1
|
Perchiazzi G, Ranieri VM. Self-Induced Lung Injury and the Quest for a Smoking Gun: A Nosologic Perspective. Acta Anaesthesiol Scand 2025; 69:e70072. [PMID: 40528503 DOI: 10.1111/aas.70072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2025] [Revised: 05/24/2025] [Accepted: 05/28/2025] [Indexed: 06/20/2025]
Affiliation(s)
- Gaetano Perchiazzi
- The Hedenstierna Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Department of Anesthesia, Operation and Intensive Care, Uppsala University Hospital, Uppsala, Sweden
| | - V Marco Ranieri
- Dipartimento di Anestesia, Rianimazione e Terapia Intensiva, Università Degli Studi di Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
2
|
Xu X, Liu W, Xu Y, Fan Y, Han F, Pan J, Lu G, Yi C, Zhang Q. IL-37 Protects Against Ventilator-Induced Lung Injury by Inhibiting NLRP3 Activation. Cell Biochem Funct 2025; 43:e70080. [PMID: 40325829 DOI: 10.1002/cbf.70080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 04/02/2025] [Accepted: 04/25/2025] [Indexed: 05/07/2025]
Abstract
Mechanical ventilation is an effective strategy for managing acute respiratory distress syndrome (ARDS), but it can also exacerbate lung injury, leading to ventilator-induced lung injury (VILI). To investigate the protective role of interleukin-37 (IL-37) in the pathogenesis of VILI, we used two approaches, human IL-37 transgenic (IL37tg) mice and administration of recombinant human IL-37 (rIL37) in wild-type (WT) mice subjected to mechanical ventilation. Lung histopathology was evaluated, inflammatory cytokine levels (IL-1β, IL-6, TNF-α) were measured, and inflammasome activation was assessed by analyzing NLRP3 and Caspase-1 expression. As a result, IL37tg mice exhibited significantly attenuated lung injury compared to WT controls, characterized by improved histological morphology, reduced lung injury scores, and decreased infiltration of macrophages and neutrophils. Similarly, rIL37 administration markedly reduced lung injury and decreased inflammatory cytokine levels. Immunofluorescence analysis revealed colocalization of the alveolar cell marker surfactant protein D (SP-D) and IL-37. Furthermore, IL-37 suppressed NLRP3 inflammasome activation, as evidenced by reduced NLRP3 and Cleaved-Caspase-1 levels in both the IL37tg mouse model and the rIL37 treatment group. These findings suggest that IL-37 effectively protects against VILI by inhibiting inflammation in lung tissues through inhibition of the NLRP3 inflammasome. Therefore, IL-37 may serve as a potential therapeutic target for the prevention and treatment of VILI in the future.
Collapse
Affiliation(s)
- Xingmeng Xu
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Pancreatic Center, Yangzhou University, Yangzhou, Jiangsu, China
- Yangzhou Key Laboratory of Pancreatic Disease, Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Weili Liu
- Department of Intensive Care Unit, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yan Xu
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yinghong Fan
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Pancreatic Center, Yangzhou University, Yangzhou, Jiangsu, China
- Yangzhou Key Laboratory of Pancreatic Disease, Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Fei Han
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Pancreatic Center, Yangzhou University, Yangzhou, Jiangsu, China
- Yangzhou Key Laboratory of Pancreatic Disease, Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
- Dalian Medical University, Dalian, Liaoning, China
| | - Jiajia Pan
- Department of Intensive Care Unit, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Guotao Lu
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Pancreatic Center, Yangzhou University, Yangzhou, Jiangsu, China
- Yangzhou Key Laboratory of Pancreatic Disease, Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Chenlong Yi
- Department of Cardiovascular Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Qingfen Zhang
- Department of Anesthesiology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
3
|
Tian X, Lu B, Huang Y, Zhong W, Lei X, Liu S, Tao T, Yun F, Huang S, Tan T, Liu H, Zhou Z, Peng G, Wang Y, Zhang K, Luo X, Zhong Z. Associated effects of lipopolysaccharide, oleic acid, and lung injury ventilator-induced in developing a model of moderate acute respiratory distress syndrome in New Zealand white rabbits. Front Vet Sci 2025; 12:1477554. [PMID: 40177676 PMCID: PMC11963770 DOI: 10.3389/fvets.2025.1477554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a critical pulmonary disorder with manifestations of pulmonary edema, inflammation, and impaired oxygenation. Establishing reliable animal ARDS models has been critical for investigating its mechanisms and for testing pharmacological interventions. The present study sought to induce a moderate ARDS model in New Zealand White rabbits with a model involving a mix of lipopolysaccharide (LPS), oleic acid (OA), and ventilation-induced lung injury (VILI). Four experimental groups were established: negative control (NC, n = 4), OA (OM, n = 6), LPS + OA (LOM, n = 6), and LPS + OA + VILI (LOV, n = 6). Throughout the modeling process, vital signs (MAP and HR), respiratory parameters (Cdyn), and hematological indices (WBC and P/F) were continuously monitored, and lung ultrasound was performed. After the experiment, bronchoalveolar lavage fluid (BALF) was collected to measure total protein content, and lung tissue samples were collected to determine the wet-to-dry (W/D) ratio. HE-stained lung tissue sections were prepared and scored according to the ATS guidelines for lung injury scoring. The LOV group showed the most severe lung injury, significantly decreasing MAP and Cdyn. Pathological and ultrasound scores were considerably higher in the LOV group compared to the OM and LOM groups (p < 0.05). The lung W/D ratio was significantly higher in the LOM (6.68 ± 0.56) and LOV (7.40 ± 0.56) groups compared to the NC group (5.20 ± 0.16) (p < 0.05). At T6, the PaO2/FiO2 ratio in the LOV group was ≤200 mmHg, significantly lower than that in the NC group (p < 0.05). Some rabbits in the OM and LOM groups also had PaO2/FiO2 ratios ≤200 mmHg, but the difference compared to the NC group was not statistically significant. In conclusion, this study established a novel moderate ARDS model in New Zealand White rabbits using LPS, OA, and VILI. The model demonstrates severe lung damage, pulmonary edema, and sustained hypoxemia, providing a basis for future research.
