-organ effects in both animal models and clinical studies. We also explore the association between elevated CO2, acute cor pulmonale, and ICU mortality. We conclude with a brief review of alternative therapies for CO2 management currently under investigation in patients with moderate to severe ARDS.
Collapse
Affiliation(s)
- Tavish Barnes
- Department of Critical Care Medicine, University of Calgary, Calgary, AB, Canada
| | - Vasileios Zochios
- Department of Critical Care Medicine, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, College of Medical and Dental Sciences, University of Birmingham, Birmingham, England
| | - Ken Parhar
- Department of Critical Care Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
15
|
Fuller BM, Mohr NM, Drewry AM, Ferguson IT, Trzeciak S, Kollef MH, Roberts BW. Partial pressure of arterial carbon dioxide and survival to hospital discharge among patients requiring acute mechanical ventilation: A cohort study. J Crit Care 2017; 41:29-35. [PMID: 28472700 PMCID: PMC5633513 DOI: 10.1016/j.jcrc.2017.04.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 04/18/2017] [Accepted: 04/21/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE To describe the prevalence of hypocapnia and hypercapnia during the earliest period of mechanical ventilation, and determine the association between PaCO2 and mortality. MATERIALS AND METHODS A cohort study using an emergency department registry of mechanically ventilated patients. PaCO2 was categorized: hypocapnia (<35mmHg), normocapnia (35-45mmHg), and hypercapnia (>45mmHg). The primary outcome was survival to hospital discharge. RESULTS A total of 1,491 patients were included. Hypocapnia occurred in 375 (25%) patients and hypercapnia in 569 (38%). Hypercapnia (85%) had higher survival rate compared to normocapnia (74%) and hypocapnia (66%), P<0.001. PaCO2 was an independent predictor of survival to hospital discharge [hypocapnia (aOR 0.65 (95% confidence interval [CI] 0.48-0.89), normocapnia (reference category), hypercapnia (aOR 1.83 (95% CI 1.32-2.54)]. Over ascending ranges of PaCO2, there was a linear trend of increasing survival up to a PaCO2 range of 66-75mmHg, which had the strongest survival association, aOR 3.18 (95% CI 1.35-7.50). CONCLUSIONS Hypocapnia and hypercapnia occurred frequently after initiation of mechanical ventilation. Higher PaCO2 levels were associated with increased survival. These data provide rationale for a trial examining the optimal PaCO2 in the critically ill.
Collapse
Affiliation(s)
- Brian M Fuller
- Departments of Emergency Medicine and Anesthesiology, Division of Critical Care Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States.
| | - Nicholas M Mohr
- Departments of Emergency Medicine and Anesthesiology, Division of Critical Care Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 200 Hawkins Drive, 1008 RCP, Iowa City, IA 52242, United States.
| | - Anne M Drewry
- Department of Anesthesiology, Division of Critical Care Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States.
| | - Ian T Ferguson
- School of Medicine and Medical Science, University College Dublin, Dublin 4, Ireland.
| | - Stephen Trzeciak
- Departments of Medicine and Emergency Medicine, Division of Critical Care Medicine, Cooper University Hospital, One Cooper Plaza, K152, Camden, NJ 08103, United States.
| | - Marin H Kollef
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, United States.
| | - Brian W Roberts
- Department of Emergency Medicine, Cooper University Hospital, One Cooper Plaza, K152, Camden, NJ 08103, United States.
| |
Collapse
|
16
|
Fuchs H, Rossmann N, Schmid MB, Hoenig M, Thome U, Mayer B, Klotz D, Hummler HD. Permissive hypercapnia for severe acute respiratory distress syndrome in immunocompromised children: A single center experience. PLoS One 2017. [PMID: 28632754 PMCID: PMC5478142 DOI: 10.1371/journal.pone.0179974] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Controlled hypoventilation while accepting hypercapnia has been advocated to reduce ventilator-induced lung injury. The aim of the study was to analyze outcomes of a cohort of immunocompromised children with acute respiratory distress syndrome (ARDS) ventilated with a strategy of stepwise increasing PCO2 targets up to 140 mm Hg. METHODS Retrospective analysis of outcomes of a cohort of children with oncologic disease or after stem cell transplantation and severe respiratory failure in comparison with a historical control cohort. RESULTS Out of 150 episodes of admission to the PICU 88 children underwent invasive mechanical ventilation for >24h (overall survival 75%). In a subgroup of 38 children with high ventilator requirements the PCO2 target ranges were increased stepwise. Fifteen children survived and were discharged from the PICU. Severe pulmonary hypertension was seen in two patients and no case of cerebral edema was observed. Long term outcome was available in 15 patients and 10 of these patients survived without adverse neurological sequelae. With introduction of this strategy survival of immunocompromised children undergoing mechanical ventilation for >24h increased to 48% compared to 32% prior to introduction (historical cohort). CONCLUSIONS A ventilation strategy incorporating very high carbon dioxide levels to allow for low tidal volumes and limited inspiratory pressures is feasible in children. Even severe hypercapnia may be well tolerated. No severe side effects associated with hypercapnia were observed. This strategy could potentially increase survival in immunocompromised children with severe ARDS.
Collapse
Affiliation(s)
- Hans Fuchs
- Center for Pediatrics, Department of Neonatology and Pediatric Intensive Care, Medical Center – Albert Ludwig University of Freiburg, Faculty of Medicine, Freiburg, Germany
- * E-mail:
| | - Nicola Rossmann
- Division of Neonatology and Pediatric Critical Care, Department for Pediatrics and Adolescent Medicine, Ulm University, Ulm, Germany
| | - Manuel B. Schmid
- Department of Neonatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Manfred Hoenig
- Oncology and stem cell transplantation, Department for Pediatrics and Adolescent Medicine, Ulm University, Ulm, Germany
| | - Ulrich Thome
- Division of Neonatology, University Hospital of Leipzig, Leipzig, Germany
| | - Benjamin Mayer
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Daniel Klotz
- Center for Pediatrics, Department of Neonatology and Pediatric Intensive Care, Medical Center – Albert Ludwig University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Helmut D. Hummler
- Division of Neonatology and Pediatric Critical Care, Department for Pediatrics and Adolescent Medicine, Ulm University, Ulm, Germany
| |
Collapse
|
17
|
Otulakowski G, Engelberts D, Arima H, Hirate H, Bayir H, Post M, Kavanagh BP. α-Tocopherol transfer protein mediates protective hypercapnia in murine ventilator-induced lung injury. Thorax 2017; 72:538-549. [PMID: 28159772 DOI: 10.1136/thoraxjnl-2016-209501] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 12/27/2022]
Abstract
RATIONALE Hypercapnia is common in mechanically ventilated patients. Experimentally, 'therapeutic hypercapnia' can protect, but it can also cause harm, depending on the mechanism of injury. Hypercapnia suppresses multiple signalling pathways. Previous investigations have examined mechanisms that were known a priori, but only a limited number of pathways, each suppressed by CO2, have been reported. OBJECTIVE Because of the complexity and interdependence of processes in acute lung injury, this study sought to fill in knowledge gaps using an unbiased screen, aiming to identify a specifically upregulated pathway. METHODS AND RESULTS Using genome-wide gene expression analysis in a mouse model of ventilator-induced lung injury, we discovered a previously unsuspected mechanism by which CO2 can protect against injury: induction of the transporter protein for α-tocopherol, α-tocopherol transfer protein (αTTP). Pulmonary αTTP was induced by inspired CO2 in two in vivo murine models of ventilator-induced lung injury; the level of αTTP expression correlated with degree of lung protection; and, absence of the αTTP gene significantly reduced the protective effects of CO2. α-Tocopherol is a potent antioxidant and hypercapnia increased lung α-tocopherol in wild-type mice, but this did not alter superoxide generation or expression of NRF2-dependent antioxidant response genes in wild-type or in αTTP-/- mice. In concordance with a regulatory role for α-tocopherol in lipid mediator synthesis, hypercapnia attenuated 5-lipoxygenase activity and this was dependent on the presence of αTTP. CONCLUSIONS Inspired CO2 upregulates αTTP which increases lung α-tocopherol levels and inhibits synthesis of a pathogenic chemoattractant.