Collapse
Affiliation(s)
- Xingyu Tian
- Key Laboratory of Animal Disease and Human Health of Sichuan, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Bin Lu
- Department of Pediatric Critical Medicine, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuyan Huang
- Key Laboratory of Animal Disease and Human Health of Sichuan, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Wenhao Zhong
- Key Laboratory of Animal Disease and Human Health of Sichuan, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xin Lei
- Key Laboratory of Animal Disease and Human Health of Sichuan, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Siyu Liu
- Key Laboratory of Animal Disease and Human Health of Sichuan, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Tao Tao
- Department of Pediatric Critical Medicine, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Fengning Yun
- Department of Pediatric Critical Medicine, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shiyong Huang
- Department of Pediatric Critical Medicine, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Tiqing Tan
- Department of Pediatric Critical Medicine, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Haifeng Liu
- Key Laboratory of Animal Disease and Human Health of Sichuan, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ziyao Zhou
- Key Laboratory of Animal Disease and Human Health of Sichuan, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Guangneng Peng
- Key Laboratory of Animal Disease and Human Health of Sichuan, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ya Wang
- Key Laboratory of Animal Disease and Human Health of Sichuan, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Kun Zhang
- Key Laboratory of Animal Disease and Human Health of Sichuan, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xiaoli Luo
- Department of Pediatric Critical Medicine, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhijun Zhong
- Key Laboratory of Animal Disease and Human Health of Sichuan, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
4
|
Zhang NR, Zhang LZ, Chen Y, Zhang S, Li S, Gu XK, Li J, Li H. Intraoperative protective ventilation with or without periodic lung recruitment manoeuvres on pulmonary complications after major abdominal surgery (REMAIN-1): protocol for a randomised controlled trial. BMJ Open 2025; 15:e093360. [PMID: 40082005 PMCID: PMC11906986 DOI: 10.1136/bmjopen-2024-093360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/24/2025] [Indexed: 03/16/2025] Open
Abstract
INTRODUCTION Postoperative pulmonary complications (PPCs) are frequent after abdominal surgery and significantly affect postoperative outcomes. Intraoperative protective ventilation (IPV) has been demonstrated to mitigate PPCs. However, the comparative effectiveness of two common IPV regimens-IPV with or without periodic lung recruitment manoeuvres (PLRM)-in preventing PPCs is unclear. This study aims to compare the effects of these two IPV regimens on PPCs. METHODS AND ANALYSIS This study is a prospective, double-blinded, randomised controlled trial. A total of 1060 patients at intermediate or high risk for PPCs, scheduled to undergo major abdominal surgery, will be enrolled and randomly assigned to receive either an IPV with PLRM (intensive IPV group) or an IPV without PLRM (moderate IPV group). Patients assigned to the intensive IPV group will receive positive end-expiratory pressure (PEEP) of 6-10 cm H2O with lung recruitment manoeuvres performed every 30 min. Patients in the moderate IPV group will receive the same level of PEEP without lung recruitment manoeuvres. Both groups will receive a tidal volume of 7 mL/kg predicted body weight and an inspired oxygen fraction of 0.3-0.4. The primary outcome is respiratory failure within the first 7 postoperative days. Secondary outcomes include other PPCs, extrapulmonary complications, unplanned admissions to the intensive care unit, length of postoperative hospital stay and mortality from any cause. ETHICS AND DISSEMINATION This protocol has been approved by the Ethics Committee of the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China. The first participant was recruited on 9 October 2022, with an estimated completion date of 30 May 2025. The results of this trial are expected to be published in peer-reviewed journals. TRIAL REGISTRATION NUMBER NCT05556174.