Collapse
Affiliation(s)
- Gail Otulakowski
- Physiology and Experimental Medicine, Hospital for Sick Children, Toronto, Canada
| | - Doreen Engelberts
- Physiology and Experimental Medicine, Hospital for Sick Children, Toronto, Canada
| | - Hajime Arima
- Physiology and Experimental Medicine, Hospital for Sick Children, Toronto, Canada.,Department of Critical Care Medicine, Hospital for Sick Children, Toronto, Canada.,Department of Anesthesiology and Intensive Care Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroyuki Hirate
- Physiology and Experimental Medicine, Hospital for Sick Children, Toronto, Canada.,Department of Critical Care Medicine, Hospital for Sick Children, Toronto, Canada.,Department of Anesthesiology and Intensive Care Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hülya Bayir
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Critical Care Medicine, Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Martin Post
- Physiology and Experimental Medicine, Hospital for Sick Children, Toronto, Canada
| | - Brian P Kavanagh
- Physiology and Experimental Medicine, Hospital for Sick Children, Toronto, Canada.,Department of Critical Care Medicine, Hospital for Sick Children, Toronto, Canada.,Department of Anesthesia, University of Toronto, Toronto, Canada
| |
Collapse
|
18
|
Abstract
This article reviews aspects of mechanical ventilation in patients without lung injury, patients in the perioperative period, and those with neurologic injury or disease including spinal cord injury. Specific emphasis is placed on ventilator strategies, including timing and indications for tracheostomy. Lung protective ventilation, using low tidal volumes and modest levels of positive end-expiratory pressure, should be the default consideration in all patients requiring mechanical ventilatory support. The exception may be the patient with high cervical spinal cord injuries who requires mechanical ventilatory support. There is no consensus on the timing of tracheostomy in patients with neurologic diseases.
Collapse
|
19
|
Horie S, Ansari B, Masterson C, Devaney J, Scully M, O'Toole D, Laffey JG. Hypercapnic acidosis attenuates pulmonary epithelial stretch-induced injury via inhibition of the canonical NF-κB pathway. Intensive Care Med Exp 2016; 4:8. [PMID: 27001525 PMCID: PMC4801837 DOI: 10.1186/s40635-016-0081-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 03/14/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hypercapnia, with its associated acidosis (HCA), is a consequence of respiratory failure and is also seen in critically ill patients managed with conventional "protective" ventilation strategies. Nuclear factor kappa-B (NF-κB), a pivotal transcription factor, is activated in the setting of injury and repair and is central to innate immunity. We have previously established that HCA protects against ventilation-induced lung injury in vivo, potentially via a mechanism involving inhibition of NF-κB signaling. We wished to further elucidate the role and mechanism of HCA-mediated inhibition of the NF-κB pathway in attenuating stretch-induced injury in vitro. METHODS Initial experiments examined the effect of HCA on cyclic stretch-induced inflammation and injury in human bronchial and alveolar epithelial cells. Subsequent experiments examined the role of the canonical NF-κB pathway in mediating stretch-induced injury and the mechanism of action of HCA. The contribution of pH versus CO2 in mediating this effect of HCA was also examined. RESULTS Pulmonary epithelial high cyclic stretch (22 % equibiaxial strain) activated NF-κB, enhanced interleukin-8 (IL-8) production, caused cell injury, and reduced cell survival. In contrast, physiologic stretch (10 % strain) did not activate inflammation or cause cell injury. HCA reduced cyclic mechanical stretch-induced NF-κB activation, attenuated IL-8 production, reduced injury, and enhanced survival, in bronchial and alveolar epithelial cells, following shorter (24 h) and longer (120 h) cyclic mechanical stretch. Pre-conditioning with HCA was less effective than when HCA was applied after commencement of cell stretch. HCA prevented the stretch-induced breakdown of the NF-κB cytosolic inhibitor IκBα, while IκBα overexpression "occluded" the effect of HCA. These effects were mediated by a pH-dependent mechanism rather than via CO2 per se. CONCLUSIONS HCA attenuates adverse mechanical stretch-induced epithelial injury and death, via a pH-dependent mechanism that inhibits the canonical NF-κB activation by preventing IκBα breakdown.
Collapse
Affiliation(s)
- Shahd Horie
- Discipline of Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - Bilal Ansari
- Discipline of Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - Claire Masterson
- Regenerative Medicine Institute, National University of Ireland, Galway, Ireland.,Department of Anesthesia, Critical Illness and Injury Research Centre, Keenan Research Centre for Biomedical Science, St Michael's Hospital, University of Toronto, Toronto, Canada
| | - James Devaney
- Discipline of Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - Michael Scully
- Discipline of Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - Daniel O'Toole
- Discipline of Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland.,Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - John G Laffey
- Regenerative Medicine Institute, National University of Ireland, Galway, Ireland. .,Department of Anesthesia, Critical Illness and Injury Research Centre, Keenan Research Centre for Biomedical Science, St Michael's Hospital, University of Toronto, Toronto, Canada.
| |
Collapse
|
20
|
Chung KK, Rhie RY, Lundy JB, Cartotto R, Henderson E, Pressman MA, Joe VC, Aden JK, Driscoll IR, Faucher LD, McDermid RC, Mlcak RP, Hickerson WL, Jeng JC. A Survey of Mechanical Ventilator Practices Across Burn Centers in North America. J Burn Care Res 2016; 37:e131-9. [PMID: 26135527 PMCID: PMC5312724 DOI: 10.1097/bcr.0000000000000270] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Burn injury introduces unique clinical challenges that make it difficult to extrapolate mechanical ventilator (MV) practices designed for the management of general critical care patients to the burn population. We hypothesize that no consensus exists among North American burn centers with regard to optimal ventilator practices. The purpose of this study is to examine various MV practice patterns in the burn population and to identify potential opportunities for future research. A researcher designed, 24-item survey was sent electronically to 129 burn centers. The χ, Fisher's exact, and Cochran-Mantel-Haenszel tests were used to determine if there were significant differences in practice patterns. We analyzed 46 questionnaires for a 36% response rate. More than 95% of the burn centers reported greater than 100 annual admissions. Pressure support and volume assist control were the most common initial MV modes used with or without inhalation injury. In the setting of Berlin defined mild acute respiratory distress syndrome (ARDS), ARDSNet protocol and optimal positive end-expiratory pressure were the top ventilator choices, along with fluid restriction/diuresis as a nonventilator adjunct. For severe ARDS, airway pressure release ventilation and neuromuscular blockade were the most popular. The most frequently reported time frame for mechanical ventilation before tracheostomy was 2 weeks (25 of 45, 55%); however, all respondents reported in the affirmative that there are certain clinical situations where early tracheostomy is warranted. Wide variations in clinical practice exist among North American burn centers. No single ventilator mode or adjunct prevails in the management of burn patients regardless of pulmonary insult. Movement toward American Burn Association-supported, multicenter studies to determine best practices and guidelines for ventilator management in burn patients is prudent in light of these findings.
Collapse
Affiliation(s)
- Kevin K. Chung
- From the United States Army Institute of Surgical Research, Fort Sam Houston, Texas; Uniformed Services University of the Health Sciences, Bethesda, Maryland; Sunnybrook Health Sciences Centre, Toronto, Canada; Massachusetts General Hospital, Boston; Arizona Burn Center, Phoenix; University of California Irvine Regional Burn Center, Orange; University of Wisconsin Hospital, Madison; University of Alberta, Edmonton, Canada; Shriners Hospital for Children, Galveston, Texas; Memphis Burn Center, Memphis, Tennessee; and Mount Sinai Beth Israel Medical Center, New York, New York
| | - Ryan Y. Rhie
- From the United States Army Institute of Surgical Research, Fort Sam Houston, Texas; Uniformed Services University of the Health Sciences, Bethesda, Maryland; Sunnybrook Health Sciences Centre, Toronto, Canada; Massachusetts General Hospital, Boston; Arizona Burn Center, Phoenix; University of California Irvine Regional Burn Center, Orange; University of Wisconsin Hospital, Madison; University of Alberta, Edmonton, Canada; Shriners Hospital for Children, Galveston, Texas; Memphis Burn Center, Memphis, Tennessee; and Mount Sinai Beth Israel Medical Center, New York, New York
| | - Jonathan B. Lundy
- From the United States Army Institute of Surgical Research, Fort Sam Houston, Texas; Uniformed Services University of the Health Sciences, Bethesda, Maryland; Sunnybrook Health Sciences Centre, Toronto, Canada; Massachusetts General Hospital, Boston; Arizona Burn Center, Phoenix; University of California Irvine Regional Burn Center, Orange; University of Wisconsin Hospital, Madison; University of Alberta, Edmonton, Canada; Shriners Hospital for Children, Galveston, Texas; Memphis Burn Center, Memphis, Tennessee; and Mount Sinai Beth Israel Medical Center, New York, New York
| | - Robert Cartotto
- From the United States Army Institute of Surgical Research, Fort Sam Houston, Texas; Uniformed Services University of the Health Sciences, Bethesda, Maryland; Sunnybrook Health Sciences Centre, Toronto, Canada; Massachusetts General Hospital, Boston; Arizona Burn Center, Phoenix; University of California Irvine Regional Burn Center, Orange; University of Wisconsin Hospital, Madison; University of Alberta, Edmonton, Canada; Shriners Hospital for Children, Galveston, Texas; Memphis Burn Center, Memphis, Tennessee; and Mount Sinai Beth Israel Medical Center, New York, New York
| | - Elizabeth Henderson
- From the United States Army Institute of Surgical Research, Fort Sam Houston, Texas; Uniformed Services University of the Health Sciences, Bethesda, Maryland; Sunnybrook Health Sciences Centre, Toronto, Canada; Massachusetts General Hospital, Boston; Arizona Burn Center, Phoenix; University of California Irvine Regional Burn Center, Orange; University of Wisconsin Hospital, Madison; University of Alberta, Edmonton, Canada; Shriners Hospital for Children, Galveston, Texas; Memphis Burn Center, Memphis, Tennessee; and Mount Sinai Beth Israel Medical Center, New York, New York
| | - Melissa A. Pressman
- From the United States Army Institute of Surgical Research, Fort Sam Houston, Texas; Uniformed Services University of the Health Sciences, Bethesda, Maryland; Sunnybrook Health Sciences Centre, Toronto, Canada; Massachusetts General Hospital, Boston; Arizona Burn Center, Phoenix; University of California Irvine Regional Burn Center, Orange; University of Wisconsin Hospital, Madison; University of Alberta, Edmonton, Canada; Shriners Hospital for Children, Galveston, Texas; Memphis Burn Center, Memphis, Tennessee; and Mount Sinai Beth Israel Medical Center, New York, New York
| | - Victor C. Joe
- From the United States Army Institute of Surgical Research, Fort Sam Houston, Texas; Uniformed Services University of the Health Sciences, Bethesda, Maryland; Sunnybrook Health Sciences Centre, Toronto, Canada; Massachusetts General Hospital, Boston; Arizona Burn Center, Phoenix; University of California Irvine Regional Burn Center, Orange; University of Wisconsin Hospital, Madison; University of Alberta, Edmonton, Canada; Shriners Hospital for Children, Galveston, Texas; Memphis Burn Center, Memphis, Tennessee; and Mount Sinai Beth Israel Medical Center, New York, New York
| | - James K. Aden
- From the United States Army Institute of Surgical Research, Fort Sam Houston, Texas; Uniformed Services University of the Health Sciences, Bethesda, Maryland; Sunnybrook Health Sciences Centre, Toronto, Canada; Massachusetts General Hospital, Boston; Arizona Burn Center, Phoenix; University of California Irvine Regional Burn Center, Orange; University of Wisconsin Hospital, Madison; University of Alberta, Edmonton, Canada; Shriners Hospital for Children, Galveston, Texas; Memphis Burn Center, Memphis, Tennessee; and Mount Sinai Beth Israel Medical Center, New York, New York
| | - Ian R. Driscoll
- From the United States Army Institute of Surgical Research, Fort Sam Houston, Texas; Uniformed Services University of the Health Sciences, Bethesda, Maryland; Sunnybrook Health Sciences Centre, Toronto, Canada; Massachusetts General Hospital, Boston; Arizona Burn Center, Phoenix; University of California Irvine Regional Burn Center, Orange; University of Wisconsin Hospital, Madison; University of Alberta, Edmonton, Canada; Shriners Hospital for Children, Galveston, Texas; Memphis Burn Center, Memphis, Tennessee; and Mount Sinai Beth Israel Medical Center, New York, New York
| | - Lee D. Faucher
- From the United States Army Institute of Surgical Research, Fort Sam Houston, Texas; Uniformed Services University of the Health Sciences, Bethesda, Maryland; Sunnybrook Health Sciences Centre, Toronto, Canada; Massachusetts General Hospital, Boston; Arizona Burn Center, Phoenix; University of California Irvine Regional Burn Center, Orange; University of Wisconsin Hospital, Madison; University of Alberta, Edmonton, Canada; Shriners Hospital for Children, Galveston, Texas; Memphis Burn Center, Memphis, Tennessee; and Mount Sinai Beth Israel Medical Center, New York, New York
| | - Robert C. McDermid
- From the United States Army Institute of Surgical Research, Fort Sam Houston, Texas; Uniformed Services University of the Health Sciences, Bethesda, Maryland; Sunnybrook Health Sciences Centre, Toronto, Canada; Massachusetts General Hospital, Boston; Arizona Burn Center, Phoenix; University of California Irvine Regional Burn Center, Orange; University of Wisconsin Hospital, Madison; University of Alberta, Edmonton, Canada; Shriners Hospital for Children, Galveston, Texas; Memphis Burn Center, Memphis, Tennessee; and Mount Sinai Beth Israel Medical Center, New York, New York
| | - Ronald P. Mlcak
- From the United States Army Institute of Surgical Research, Fort Sam Houston, Texas; Uniformed Services University of the Health Sciences, Bethesda, Maryland; Sunnybrook Health Sciences Centre, Toronto, Canada; Massachusetts General Hospital, Boston; Arizona Burn Center, Phoenix; University of California Irvine Regional Burn Center, Orange; University of Wisconsin Hospital, Madison; University of Alberta, Edmonton, Canada; Shriners Hospital for Children, Galveston, Texas; Memphis Burn Center, Memphis, Tennessee; and Mount Sinai Beth Israel Medical Center, New York, New York
| | - William L. Hickerson
- From the United States Army Institute of Surgical Research, Fort Sam Houston, Texas; Uniformed Services University of the Health Sciences, Bethesda, Maryland; Sunnybrook Health Sciences Centre, Toronto, Canada; Massachusetts General Hospital, Boston; Arizona Burn Center, Phoenix; University of California Irvine Regional Burn Center, Orange; University of Wisconsin Hospital, Madison; University of Alberta, Edmonton, Canada; Shriners Hospital for Children, Galveston, Texas; Memphis Burn Center, Memphis, Tennessee; and Mount Sinai Beth Israel Medical Center, New York, New York
| | - James C. Jeng
- From the United States Army Institute of Surgical Research, Fort Sam Houston, Texas; Uniformed Services University of the Health Sciences, Bethesda, Maryland; Sunnybrook Health Sciences Centre, Toronto, Canada; Massachusetts General Hospital, Boston; Arizona Burn Center, Phoenix; University of California Irvine Regional Burn Center, Orange; University of Wisconsin Hospital, Madison; University of Alberta, Edmonton, Canada; Shriners Hospital for Children, Galveston, Texas; Memphis Burn Center, Memphis, Tennessee; and Mount Sinai Beth Israel Medical Center, New York, New York
| |
Collapse
|
21
|
Yang W, Zhang X, Wang N, Tan J, Fang X, Wang Q, Tao T, Li W. Effects of Acute Systemic Hypoxia and Hypercapnia on Brain Damage in a Rat Model of Hypoxia-Ischemia. PLoS One 2016; 11:e0167359. [PMID: 27907083 PMCID: PMC5131999 DOI: 10.1371/journal.pone.0167359] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 11/12/2016] [Indexed: 12/18/2022] Open
Abstract
Therapeutic hypercapnia has the potential for neuroprotection after global cerebral ischemia. Here we further investigated the effects of different degrees of acute systemic hypoxia in combination with hypercapnia on brain damage in a rat model of hypoxia and ischemia. Adult wistar rats underwent unilateral common carotid artery (CCA) ligation for 60 min followed by ventilation with normoxic or systemic hypoxic gas containing 11%O2,13%O2,15%O2 and 18%O2 (targeted to PaO2 30-39 mmHg, 40-49 mmHg, 50-59 mmHg, and 60-69 mmHg, respectively) or systemic hypoxic gas containing 8% carbon dioxide (targeted to PaCO2 60-80 mmHg) for 180 min. The mean artery pressure (MAP), blood gas, and cerebral blood flow (CBF) were evaluated. The cortical vascular permeability and brain edema were examined. The ipsilateral cortex damage and the percentage of hippocampal apoptotic neurons were evaluated by Nissl staining and terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine 5'-triphosphate-biotin nick end labeling (TUNEL) assay as well as flow cytometry, respectively. Immunofluorescence and western blotting were performed to determine aquaporin-4 (AQP4) expression. In rats treated with severe hypoxia (PaO2 < 50 mmHg), hypercapnia augmented the decline of MAP with cortical CBF and damaged blood-brain barrier permeability (p < 0.05). In contrast, in rats treated with mild to moderate hypoxia (PaO2 > 50 mmHg), hypercapnia protected against these pathophysiological changes. Moreover, hypercapnia treatment significantly reduced brain damage in the ischemic ipsilateral cortex and decreased the percentage of apoptotic neurons in the hippocampus after the CCA ligated rats were exposed to mild or moderate hypoxemia (PaO2 > 50 mmHg); especially under mild hypoxemia (PaO2 > 60 mmHg), hypercapnia significantly attenuated the expression of AQP4 protein with brain edema (p < 0.05). Hypercapnia exerts beneficial effects under mild to moderate hypoxemia and augments detrimental effects under severe hypoxemia on brain damage in a rat model of hypoxia-ischemia.