Collapse
Affiliation(s)
- Nan-Rong Zhang
- Department of Anaesthesia, Sun Yat-sen University Sixth Affiliated Hospital, Guangzhou, Guangdong, China
- Biomedical Innovation Centre, Sun Yat-sen University Sixth Affiliated Hospital, Guangzhou, Guangdong, China
| | - Li-Zhen Zhang
- Department of Anaesthesia, Sun Yat-sen University Sixth Affiliated Hospital, Guangzhou, Guangdong, China
- Biomedical Innovation Centre, Sun Yat-sen University Sixth Affiliated Hospital, Guangzhou, Guangdong, China
| | - Yi Chen
- Department of Anaesthesia, Sun Yat-sen University Sixth Affiliated Hospital, Guangzhou, Guangdong, China
- Biomedical Innovation Centre, Sun Yat-sen University Sixth Affiliated Hospital, Guangzhou, Guangdong, China
| | - Song Zhang
- Department of Anaesthesia, Sun Yat-sen University Sixth Affiliated Hospital, Guangzhou, Guangdong, China
| | - Shan Li
- Department of Anaesthesia, Sun Yat-sen University Sixth Affiliated Hospital, Guangzhou, Guangdong, China
| | - Xiao-Ke Gu
- Department of Anaesthesia, Sun Yat-sen University Sixth Affiliated Hospital, Guangzhou, Guangdong, China
| | - Jing Li
- Department of Anaesthesia, Sun Yat-sen University Sixth Affiliated Hospital, Guangzhou, Guangdong, China
- Biomedical Innovation Centre, Sun Yat-sen University Sixth Affiliated Hospital, Guangzhou, Guangdong, China
| | - Hong Li
- Department of Anaesthesia, Sun Yat-sen University Sixth Affiliated Hospital, Guangzhou, Guangdong, China
- Biomedical Innovation Centre, Sun Yat-sen University Sixth Affiliated Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Zhou K, Qin Q, Lu J. Pathophysiological mechanisms of ARDS: a narrative review from molecular to organ-level perspectives. Respir Res 2025; 26:54. [PMID: 39948645 PMCID: PMC11827456 DOI: 10.1186/s12931-025-03137-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) remains a life-threatening pulmonary condition with persistently high mortality rates despite significant advancements in supportive care. Its complex pathophysiology involves an intricate interplay of molecular and cellular processes, including cytokine storms, oxidative stress, programmed cell death, and disruption of the alveolar-capillary barrier. These mechanisms drive localized lung injury and contribute to systemic inflammatory response syndrome and multiple organ dysfunction syndrome. Unlike prior reviews that primarily focus on isolated mechanisms, this narrative review synthesizes the key pathophysiological processes of ARDS across molecular, cellular, tissue, and organ levels. MAIN BODY By integrating classical theories with recent research advancements, we provide a comprehensive analysis of how inflammatory mediators, metabolic reprogramming, oxidative stress, and immune dysregulation synergistically drive ARDS onset and progression. Furthermore, we critically evaluate current evidence-based therapeutic strategies, such as lung-protective ventilation and prone positioning, while exploring innovative therapies, including stem cell therapy, gene therapy, and immunotherapy. We emphasize the significance of ARDS subtypes and their inherent heterogeneity in guiding the development of personalized treatment strategies. CONCLUSIONS This narrative review provides fresh perspectives for future research, ultimately enhancing patient outcomes and optimizing management approaches in ARDS.
Collapse
Affiliation(s)
- Kaihuan Zhou
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Qianqian Qin
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Junyu Lu
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China.
| |
Collapse
|
6
|
Ding X, Jin S, Tian W, Zhang Y, Xu L, Zhang T, Chen Z, Niu F, Li Q. ROLE OF CASPASE-1/CASPASE-11-HMGB1-RAGE/TLR4 SIGNALING IN THE EXACERBATION OF EXTRAPULMONARY SEPSIS-INDUCED LUNG INJURY BY MECHANICAL VENTILATION. Shock 2025; 63:299-311. [PMID: 39228020 DOI: 10.1097/shk.0000000000002471] [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: 09/05/2024]
Abstract
ABSTRACT Background: Mechanical ventilation (MV) is a clinically important measure for respiratory support in critically ill patients. Although moderate tidal volume MV does not cause lung injury, it can further exacerbate lung injury in a pathological state such as sepsis. This pathological process is known as the "two-hit" theory, whereby an initial lung injury (e.g., infection, trauma, or sepsis) triggers an inflammatory response that activates immune cells, presenting the lung tissue in a fragile state and rendering it more susceptible to subsequent injury. The second hit occurs when MV is applied to lung tissue in a fragile state, and it is noteworthy that this MV is harmless to healthy lung tissue, further aggravating preexisting lung injury through unknown mechanisms. This interaction between initial injury and subsequent MV develops a malignant cycle significantly exacerbating lung injury and severely hampering patient prognosis. The two-hit theory is critical to understanding the complicated mechanisms of ventilator-associated lung injury and facilitates the subsequent development of targeted therapeutic strategies. Methods and Results: The cecum ligation and perforation mice model was used to mimic clinical sepsis patients. After 12 h, the mice were mechanically ventilated for 2 to 6 h. MV by itself did not lead to HMGB1 release, but significantly strengthened HMGB1 in plasma and cytoplasm of lung tissue in septic mice. Plasma and lung tissue activation of cytokines and chemokines, mitogen-activated protein kinase signaling pathway, neutrophil recruitment, and acute lung injury were progressively decreased in LysM HMGB1 -/- (Hmgb1 deletion in myeloid cells) and iHMGB1 -/- mice (inducible HMGB1 -/- mouse strain where the Hmgb1 gene was globally deleted after tamoxifen treatment). Compared with C57BL/6 mice, although EC-HMGB1 -/- (Hmgb1 deletion in endothelial cells) mice did not have lower levels of inflammation, neutrophil recruitment and lung injury were reduced. Compared with LysM HMGB1 -/- mice, EC-HMGB1 -/- mice had higher levels of inflammation but significantly lower neutrophil recruitment and lung injury. Overall, iHMGB1 -/- mice had the lowest levels of all the above indicators. The level of inflammation, neutrophil recruitment, and the degree of lung injury were decreased in RAGE -/- mice, and even the above indices were further decreased in TLR4/RAGE -/- mice. Levels of inflammation and neutrophil recruitment were decreased in caspase-11 -/- and caspase-1/11 -/- mice, but there was no statistical difference between these two gene knockout mice. Conclusions: These data show for the first time that the caspase-1/caspase-11-HMGB1-TLR4/RAGE signaling pathway plays a key role in mice model of sepsis-induced lung injury exacerbated by MV. Different species of HMGB1 knockout mice have different lung-protective mechanisms in the two-hit model, and location is the key to function. Specifically, LysM HMGB1 -/- mice due to the deletion of HMGB1 in myeloid cells resulted in a pulmonary-protective mechanism that was associated with a downregulation of the inflammatory response. EC-HMGB1 -/- mice are deficient in HMGB1 owing to endothelial cells, resulting in a distinct pulmonary-protective mechanism independent of the inflammatory response and more relevant to the improvement of alveolar-capillary permeability. iHMGB1 -/- mice, which are systemically HMGB1-deficient, share both of these lung-protective mechanisms.