Collapse
Affiliation(s)
- Wanchao Yang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University P. R. China; and Anesthesiology Key Laboratory, Education Department, Harbin Medical University, Heilongjiang Province, P. R. China
| | - Xuezhong Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University P. R. China; and Anesthesiology Key Laboratory, Education Department, Harbin Medical University, Heilongjiang Province, P. R. China
| | - Nan Wang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University P. R. China; and Anesthesiology Key Laboratory, Education Department, Harbin Medical University, Heilongjiang Province, P. R. China
| | - Jing Tan
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University P. R. China; and Anesthesiology Key Laboratory, Education Department, Harbin Medical University, Heilongjiang Province, P. R. China
| | - Xianhai Fang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University P. R. China; and Anesthesiology Key Laboratory, Education Department, Harbin Medical University, Heilongjiang Province, P. R. China
| | - Qi Wang
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University P. R. China; and Anesthesiology Key Laboratory, Education Department, Harbin Medical University, Heilongjiang Province, P. R. China
| | - Tao Tao
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University P. R. China; and Anesthesiology Key Laboratory, Education Department, Harbin Medical University, Heilongjiang Province, P. R. China
| | - Wenzhi Li
- Department of Anesthesiology, Second Affiliated Hospital of Harbin Medical University P. R. China; and Anesthesiology Key Laboratory, Education Department, Harbin Medical University, Heilongjiang Province, P. R. China
- * E-mail:
| |
Collapse
|
22
|
Retamal J, Borges JB, Bruhn A, Cao X, Feinstein R, Hedenstierna G, Johansson S, Suarez-Sipmann F, Larsson A. High respiratory rate is associated with early reduction of lung edema clearance in an experimental model of ARDS. Acta Anaesthesiol Scand 2016; 60:79-92. [PMID: 26256848 DOI: 10.1111/aas.12596] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 07/08/2015] [Accepted: 07/13/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND The independent impact of respiratory rate on ventilator-induced lung injury has not been fully elucidated. The aim of this study was to investigate the effects of two clinically relevant respiratory rates on early ventilator-induced lung injury evolution and lung edema during the protective ARDSNet strategy. We hypothesized that the use of a higher respiratory rate during a protective ARDSNet ventilation strategy increases lung inflammation and, in addition, lung edema associated to strain-induced activation of transforming growth factor beta (TGF-β) in the lung epithelium. METHODS Twelve healthy piglets were submitted to a two-hit lung injury model and randomized into two groups: LRR (20 breaths/min) and HRR (40 breaths/min). They were mechanically ventilated during 6 h according to the ARDSNet strategy. We assessed respiratory mechanics, hemodynamics, and extravascular lung water (EVLW). At the end of the experiment, the lungs were excised and wet/dry ratio, TGF-β pathway markers, regional histology, and cytokines were evaluated. RESULTS No differences in oxygenation, PaCO2 levels, systemic and pulmonary arterial pressures were observed during the study. Respiratory system compliance and mean airway pressure were lower in LRR group. A decrease in EVLW over time occurred only in the LRR group (P < 0.05). Wet/dry ratio was higher in the HRR group (P < 0.05), as well as TGF-β pathway activation. Histological findings suggestive of inflammation and inflammatory tissue cytokines were higher in LRR. CONCLUSION HRR was associated with more pulmonary edema and higher activation of the TGF-β pathway. In contrast with our hypothesis, HRR was associated with less lung inflammation.
Collapse
Affiliation(s)
- J. Retamal
- Hedenstierna Laboratory; Department of Surgical Sciences; Section of Anaesthesiology & Critical Care; Uppsala University; Uppsala Sweden
- Departamento de Medicina Intensiva; Pontificia Universidad Cat ó lica de Chile; Santiago Chile
| | - J. B. Borges
- Hedenstierna Laboratory; Department of Surgical Sciences; Section of Anaesthesiology & Critical Care; Uppsala University; Uppsala Sweden
- Cardio-Pulmonary Department; Pulmonary Divison; Heart Institute (Incor); University of São Paulo; São Paulo Brazil
| | - A. Bruhn
- Departamento de Medicina Intensiva; Pontificia Universidad Cat ó lica de Chile; Santiago Chile
| | - X. Cao
- Department of Medical Biochemistry and Microbiology; Uppsala University; Uppsala Sweden
| | - R. Feinstein
- Department of Pathology and Wildlife Diseases; National Veterinary Institute; Uppsala Sweden
| | - G. Hedenstierna
- Department of Medical Science, Clinical Physiology; Uppsala University Hospital; Uppsala Sweden
| | - S. Johansson
- Department of Medical Biochemistry and Microbiology; Uppsala University; Uppsala Sweden
| | - F. Suarez-Sipmann
- Hedenstierna Laboratory; Department of Surgical Sciences; Section of Anaesthesiology & Critical Care; Uppsala University; Uppsala Sweden
| | - A. Larsson
- Hedenstierna Laboratory; Department of Surgical Sciences; Section of Anaesthesiology & Critical Care; Uppsala University; Uppsala Sweden
| |
Collapse
|
23
|
Hayes M, Curley GF, Masterson C, Devaney J, O'Toole D, Laffey JG. Mesenchymal stromal cells are more effective than the MSC secretome in diminishing injury and enhancing recovery following ventilator-induced lung injury. Intensive Care Med Exp 2015; 3:29. [PMID: 26472334 PMCID: PMC4607685 DOI: 10.1186/s40635-015-0065-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 10/14/2014] [Indexed: 12/19/2022] Open
Abstract
Background The potential for mesenchymal stem cells (MSCs) to reduce the severity of experimental lung injury has been established in several pre-clinical studies. We have recently demonstrated that MSCs, and MSC-secreted factors (secretome), enhance lung repair and regeneration at 48 h following ventilation-induced lung injury (VILI). We wished to determine the potential for MSC therapy to exert beneficial effects in the early recovery phase following VILI when ongoing injury coexists with processes of repair, and to compare the efficacy of MSC therapy to the use of the secretome alone. Methods Male Sprague–Dawley rats were anesthetized, oro-tracheally intubated, and subjected to high stretch mechanical ventilation until lung compliance had declined by 50 % of baseline. Animals were then weaned from mechanical ventilation, and anesthesia discontinued. Once awake and spontaneously ventilating, animals received an intravenous injection of either rodent MSCs (10 million/kg), MSC-conditioned medium, fibroblasts (10 million/kg), or vehicle. Thereafter, the animals were allowed to recover and the extent of lung injury/repair was determined after 4 h. Results Treatment with MSCs diminished injury and enhanced recovery following VILI to a greater extent compared to MSC-conditioned medium, with fibroblasts proving ineffective. MSCs, but not their conditioned medium, attenuated indices of lung injury including oxygenation, respiratory compliance, and lung edema. Total lung water as assessed by wet:dry ratio, bronchoalveolar lavage total inflammatory cell, neutrophil counts, and alveolar IL-6 concentrations were reduced in the animals that received MSC therapy. Conclusions The immunomodulating and/or reparative effect of MSCs is evident early after VILI in this model. MSC-conditioned medium was not as effective as the cells themselves in diminishing injury and restoring lung structure and function.