Collapse
Affiliation(s)
| | | | - Weitian Tian
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yizhe Zhang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | - Tong Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Zhixia Chen
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Fangfang Niu
- Department of Anesthesiology, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Quan Li
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| |
Collapse
|
7
|
Marron A, Totapally A, Weatherly A, De S, Barber M, Lifsey A, Boyle K. Pediatric Health Assessment Tracker: A Quality Improvement Initiative to Obtain Weights Consistently and Appropriately in a Tertiary Pediatric Intensive Care Unit. Pediatr Qual Saf 2025; 10:e791. [PMID: 39822276 PMCID: PMC11737482 DOI: 10.1097/pq9.0000000000000791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 12/30/2024] [Indexed: 01/19/2025] Open
Abstract
Introduction Weight is vital for tracking fluid status and nutrition and assuring patients have accurate dosing weights in the pediatric intensive care unit (PICU). Challenges in acquiring weights in critically ill patients include clinical instability, limited equipment, and lack of appropriate orders in the electronic medical record (EMR). Methods We implemented interventions that targeted EMR weight orders and actual collection of weights in the 42-bed PICU of a children's hospital. Preintervention data were collected from February to March 2023 for all patients admitted to the PICU with a length of stay (LOS) ≥3 days. We surveyed PICU nurses to identify barriers to collecting weights. Interventions included a multidisciplinary team approach, safety checklist, nursing education, and automatization of weight orders. The study team monitored the number of patients with weight orders and weights obtained as ordered twice weekly from March 2023 to April 2024 using statistical process control charts. Results There were 1728 patient instances of LOS ≥3 days. Preintervention data showed 70.4% of patients with appropriate weight orders and 35.5% with weights obtained. Implementing a safety checklist, nursing education, EMR changes, and automatizing weight orders, the centerline for weight orders shifted to 94.3% and for weights obtained to 69.5%. Reminder emails to all ICU providers and nursing check-ins maintained the centerline. No increase in unplanned extubations occurred. Conclusions Through interventions involving rounding providers, nurses, and the EMR, the frequency of weights ordered and obtained in a busy PICU sustainably increased.
Collapse
Affiliation(s)
- Alonso Marron
- From the Department of Pediatrics, Monroe Carell Jr Children’s Hospital at Vanderbilt, Vanderbilt University Medical Center, Nashville, Tenn
| | - Abhinav Totapally
- From the Department of Pediatrics, Monroe Carell Jr Children’s Hospital at Vanderbilt, Vanderbilt University Medical Center, Nashville, Tenn
| | - Allison Weatherly
- From the Department of Pediatrics, Monroe Carell Jr Children’s Hospital at Vanderbilt, Vanderbilt University Medical Center, Nashville, Tenn
| | - Subhendu De
- From the Department of Pediatrics, Monroe Carell Jr Children’s Hospital at Vanderbilt, Vanderbilt University Medical Center, Nashville, Tenn
| | - Madeline Barber
- From the Department of Pediatrics, Monroe Carell Jr Children’s Hospital at Vanderbilt, Vanderbilt University Medical Center, Nashville, Tenn
| | - Allyson Lifsey
- From the Department of Pediatrics, Monroe Carell Jr Children’s Hospital at Vanderbilt, Vanderbilt University Medical Center, Nashville, Tenn
| | - Katharine Boyle
- From the Department of Pediatrics, Monroe Carell Jr Children’s Hospital at Vanderbilt, Vanderbilt University Medical Center, Nashville, Tenn
| |
Collapse
|
8
|
Monet C, Renault T, Aarab Y, Pensier J, Prades A, Lakbar I, Le Bihan C, Capdevila M, De Jong A, Molinari N, Jaber S. Feasibility and safety of ultra-low volume ventilation (≤ 3 ml/kg) combined with extra corporeal carbon dioxide removal (ECCO 2R) in acute respiratory failure patients. Crit Care 2024; 28:433. [PMID: 39731126 PMCID: PMC11674201 DOI: 10.1186/s13054-024-05168-8] [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: 09/26/2024] [Accepted: 11/11/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND Ultra-protective ventilation is the combination of low airway pressures and tidal volume (Vt) combined with extra corporeal carbon dioxide removal (ECCO2R). A recent large study showed no benefit of ultra-protective ventilation compared to standard ventilation in ARDS (Acute Respiratory Distress Syndrome) patients. However, the reduction in Vt failed to achieve the objective of less than or equal to 3 ml/kg predicted body weight (PBW). The main objective of our study was to assess the feasibility of the ultra-low volume ventilation (Vt ≤ 3 ml/kg PBW) facilitated by ECCO2R in acute respiratory failure patients. METHODS Retrospective analysis of a prospective cohort of patients with either high or low blood flow veno-venous ECCO2R devices. A session was defined as a treatment of ECCO2R from the start to the removal of the device (one patient could have one more than one session). Primary endpoint was the proportion of sessions during which a Vt less or equal to 3 ml/kg PBW at 24 h after the start of ECCO2R was successfully achieved for at least 12 h. Secondary endpoints were respiratory variables, rate of adverse events and outcomes. RESULTS Forty-five ECCO2R sessions were recorded among 41 patients. Ultra-low volume ventilation (tidal volume ≤ 3 ml/kg PBW, success group) was successfully achieved at 24 h in 40.0% sessions (18 out of 45 sessions, confidence interval 25.3-54.6%). At 24 h, tidal volume in the failure group was 4.1 [3.8-4.5] ml/kg PBW compared to 2.1 [1.9-2.5] in the success group (p < 0.001). After multivariate analysis, blood flow rate was significantly associated with success of ultra-low volume ventilation (adjusted OR per 100 ml/min increase 1.51 (95%CI 1.21-1.90, p = 0.0003). CONCLUSION Ultra-low volume ventilation (≤ 3 ml/kg PBW) was feasible in 18 out of 45 sessions. Higher blood flow rates were associated with the success of ultra-low volume ventilation.
Collapse
Affiliation(s)
- Clément Monet
- Department of Anesthesia and Intensive Care Unit, Regional University Hospital of Montpellier, St-Eloi Hospital, PhyMedExp, INSERM U1046, CNRS UMR, University of Montpellier, 9214, Montpellier Cedex 5, France
- PhyMedExp, INSERM U1046, CNRS UMR, University of Montpellier, 9214, Montpellier, France
| | - Thomas Renault
- Department of Anesthesia and Intensive Care Unit, Regional University Hospital of Montpellier, St-Eloi Hospital, PhyMedExp, INSERM U1046, CNRS UMR, University of Montpellier, 9214, Montpellier Cedex 5, France
| | - Yassir Aarab
- Department of Anesthesia and Intensive Care Unit, Regional University Hospital of Montpellier, St-Eloi Hospital, PhyMedExp, INSERM U1046, CNRS UMR, University of Montpellier, 9214, Montpellier Cedex 5, France
- PhyMedExp, INSERM U1046, CNRS UMR, University of Montpellier, 9214, Montpellier, France
| | - Joris Pensier
- Department of Anesthesia and Intensive Care Unit, Regional University Hospital of Montpellier, St-Eloi Hospital, PhyMedExp, INSERM U1046, CNRS UMR, University of Montpellier, 9214, Montpellier Cedex 5, France
- PhyMedExp, INSERM U1046, CNRS UMR, University of Montpellier, 9214, Montpellier, France
| | - Albert Prades
- Department of Anesthesia and Intensive Care Unit, Regional University Hospital of Montpellier, St-Eloi Hospital, PhyMedExp, INSERM U1046, CNRS UMR, University of Montpellier, 9214, Montpellier Cedex 5, France
| | - Ines Lakbar
- Department of Anesthesia and Intensive Care Unit, Regional University Hospital of Montpellier, St-Eloi Hospital, PhyMedExp, INSERM U1046, CNRS UMR, University of Montpellier, 9214, Montpellier Cedex 5, France
- PhyMedExp, INSERM U1046, CNRS UMR, University of Montpellier, 9214, Montpellier, France
| | - Clément Le Bihan
- Department of Anesthesia and Intensive Care Unit, Regional University Hospital of Montpellier, St-Eloi Hospital, PhyMedExp, INSERM U1046, CNRS UMR, University of Montpellier, 9214, Montpellier Cedex 5, France
| | - Mathieu Capdevila
- Department of Anesthesia and Intensive Care Unit, Regional University Hospital of Montpellier, St-Eloi Hospital, PhyMedExp, INSERM U1046, CNRS UMR, University of Montpellier, 9214, Montpellier Cedex 5, France
- PhyMedExp, INSERM U1046, CNRS UMR, University of Montpellier, 9214, Montpellier, France
| | - Audrey De Jong
- Department of Anesthesia and Intensive Care Unit, Regional University Hospital of Montpellier, St-Eloi Hospital, PhyMedExp, INSERM U1046, CNRS UMR, University of Montpellier, 9214, Montpellier Cedex 5, France.