Collapse
Affiliation(s)
- Mairead Hayes
- Regenerative Medicine Institute, National University of Ireland, Galway, Ireland. .,Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland.
| | - Gerard F Curley
- Department of Anesthesia, Keenan Research Centre for Biomedical Science of St Michael's Hospital, St. Michael's Hospital, 30 Bond Street, Toronto, ON, M5B 1W8, Canada. .,Department of Anesthesia, University of Toronto, Toronto, Canada.
| | - Claire Masterson
- Department of Anesthesia, Keenan Research Centre for Biomedical Science of St Michael's Hospital, St. Michael's Hospital, 30 Bond Street, Toronto, ON, M5B 1W8, Canada. .,Department of Anesthesia, University of Toronto, Toronto, Canada.
| | - James Devaney
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland.
| | - Daniel O'Toole
- Regenerative Medicine Institute, National University of Ireland, Galway, Ireland. .,Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland.
| | - John G Laffey
- Regenerative Medicine Institute, National University of Ireland, Galway, Ireland. .,Department of Anesthesia, Keenan Research Centre for Biomedical Science of St Michael's Hospital, St. Michael's Hospital, 30 Bond Street, Toronto, ON, M5B 1W8, Canada. .,Department of Anesthesia, University of Toronto, Toronto, Canada.
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Multiple clinical and laboratory studies have been conducted to illustrate the effects of hypercapnia in a range of injuries, and to understand the mechanisms underlying these effects. The aim of this review is to highlight and interpret information obtained from these recent reports and discuss how they may inform the clinical context. RECENT FINDINGS In the last decade, several important articles have addressed key elements of how carbon dioxide interacts in critical illness states. Among them the most important insights relate to how hypercapnia affects critical illness and include the effects and mechanisms of carbon dioxide in pulmonary hypertension, infection, inflammation, diaphragm dysfunction, and cerebral ischemia. In addition, we discuss molecular insights that apply to multiple aspects of critical illness. SUMMARY Experiments involving hypercapnia have covered a wide range of illness models with varying degrees of success. It is becoming evident that deliberate hypercapnia in the clinical setting should seldom be used, except wherever necessitated to avoid ventilator-associated lung injury. A more complete understanding of the molecular mechanisms must be established.
Collapse
|
25
|
Wang C, Gui Q, Zhang K. Functional polymorphisms in CD86 gene are associated with susceptibility to pneumonia-induced sepsis. APMIS 2015; 123:433-8. [PMID: 25912130 DOI: 10.1111/apm.12364] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 12/17/2014] [Indexed: 11/30/2022]
Abstract
Sepsis is an illness in which the body has a severe response to bacteria or other germs. A bacterial infection in the body such as lungs may set off the response that leads to the disease. CD86 (B7-2) is expressed on various immune cells and plays critical roles in immune responses. Genetic polymorphisms in CD86 gene may affect the development of several diseases. Here, we evaluated the association between two CD86 polymorphisms (rs1915087C/T and rs2332096T/G) and susceptibility to pneumonia-induced sepsis. CD86 rs1915087C/T and rs2332096T/G were identified in 186 pneumonia-induced septic patients and 196 healthy controls in the Chinese population. Results revealed that subjects with rs1915087CT and TT genotypes had significantly lower risk of pneumonia-induced sepsis than those with CC genotype [odds ratio (OR) = 0.58, 95% confidence interval (CI), 0.37-0.91, p = 0.017, and OR = 0.40, 95%CI, 0.21-0.76, p = 0.005]. However, prevalence of rs2332096GG genotype and G allele were significantly increased in patients than in healthy controls (OR = 2.75, 95%CI, 1.46-5.16, p = 0.001, and OR = 1.65, 95%CI, 1.21-2.24, p = 0.001]. We further investigated functions of these two polymorphisms by assessing gene expression in peripheral blood mononuclear cells and in monocytes. Data showed subjects carrying rs2332096GG genotype had significantly decreased level of CD86 in monocytes than those carrying rs2332096TT genotype. These results indicate that CD86 polymorphisms are associated with susceptibility to pneumonia-induced sepsis and may affect gene expression in monocytes.
Collapse
Affiliation(s)
- Chenfei Wang
- Department of Emergency, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | | | |
Collapse
|
26
|
|
27
|
Nardelli L, Rzezinski A, Silva J, Maron-Gutierrez T, Ornellas D, Henriques I, Capelozzi V, Teodoro W, Morales M, Silva P, Pelosi P, Garcia C, Rocco P. Effects of acute hypercapnia with and without acidosis on lung inflammation and apoptosis in experimental acute lung injury. Respir Physiol Neurobiol 2015; 205:1-6. [DOI: 10.1016/j.resp.2014.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/04/2014] [Accepted: 09/14/2014] [Indexed: 12/24/2022]
|
28
|
Curley GF, Laffey JG, Kavanagh BP. CrossTalk proposal: there is added benefit to providing permissive hypercapnia in the treatment of ARDS. J Physiol 2013; 591:2763-5. [PMID: 23729790 DOI: 10.1113/jphysiol.2013.252601] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Gerard F Curley
- Department of Anesthesia, Keenan Research Centre in the Li Ka Shing Knowledge Institute, St Michael’s Hospital, Toronto, Ontario, Canada
| | | | | |
Collapse
|
29
|
Gates KL, Howell HA, Nair A, Vohwinkel CU, Welch LC, Beitel GJ, Hauser AR, Sznajder JI, Sporn PHS. Hypercapnia impairs lung neutrophil function and increases mortality in murine pseudomonas pneumonia. Am J Respir Cell Mol Biol 2013; 49:821-8. [PMID: 23777386 DOI: 10.1165/rcmb.2012-0487oc] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Hypercapnia, an elevation of the level of carbon dioxide (CO2) in blood and tissues, is a marker of poor prognosis in chronic obstructive pulmonary disease and other pulmonary disorders. We previously reported that hypercapnia inhibits the expression of TNF and IL-6 and phagocytosis in macrophages in vitro. In the present study, we determined the effects of normoxic hypercapnia (10% CO2, 21% O2, and 69% N2) on outcomes of Pseudomonas aeruginosa pneumonia in BALB/c mice and on pulmonary neutrophil function. We found that the mortality of P. aeruginosa pneumonia was increased in 10% CO2-exposed compared with air-exposed mice. Hypercapnia increased pneumonia mortality similarly in mice with acute and chronic respiratory acidosis, indicating an effect unrelated to the degree of acidosis. Exposure to 10% CO2 increased the burden of P. aeruginosa in the lungs, spleen, and liver, but did not alter lung injury attributable to pneumonia. Hypercapnia did not reduce pulmonary neutrophil recruitment during infection, but alveolar neutrophils from 10% CO2-exposed mice phagocytosed fewer bacteria and produced less H2O2 than neutrophils from air-exposed mice. Secretion of IL-6 and TNF in the lungs of 10% CO2-exposed mice was decreased 7 hours, but not 15 hours, after the onset of pneumonia, indicating that hypercapnia inhibited the early cytokine response to infection. The increase in pneumonia mortality caused by elevated CO2 was reversible when hypercapnic mice were returned to breathing air before or immediately after infection. These results suggest that hypercapnia may increase the susceptibility to and/or worsen the outcome of lung infections in patients with severe lung disease.
Collapse
Affiliation(s)
- Khalilah L Gates
- 1 Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Bautista AF, Akca O. Hypercapnia: is it protective in lung injury? Med Gas Res 2013; 3:23. [PMID: 24209944 PMCID: PMC3833649 DOI: 10.1186/2045-9912-3-23] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 11/05/2013] [Indexed: 11/10/2022] Open
Abstract
Hypercapnic acidosis has been regarded as a tolerated side effect of protective lung ventilation strategies. Various in vivo and ex vivo animal studies have shown beneficial effects in acute lung injury setting, but some recent work raised concerns about its anti-inflammatory properties. This mini-review article aims to expand the potential clinical spectrum of hypercapnic acidosis in critically ill patients with lung injury. Despite the proven benefits of hypercapnic acidosis, further safety studies including dose-effect, level-and-onset of anti-inflammatory effect, and safe applicability period need to be performed in various models of lung injury in animals and humans to further elucidate its protective role.