- PhyMedExp, INSERM U1046, CNRS UMR, University of Montpellier, 9214, Montpellier, France.
| | - Nicolas Molinari
- Medical Information, IMAG, CNRS, Centre Hospitalier Regional Universitaire de Montpellier, Univ Montpellier, Montpellier, France
- Département d'informatique Médicale, CHRU Montpellier, Institut Desbrest de Santé Publique (IDESP) INSERM, Université de Montpellier, Montpellier, France
| | - Samir Jaber
- Department of Anesthesia and Intensive Care Unit, Regional University Hospital of Montpellier, St-Eloi Hospital, PhyMedExp, INSERM U1046, CNRS UMR, University of Montpellier, 9214, Montpellier Cedex 5, France.
- PhyMedExp, INSERM U1046, CNRS UMR, University of Montpellier, 9214, Montpellier, France.
| |
Collapse
|
9
|
Hong Y, Shin S, Nasim U, Roberts KG, Potchernikov AS, Liu KY, Dufendach KA, Skoog DJ, Bacchetta M, Cook KE. Hemocompatibility Evaluation of a Novel Ambulatory Pulmonary Assist System Using a Lightweight Axial-Flow Pump. ASAIO J 2024; 70:971-978. [PMID: 38728744 PMCID: PMC11527577 DOI: 10.1097/mat.0000000000002227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024] Open
Abstract
The Pulmonary Assist System (PAS) is currently under development as a wearable respiratory assist system. In this study, the hemocompatibility of the PAS's axial-flow mechanical pump (AFP) was compared to other contemporary mechanical pumps in an acute ovine model. The PAS was attached to a normal sheep in a venovenous configuration using one of three pumps: 1) AFP, 2) ReliantHeart HeartAssist 5 (control), or 3) Abbott Pedimag (control) (n = 5 each). Each sheep was supported on the PAS for 12 hours with two L/minute of blood flow and four L/minute of sweep gas. Hemolysis, coagulation, inflammation, and platelet activation and loss were compared among the groups. In this study, the plasma-free hemoglobin (pfHb) was less than 10 mg/dl in all groups. The pfHb was significantly lower in the AFP group compared to other groups. There was no significant clot formation in the pumps and oxygenators in all groups. Furthermore, no significant differences in coagulation (oxygenator resistance, fibrinopeptide A), inflammation (white blood cell counts, IL-8), and platelet activation and loss (p-selectin, platelet counts) were observed among the groups (all, p > 0.05). This study demonstrates equivalent hemocompatibility of the PAS's AFP to other contemporary mechanical pumps with a reduced level of hemolysis on startup.
Collapse
Affiliation(s)
- Yeahwa Hong
- From the Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
- Surgery
| | - Suji Shin
- From the Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Umar Nasim
- From the Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Kalliope G Roberts
- From the Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Alexander S Potchernikov
- From the Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Kimberly Y Liu
- From the Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Keith A Dufendach
- Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - David J Skoog
- Advanced Respiratory Technologies, Inc, Pittsburgh, Pennsylvania
| | - Matthew Bacchetta
- Cardiac Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Keith E Cook
- From the Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| |
Collapse
|
10
|
Mo J, Wang D, Xiao J, Chen Q, An R, Liu HL. Effects of lung protection ventilation strategies on postoperative pulmonary complications after noncardiac surgery: a network meta-analysis of randomized controlled trials. BMC Anesthesiol 2024; 24:346. [PMID: 39342110 PMCID: PMC11437922 DOI: 10.1186/s12871-024-02737-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND The purpose of this network meta-analysis was to assess the impact of different protective ventilatory strategies on postoperative pulmonary complications (PPCs). METHODS Several databases were searched for randomized controlled trials (RCTs) that were published before October 2023 in a network meta-analysis. We assessed the effect of different lung-protective ventilation strategies on the incidence of PPCs using Bayesian network meta-analysis. RESULTS We included 58 studies (11610 patients) in this meta-analysis. The network meta-analysis showed that low tidal volumes (LTVs) combined with iPEEP and recruitment manoeuvres (RM) was associated with significantly lower incidence of PPCs [HTVs: OR = 0.38, 95%CrI (0.19, 0.75), LTVs: OR = 0.33, 95%CrI (0.12, 0.82)], postoperative atelectasis[HTVs: OR = 0.2, 95%CrI (0.08, 0.48), LTVs: OR = 0.47, 95%CrI (0.11, 0.93)], and pneumonia[HTVs: OR = 0.22, 95%CrI (0.09, 0.48), LTVs: OR = 0.27, 95%CrI (0.08,0.89)] than was High tidal volumes (HTVs) or LTVs. LTVs combined with medium-to-high PEEP and RM were associated with significantly lower incidence of postoperative atelectasis, and pneumonia. CONCLUSION LTVs combined with iPEEP and RM decreased the incidence of PPCs, postoperative atelectasis, and pneumonia in noncardiac surgery patients. Individual PEEP-guided ventilation was the optimal lung protection ventilation strategy. The quality of evidence is moderate. TRIAL REGISTRATION PROSPERO identifier CRD42023399485.