Collapse
Affiliation(s)
| | - Ozan Akca
- Department of Anesthesiology & Perioperative Medicine, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|
31
|
Akça O, Kurz A, Fleischmann E, Buggy D, Herbst F, Stocchi L, Galandiuk S, Iscoe S, Fisher J, Apfel C, Sessler D. Hypercapnia and surgical site infection: a randomized trial †. Br J Anaesth 2013; 111:759-67. [DOI: 10.1093/bja/aet233] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
32
|
Cummins EP, Selfridge AC, Sporn PH, Sznajder JI, Taylor CT. Carbon dioxide-sensing in organisms and its implications for human disease. Cell Mol Life Sci 2013; 71:831-45. [PMID: 24045706 DOI: 10.1007/s00018-013-1470-6] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 08/22/2013] [Accepted: 08/30/2013] [Indexed: 12/29/2022]
Abstract
The capacity of organisms to sense changes in the levels of internal and external gases and to respond accordingly is central to a range of physiologic and pathophysiologic processes. Carbon dioxide, a primary product of oxidative metabolism is one such gas that can be sensed by both prokaryotic and eukaryotic cells and in response to altered levels, elicit the activation of multiple adaptive pathways. The outcomes of activating CO2-sensitive pathways in various species include increased virulence of fungal and bacterial pathogens, prey-seeking behavior in insects as well as taste perception, lung function, and the control of immunity in mammals. In this review, we discuss what is known about the mechanisms underpinning CO2 sensing across a range of species and consider the implications of this for physiology, disease progression, and the possibility of developing new therapeutics for inflammatory and infectious disease.
Collapse
Affiliation(s)
- Eoin P Cummins
- School of Medicine and Medical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | | | |
Collapse
|
33
|
Tao T, Liu Y, Zhang J, Xu Y, Li W, Zhao M. Therapeutic hypercapnia improves functional recovery and attenuates injury via antiapoptotic mechanisms in a rat focal cerebral ischemia/reperfusion model. Brain Res 2013; 1533:52-62. [PMID: 23939225 DOI: 10.1016/j.brainres.2013.08.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 08/01/2013] [Accepted: 08/05/2013] [Indexed: 12/13/2022]
Abstract
Recent studies have demonstrated neuroprotective effects of therapeutic hypercapnia for different forms of brain injury. However, few studies have assessed the neuroprotective and neurobehavioral effects of hypercapnia in focal cerebral ischemia, and the underlying mechanisms are still unclear. Here, we investigated the effects of therapeutic hypercapnia in focal cerebral ischemia in the rat middle cerebral artery occlusion/reperfusion (MCAO/R) model. Adult male Sprague Dawley rats were subjected to 90 min of MCAO/R and subsequently exposed to increased carbon dioxide (CO2) levels to maintain arterial blood CO2 tension (PaCO2) between 80 and 100 mmHg for 2h. Neurological deficits were evaluated with the corner test at days 1, 7, 14, and 28. Infarction volume and apoptotic changes were assessed by 2, 3, 7-triphenyltetrazolium chloride (TTC) staining, and terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine 5'-triphosphate-biotin nick end labeling (TUNEL) staining at 24h after reperfusion. Apoptosis-related proteins (Bcl-2, Bax, cytochrome c, and caspase-3) were investigated by western blotting. The results of this study showed that therapeutic hypercapnia significantly reduced infarct volume and improved neurological scores after MCAO/R. Moreover, hypercapnia treatment increased the survival rate at 28 days after reperfusion. The TUNEL-positive neurons in the ipsilateral cortex were significantly decreased in the hypercapnia group. Mitochondrial Bcl-2 and Bax cortical expression levels were significantly higher and lower, respectively, in hypercapnia-treated rats. In addition, hypercapnia treatment decreased cytosolic cytochrome c and cleaved caspase-3 expression and increased cytosolic Bax expression. These findings indicate that therapeutic hypercapnia preserves brain tissue and promotes functional neurological recovery through antiapoptotic mechanisms.
Collapse
Affiliation(s)
- Tao Tao
- Department of Anesthesiology, the Second Affiliated Hospital of Harbin Medical University, the Hei Long Jiang Province key Lab of Research on Anesthesiology and Critical Care Medicine, Harbin 150081, China.
| | | | | | | | | | | |
Collapse
|
34
|
Devaney J, Curley GF, Hayes M, Masterson C, Ansari B, O'Brien T, O'Toole D, Laffey JG. Inhibition of pulmonary nuclear factor kappa-B decreases the severity of acute Escherichia coli pneumonia but worsens prolonged pneumonia. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2013; 17:R82. [PMID: 23622108 PMCID: PMC4056114 DOI: 10.1186/cc12696] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 04/27/2013] [Indexed: 12/19/2022]
Abstract
Introduction Nuclear factor (NF)-κB is central to the pathogenesis of inflammation in acute lung injury, but also to inflammation resolution and repair. We wished to determine whether overexpression of the NF-κB inhibitor IκBα could modulate the severity of acute and prolonged pneumonia-induced lung injury in a series of prospective randomized animal studies. Methods Adult male Sprague-Dawley rats were randomized to undergo intratracheal instillation of (a) 5 × 109 adenoassociated virus (AAV) vectors encoding the IκBα transgene (5 × 109 AAV-IκBα); (b) 1 × 1010 AAV-IκBα; (c) 5 × 1010 AAV-IκBα; or (d) vehicle alone. After intratracheal inoculation with Escherichia coli, the severity of the lung injury was measured in one series over a 4-hour period (acute pneumonia), and in a second series after 72 hours (prolonged pneumonia). Additional experiments examined the effects of IκBα and null-gene overexpression on E. coli-induced and sham pneumonia. Results In acute pneumonia, IκBα dose-dependently decreased lung injury, improving arterial oxygenation and lung static compliance, reducing alveolar protein leak and histologic injury, and decreasing alveolar IL-1β concentrations. Benefit was maximal at the intermediate (1 × 1010) IκBα vector dose; however, efficacy was diminished at the higher (5 × 1010) IκBα vector dose. In contrast, IκBα worsened prolonged pneumonia-induced lung injury, increased lung bacterial load, decreased lung compliance, and delayed resolution of the acute inflammatory response. Conclusions Inhibition of pulmonary NF-κB activity reduces early pneumonia-induced injury, but worsens injury and bacterial load during prolonged pneumonia.
Collapse
|
35
|
Abstract
In the previous issue of Critical Care, Jung and colleagues report on the preventive effects of hypercapnia on ventilator-induced diaphragmatic dysfunction (VIDD) under controlled ventilation. Possibly, a combination of controlled hypercapnia and allowed spontaneous breathing efforts may provide complementary protection for diaphragm and respiratory functionality during mechanical ventilation. However, further safety and efficacy studies need to be performed in various different animal models and patients before a universal application of hypercapnia in the critical care setting for the prevention of VIDD can be considered.
Collapse
|
36
|
Hypercapnic acidosis attenuates ventilation-induced lung injury by a nuclear factor-κB–dependent mechanism. Crit Care Med 2012; 40:2622-30. [DOI: 10.1097/ccm.0b013e318258f8b4] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
37
|
Fuchs H, Mendler MR, Scharnbeck D, Ebsen M, Hummler HD. Very low tidal volume ventilation with associated hypercapnia--effects on lung injury in a model for acute respiratory distress syndrome. PLoS One 2011; 6:e23816. [PMID: 21886825 PMCID: PMC3158784 DOI: 10.1371/journal.pone.0023816] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 07/27/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Ventilation using low tidal volumes with permission of hypercapnia is recommended to protect the lung in acute respiratory distress syndrome. However, the most lung protective tidal volume in association with hypercapnia is unknown. The aim of this study was to assess the effects of different tidal volumes with associated hypercapnia on lung injury and gas exchange in a model for acute respiratory distress syndrome. METHODOLOGY/PRINCIPAL FINDINGS In this randomized controlled experiment sixty-four surfactant-depleted rabbits were exposed to 6 hours of mechanical ventilation with the following targets: Group 1: tidal volume = 8-10 ml/kg/PaCO(2) = 40 mm Hg; Group 2: tidal volume = 4-5 ml/kg/PaCO(2) = 80 mm Hg; Group 3: tidal volume = 3-4 ml/kg/PaCO(2) = 120 mm Hg; Group 4: tidal volume = 2-3 ml/kg/PaCO(2) = 160 mm Hg. Decreased wet-dry weight ratios of the lungs, lower histological lung injury scores and higher PaO(2) were found in all low tidal volume/hypercapnia groups (group 2, 3, 4) as compared to the group with conventional tidal volume/normocapnia (group 1). The reduction of the tidal volume below 4-5 ml/kg did not enhance lung protection. However, oxygenation and lung protection were maintained at extremely low tidal volumes in association with very severe hypercapnia and no adverse hemodynamic effects were observed with this strategy. CONCLUSION Ventilation with low tidal volumes and associated hypercapnia was lung protective. A tidal volume below 4-5 ml/kg/PaCO(2) 80 mm Hg with concomitant more severe hypercapnic acidosis did not increase lung protection in this surfactant deficiency model. However, even at extremely low tidal volumes in association with severe hypercapnia lung protection and oxygenation were maintained.
Collapse
Affiliation(s)
- Hans Fuchs
- Division of Neonatology and Pediatric Critical Care, Department of Pediatrics and Adolescent Medicine, Ulm University, Ulm, Germany.
| | | | | | | | | |
Collapse
|
38
|
Chuang IC, Yang RC, Chou SH, Huang LR, Tsai TN, Dong HP, Huang MS. Effect of carbon dioxide inhalation on pulmonary hypertension induced by increased blood flow and hypoxia. Kaohsiung J Med Sci 2011; 27:336-43. [DOI: 10.1016/j.kjms.2011.03.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Accepted: 11/10/2010] [Indexed: 11/16/2022] Open
|
39
|
Hassett P, Curley GF, Contreras M, Masterson C, Higgins BD, O'Brien T, Devaney J, O'Toole D, Laffey JG. Overexpression of pulmonary extracellular superoxide dismutase attenuates endotoxin-induced acute lung injury. Intensive Care Med 2011; 37:1680-7. [PMID: 21755396 PMCID: PMC7095197 DOI: 10.1007/s00134-011-2309-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 06/23/2011] [Indexed: 11/02/2022]
Abstract
PURPOSE Superoxide is produced by activated neutrophils during the inflammatory response to stimuli such as endotoxin, can directly or indirectly injure host cells, and has been implicated in the pathogenesis of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS). We wished to determine the potential for pulmonary overexpression of the extracellular isoform of superoxide dismutase (EC-SOD) to reduce the severity of endotoxin-induced lung injury. METHODS Animals were randomly allocated to undergo intratracheal instillation of (1) surfactant alone (vehicle); (2) adeno-associated virus (AAV) vectors containing a null transgene (AAV-null); and (3) adeno-associated virus vectors containing the EC-SOD transgene (AAV-EC-SOD) and endotoxin was subsequently administered intratracheally. Two additional groups were randomized to receive (1) vehicle or (2) AAV-EC-SOD, and to undergo sham (vehicle) injury. The severity of the lung injury was assessed in all animals 24 h later. RESULTS Endotoxin produced a severe lung injury compared to sham injury. The AAV vector encoding EC-SOD increased lung EC-SOD concentrations, and enhanced the antioxidant capacity of the lung. EC-SOD overexpression decreased the severity of endotoxin-induced ALI, reducing the decrement in systemic oxygenation and lung compliance, decreasing lung permeability and decreasing histologic injury. EC-SOD attenuated pulmonary inflammation, decreased bronchoalveolar lavage neutrophil counts, and reduced interleukin-6 and CINC-1 concentrations. The AAV vector itself did not contribute to inflammation or to lung injury. CONCLUSIONS Pulmonary overexpression of EC-SOD protects the lung against endotoxin-induced ALI.
Collapse
Affiliation(s)
- Patrick Hassett
- Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Curley G, Hayes M, Laffey JG. Can 'permissive' hypercapnia modulate the severity of sepsis-induced ALI/ARDS? CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2011; 15:212. [PMID: 21457509 PMCID: PMC3219408 DOI: 10.1186/cc9994] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Gerard Curley
- Department of Anestheisa, Clinical Sciences Institute, National University, Galway, Ireland
| | | | | |
Collapse
|
41
|
Bench-to-bedside review: hypercapnic acidosis in lung injury--from 'permissive' to 'therapeutic'. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2010; 14:237. [PMID: 21067531 PMCID: PMC3220022 DOI: 10.1186/cc9238] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Modern ventilation strategies for patients with acute lung injury and acute respiratory distress syndrome frequently result in hypercapnic acidosis (HCA), which is regarded as an acceptable side effect ('permissive hypercapnia'). Multiple experimental studies have demonstrated advantageous effects of HCA in several lung injury models. To date, however, human trials studying the effect of carbon dioxide per se on outcome in patients with lung injury have not been performed. While significant concerns regarding HCA remain, in particular the possible unfavorable effects on bacterial killing and the inhibition of pulmonary epithelial wound repair, the potential for HCA in attenuating lung injury is promising. The underlying mechanisms by which HCA exerts its protective effects are complex, but dampening of the inflammatory response seems to play a pivotal role. After briefly summarizing the physiological effects of HCA, a critical analysis of the available evidence on the potential beneficial effects of therapeutic HCA from in vitro, ex vivo and in vivo lung injury models and from human studies will be reviewed. In addition, the potential concerns in the clinical setting will be outlined.
Collapse
|
42
|
Abstract
Carbon dioxide is a waste product of aerobic cellular respiration in all aerobic life forms. PaCO2 represents the balance between the carbon dioxide produced and that eliminated. Hypocapnia remains a common - and generally underappreciated - component of many disease states, including early asthma, high-altitude pulmonary edema, and acute lung injury. Induction of hypocapnia remains a common, if controversial, practice in both adults and children with acute brain injury. In contrast, hypercapnia has traditionally been avoided in order to keep parameters normal. More recently, advances in our understanding of the role of excessive tidal volume has prompted clinicians to use ventilation strategies that result in hypercapnia. Consequently, hypercapnia has become increasingly prevalent in the critically ill patient. Hypercapnia may play a beneficial role in the pathogenesis of inflammation and tissue injury, but may hinder the host response to sepsis and reduce repair. In contrast, hypocapnia may be a pathogenic entity in the setting of critical illness. The present paper reviews the current clinical status of low and high PaCO2 in the critically ill patient, discusses the insights gained to date from studies of carbon dioxide, identifies key concerns regarding hypocapnia and hypercapnia, and considers the potential clinical implications for the management of patients with acute lung injury.
Collapse
Affiliation(s)
- Gerard Curley
- Department of Anaesthesia, Clinical Sciences Institute, National University of Ireland, Galway, Ireland
- Lung Biology Group, National Centre of Biomedical Engineering Sciences, National University of Ireland, Galway, Ireland
| | - John G Laffey
- Department of Anaesthesia, Clinical Sciences Institute, National University of Ireland, Galway, Ireland
- Lung Biology Group, National Centre of Biomedical Engineering Sciences, National University of Ireland, Galway, Ireland
| | - Brian P Kavanagh
- Departments of Critical Care Medicine and Anesthesia and the Program in Physiology and Experimental Medicine, The Hospital for Sick Children, University of Toronto, 555 university Avenue, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
43
|
Chuang IC, Dong HP, Yang RC, Wang TH, Tsai JH, Yang PH, Huang MS. Effect of Carbon Dioxide on Pulmonary Vascular Tone at Various Pulmonary Arterial Pressure Levels Induced by Endothelin-1. Lung 2010; 188:199-207. [DOI: 10.1007/s00408-010-9234-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2009] [Accepted: 02/16/2010] [Indexed: 10/19/2022]
|
44
|
Peltekova V, Engelberts D, Otulakowski G, Uematsu S, Post M, Kavanagh BP. Hypercapnic acidosis in ventilator-induced lung injury. Intensive Care Med 2010; 36:869-78. [PMID: 20213072 DOI: 10.1007/s00134-010-1787-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Accepted: 02/09/2010] [Indexed: 10/19/2022]
Abstract
RATIONALE Permissive hypercapnia is established in lung injury management. Therapeutic hypercapnia causes benefit or harm, depending on the context. Ventilator-associated lung injury has a wide spectrum of candidate mechanisms, affording multiple opportunities for intervention such as hypercapnia to exert benefit or harm. OBJECTIVES To confirm (1) that hypercapnia attenuates in vivo ventilator-induced lung injury (VILI); (2) biological plausibility of such protection (e.g., dose-response, time series, inflammatory profile); and (3) that the associated biochemical events are consistently beneficial. METHODS A mouse model of VILI was established in vivo. Injurious ventilation was established, hypercapnia applied and markers of inflammation measured. MEASUREMENTS Lung injury was quantified by gas exchange, elastance, microvascular leak, histology and levels of cytokines and eicosanoids, cyclooxygenase and tissue nitrotyrosine. MAIN RESULTS Injurious ventilation caused significant lung injury (mechanics, microvascular leak, histology) and release of inflammatory cytokines, chemokines and eicosanoids. Hypercapnia attenuated these responses, with dose-response and time-dependent effects. No adverse effects of hypercapnia were observed in controls. Hypercapnia suppressed the transcription (mRNA) and translation (protein) of the major inducible prostanoid-generating enzyme (COX-2), but the effects on the downstream eicosanoids were modest. However, hypercapnia significantly increased lung tissue nitrotyrosine-at PaCO(2) levels that were protective. CONCLUSIONS Hypercapnia provided consistent and biologically plausible in vivo protection against VILI, but elevated lung tissue levels of nitro-tyrosine as previously described in sepsis. Clinicians and those designing clinical trials need to be aware of the potential for detrimental effects when using hypercapnia in order to balance benefits versus harm with this approach.