Collapse
Affiliation(s)
- Jun Mo
- Department of Anesthesiology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Dan Wang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital-Chongqing, Chongqing, 40030, China
| | - Jingyu Xiao
- Department of Anesthesiology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Qi Chen
- Department of Anesthesiology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Ran An
- Department of Anesthesiology, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| | - Hong Liang Liu
- Department of Anesthesiology, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| |
Collapse
|
11
|
Gabela-Zuniga B, Shukla VC, Bobba C, Higuita-Castro N, Powell HM, Englert JA, Ghadiali SN. A micro-scale humanized ventilator-on-a-chip to examine the injurious effects of mechanical ventilation. LAB ON A CHIP 2024; 24:4390-4402. [PMID: 39161999 PMCID: PMC11407794 DOI: 10.1039/d4lc00143e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Patients with compromised respiratory function frequently require mechanical ventilation to survive. Unfortunately, non-uniform ventilation of injured lungs generates complex mechanical forces that lead to ventilator induced lung injury (VILI). Although investigators have developed lung-on-a-chip systems to simulate normal respiration, modeling the complex mechanics of VILI as well as the subsequent recovery phase is a challenge. Here we present a novel humanized in vitro ventilator-on-a-chip (VOC) model of the lung microenvironment that simulates the different types of injurious forces generated in the lung during mechanical ventilation. We used transepithelial/endothelial electrical impedance measurements to investigate how individual and simultaneous application of mechanical forces alters real-time changes in barrier integrity during and after injury. We find that compressive stress (i.e. barotrauma) does not significantly alter barrier integrity while over-distention (20% cyclic radial strain, volutrauma) results in decreased barrier integrity that quickly recovers upon removal of mechanical stress. Conversely, surface tension forces generated during airway reopening (atelectrauma), result in a rapid loss of barrier integrity with a delayed recovery relative to volutrauma. Simultaneous application of cyclic stretching (volutrauma) and airway reopening (atelectrauma), indicates that the surface tension forces associated with reopening fluid-occluded lung regions are the primary driver of barrier disruption. Thus, our novel VOC system can monitor the effects of different types of injurious forces on barrier disruption and recovery in real-time and can be used to interogate the biomechanical mechanisms of VILI.
Collapse
Affiliation(s)
- Basia Gabela-Zuniga
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
| | - Vasudha C Shukla
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
| | - Christopher Bobba
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
- Department of Neurosurgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Heather M Powell
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.
- Department of Materials Science and Engineering, The Ohio State University, Columbus, Ohio, USA
- Scientific Staff, Shriners Children's Ohio, Dayton, Ohio, USA
| | - Joshua A Englert
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Samir N Ghadiali
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA.
- The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State Wexner Medical Center, Columbus, Ohio, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
12
|
Itagaki T, Akimoto Y, Takashima T, Oto J. Ultrasonographic Assessment of the Diaphragm. Diagnostics (Basel) 2024; 14:1481. [PMID: 39061618 PMCID: PMC11276413 DOI: 10.3390/diagnostics14141481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Mechanical ventilation injures not only the lungs but also the diaphragm, resulting in dysfunction associated with poor outcomes. Diaphragm ultrasonography is a noninvasive, cost-effective, and reproducible diagnostic method used to monitor the condition and function of the diaphragm. With advances in ultrasound technology and the expansion of its clinical applications, diaphragm ultrasonography has become increasingly important as a tool to visualize and quantify diaphragmatic morphology and function across multiple medical specialties, including pulmonology, critical care, and rehabilitation medicine. This comprehensive review aims to provide an in-depth analysis of the role and limitations of ultrasonography in assessing the diaphragm, especially among critically ill patients. Furthermore, we discuss a recently published expert consensus and provide a perspective for the future.
Collapse
Affiliation(s)
- Taiga Itagaki
- Department of Emergency and Disaster Medicine, Tokushima University Hospital, 2-50-1 Kuramoto, Tokushima 770-8503, Japan
| | - Yusuke Akimoto
- Emergency Department, Tokushima Prefectural Miyoshi Hospital, 815-2 Ikedacho Shima, Miyoshi 778-0005, Japan;
| | - Takuya Takashima
- Department of Emergency and Critical Care Medicine, Tokushima University Graduate Hospital of Biomedical Sciences, 3-18-15 Kuramoto, Tokushima 770-8503, Japan; (T.T.); (J.O.)
| | - Jun Oto
- Department of Emergency and Critical Care Medicine, Tokushima University Graduate Hospital of Biomedical Sciences, 3-18-15 Kuramoto, Tokushima 770-8503, Japan; (T.T.); (J.O.)
| |
Collapse
|
13
|
Adrish M, Doppalapudi S, Lvovsky D. Driving pressure decoded: Precision strategies in adult respiratory distress syndrome management. World J Crit Care Med 2024; 13:92441. [PMID: 38855266 PMCID: PMC11155505 DOI: 10.5492/wjccm.v13.i2.92441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/17/2024] [Accepted: 04/24/2024] [Indexed: 06/03/2024] Open
Abstract
Mechanical ventilation (MV) is an important strategy for improving the survival of patients with respiratory failure. However, MV is associated with aggravation of lung injury, with ventilator-induced lung injury (VILI) becoming a major concern. Thus, ventilation protection strategies have been developed to minimize complications from MV, with the goal of relieving excessive breathing workload, improving gas exchange, and minimizing VILI. By opting for lower tidal volumes, clinicians seek to strike a balance between providing adequate ventilation to support gas exchange and preventing overdistension of the alveoli, which can contribute to lung injury. Additionally, other factors play a role in optimizing lung protection during MV, including adequate positive end-expiratory pressure levels, to maintain alveolar recruitment and prevent atelectasis as well as careful consideration of plateau pressures to avoid excessive stress on the lung parenchyma.