Collapse
Affiliation(s)
- Vanya Peltekova
- Physiology and Experimental Medicine, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | | | | | | | | | | |
Collapse
|
45
|
Wang N, Gates KL, Trejo H, Favoreto S, Schleimer RP, Sznajder JI, Beitel GJ, Sporn PHS. Elevated CO2 selectively inhibits interleukin-6 and tumor necrosis factor expression and decreases phagocytosis in the macrophage. FASEB J 2010; 24:2178-90. [PMID: 20181940 DOI: 10.1096/fj.09-136895] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Elevated blood and tissue CO(2), or hypercapnia, is common in severe lung disease. Patients with hypercapnia often develop lung infections and have an increased risk of death following pneumonia. To explore whether hypercapnia interferes with host defense, we studied the effects of elevated P(CO2) on macrophage innate immune responses. In differentiated human THP-1 macrophages and human and mouse alveolar macrophages stimulated with lipopolysaccharide (LPS) and other Toll-like receptor ligands, hypercapnia inhibited expression of tumor necrosis factor and interleukin (IL)-6, nuclear factor (NF)-kappaB-dependent cytokines critical for antimicrobial host defense. Inhibition of IL-6 expression by hypercapnia was concentration dependent, rapid, reversible, and independent of extracellular and intracellular acidosis. In contrast, hypercapnia did not down-regulate IL-10 or interferon-beta, which do not require NF-kappaB. Notably, hypercapnia did not affect LPS-induced degradation of IkappaB alpha, nuclear translocation of RelA/p65, or activation of mitogen-activated protein kinases, but it did block IL-6 promoter-driven luciferase activity in mouse RAW 264.7 macrophages. Elevated P(CO2) also decreased phagocytosis of opsonized polystyrene beads and heat-killed bacteria in THP-1 and human alveolar macrophages. By interfering with essential innate immune functions in the macrophage, hypercapnia may cause a previously unrecognized defect in resistance to pulmonary infection in patients with advanced lung disease.
Collapse
Affiliation(s)
- Naizhen Wang
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, 240 E. Huron St., Chicago, IL 60611, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Masood A, Yi M, Lau M, Belcastro R, Shek S, Pan J, Kantores C, McNamara PJ, Kavanagh BP, Belik J, Jankov RP, Tanswell AK. Therapeutic effects of hypercapnia on chronic lung injury and vascular remodeling in neonatal rats. Am J Physiol Lung Cell Mol Physiol 2009; 297:L920-30. [DOI: 10.1152/ajplung.00139.2009] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Permissive hypercapnia, achieved using low tidal volume ventilation, has been an effective protective strategy in patients with acute respiratory distress syndrome. To date, no such protective effect has been demonstrated for the chronic neonatal lung injury, bronchopulmonary dysplasia. The objective of our study was to determine whether evolving chronic neonatal lung injury, using a rat model, is resistant to the beneficial effects of hypercapnia or simply requires a less conservative approach to hypercapnia than that applied clinically to date. Neonatal rats inhaled air or 60% O2 for 14 days with or without 5.5% CO2. Lung parenchymal neutrophil and macrophage numbers were significantly increased by hyperoxia alone, which was associated with interstitial thickening and reduced secondary crest formation. The phagocyte influx, interstitial thickening, and impaired alveolar formation were significantly attenuated by concurrent hypercapnia. Hyperoxic pups that received 5.5% CO2 had a significant increase in alveolar number relative to air-exposed pups. Increased tyrosine nitration, a footprint for peroxynitrite-mediated reactions, arteriolar medial wall thickening, and both reduced small peripheral pulmonary vessel number and VEGF and angiopoietin-1 (Ang-1) expression, which were observed with hyperoxia, was attenuated by concurrent hypercapnia. We conclude that evolving chronic neonatal lung injury in a rat model is responsive to the beneficial effects of hypercapnia. Inhaled 5.5% CO2 provided a significant degree of protection against parenchymal and vascular injury in an animal model of chronic neonatal lung injury likely due, at least in part, to its inhibition of a phagocyte influx.
Collapse
Affiliation(s)
- Azhar Masood
- Canadian Institutes of Health Research Group in Lung Development, and
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
- Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Man Yi
- Canadian Institutes of Health Research Group in Lung Development, and
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
- Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Mandy Lau
- Canadian Institutes of Health Research Group in Lung Development, and
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
- Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Rosetta Belcastro
- Canadian Institutes of Health Research Group in Lung Development, and
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
| | - Samuel Shek
- Canadian Institutes of Health Research Group in Lung Development, and
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
| | - Jingyi Pan
- Canadian Institutes of Health Research Group in Lung Development, and
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
| | - Crystal Kantores
- Clinical Integrative Biology, Sunnybrook Research Institute; and
| | - Patrick J. McNamara
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
- Paediatrics, and
| | - Brian P. Kavanagh
- Canadian Institutes of Health Research Group in Lung Development, and
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
- Departments of 4Anaesthesia,
- Critical Care Medicine,
- Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Jaques Belik
- Canadian Institutes of Health Research Group in Lung Development, and
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
- Paediatrics, and
- Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Robert P. Jankov
- Clinical Integrative Biology, Sunnybrook Research Institute; and
- Paediatrics, and
- Physiology, University of Toronto, Toronto, Ontario, Canada
| | - A. Keith Tanswell
- Canadian Institutes of Health Research Group in Lung Development, and
- Lung Biology Programme, Physiology and Experimental Medicine, Hospital for Sick Children Research Institute
- Paediatrics, and
- Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
47
|
Hypercapnic acidosis attenuates shock and lung injury in early and prolonged systemic sepsis. Crit Care Med 2009; 37:2412-20. [DOI: 10.1097/ccm.0b013e3181a385d3] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
48
|
Gnaegi A, Feihl F, Boulat O, Waeber B, Liaudet L. Moderate hypercapnia exerts beneficial effects on splanchnic energy metabolism during endotoxemia. Intensive Care Med 2009; 35:1297-304. [PMID: 19373455 DOI: 10.1007/s00134-009-1488-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Accepted: 03/22/2009] [Indexed: 02/07/2023]
Abstract
PURPOSE Low tidal volume ventilation and permissive hypercapnia are required in patients with sepsis complicated by ARDS. The effects of hypercapnia on tissue oxidative metabolism in this setting are unknown. We therefore determined the effects of moderate hypercapnia on markers of systemic and splanchnic oxidative metabolism in an animal model of endotoxemia. METHODS Anesthetized rats maintained at a PaCO(2) of 30, 40 or 60 mmHg were challenged with endotoxin. A control group (PaCO(2) 40 mmHg) received isotonic saline. Hemodynamic variables, arterial lactate, pyruvate, and ketone bodies were measured at baseline and after 4 h. Tissue adenosine triphosphate (ATP) and lactate were measured in the small intestine and the liver after 4 h. RESULTS Endotoxin resulted in low cardiac output, increased lactate/pyruvate ratio and decreased ketone body ratio. These changes were not influenced by hypercapnia, but were more severe with hypocapnia. In the liver, ATP decreased and lactate increased independently from PaCO(2) after endotoxin. In contrast, the drop of ATP and the rise in lactate triggered by endotoxin in the intestine were prevented by hypercapnia. CONCLUSIONS During endotoxemia in rats, moderate hypercapnia prevents the deterioration of tissue energetics in the intestine.
Collapse
Affiliation(s)
- Alex Gnaegi
- Division of Clinical Pathophysiology and Medical Teaching, Faculty of Biology and Medicine, University Hospital Center, CHUV-BH 08-621, 1011, Lausanne, Switzerland
| | | | | | | | | |
Collapse
|
49
|
Hypercapnic acidosis in acute lung injury: inevitable side effect or unexpected benefit? Crit Care Med 2008; 36:3268-9. [PMID: 19020438 DOI: 10.1097/ccm.0b013e31818f2477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|