Collapse
Affiliation(s)
- Muhammad Adrish
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, United States
| | - Sai Doppalapudi
- Department of Medicine, BronxCare Health System/Icahn School of Medicine at Mount Sinai, Bronx, NY 10467, United States
| | - Dmitry Lvovsky
- Department of Medicine, BronxCare Health System/Icahn School of Medicine at Mount Sinai, Bronx, NY 10467, United States
| |
Collapse
|
14
|
Gabela-Zuniga B, Shukla VC, Bobba C, Higuita-Castro N, Powell HM, Englert JA, Ghadiali SN. A Micro-scale Humanized Ventilator-on-a-Chip to Examine the Injurious Effects of Mechanical Ventilation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582200. [PMID: 38464068 PMCID: PMC10925162 DOI: 10.1101/2024.02.26.582200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Patients with compromised respiratory function frequently require mechanical ventilation to survive. Unfortunately, non-uniform ventilation of injured lungs generates complex mechanical forces that lead to ventilator induced lung injury (VILI). Although investigators have developed lung-on-a-chip systems to simulate normal respiration, modeling the complex mechanics of VILI as well as the subsequent recovery phase is a challenge. Here we present a novel humanized in vitro ventilator-on-a-chip (VOC) model of the lung microenvironment that simulates the different types of injurious forces generated in the lung during mechanical ventilation. We used transepithelial/endothelial electrical resistance (TEER) measurements to investigate how individual and simultaneous application of the different mechanical forces alters real-time changes in barrier integrity during and after injury. We find that compressive stress (i.e. barotrauma) does not significantly alter barrier integrity while over-distention (20% cyclic radial strain, volutrauma) results in decreased barrier integrity that quickly recovers upon removal of mechanical stress. Conversely, surface tension forces generated during airway reopening (atelectrauma), result in a rapid loss of barrier integrity with a delayed recovery relative to volutrauma. Simultaneous application of cyclic stretching (volutrauma) and airway reopening (atelectrauma), indicate that the surface tension forces associated with reopening fluid-occluded lung regions is the primary driver of barrier disruption. Thus, our novel VOC system can monitor the effects of different types of injurious forces on barrier disruption and recovery in real-time and can be used to identify the biomechanical mechanisms of VILI.
Collapse
|
15
|
Spinelli E, Damia A, Damarco F, Gregori B, Occhipinti F, Busani Z, Leali M, Battistin M, Lonati C, Zhao Z, Storaci AM, Lopez G, Vaira V, Ferrero S, Rosso L, Gatti S, Mauri T. Pathophysiological profile of non-ventilated lung injury in healthy female pigs undergoing mechanical ventilation. COMMUNICATIONS MEDICINE 2024; 4:18. [PMID: 38361130 PMCID: PMC10869686 DOI: 10.1038/s43856-024-00449-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/01/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND Lung regions excluded from mechanical insufflation are traditionally assumed to be spared from ventilation-associated lung injury. However, preliminary data showed activation of potential mechanisms of injury within these non-ventilated regions (e.g., hypoperfusion, inflammation). METHODS In the present study, we hypothesized that non-ventilated lung injury (NVLI) may develop within 24 h of unilateral mechanical ventilation in previously healthy pigs, and we performed extended pathophysiological measures to profile NVLI. We included two experimental groups undergoing exclusion of the left lung from the ventilation with two different tidal volumes (15 vs 7.5 ml/kg) and a control group on bilateral ventilation. Pathophysiological alteration including lung collapse, changes in lung perfusion, lung stress and inflammation were measured. Lung injury was quantified by histological score. RESULTS Histological injury score of the non-ventilated lung is significantly higher than normally expanded lung from control animals. The histological score showed lower intermediate values (but still higher than controls) when the tidal volume distending the ventilated lung was reduced by 50%. Main pathophysiological alterations associated with NVLI were: extensive lung collapse; very low pulmonary perfusion; high inspiratory airways pressure; and higher concentrations of acute-phase inflammatory cytokines IL-6, IL-1β and TNF-α and of Angiopoietin-2 (a marker of endothelial activation) in the broncho-alveolar lavage. Only the last two alterations were mitigated by reducing tidal volume, potentially explaining partial protection. CONCLUSIONS Non-ventilated lung injury develops within 24 h of controlled mechanical ventilation due to multiple pathophysiological alterations, which are only partially prevented by low tidal volume.
Collapse
Grants
- This study was supported, in part, by Current Research from the Italian Ministry of Health, Rome, Italy; by EuroELSO Research grant 2021; by the “Hub Life Science-Diagnostica Avanzata (HLS-DA), PNC-E3-2022-23683266-CUP: C43C22001630001/MI-0117” Project from the Italian Ministry of Health (Piano Nazionale Complementare Ecosistema Innovativo della Salute), Rome, Italy; by the “Dipartimenti di Eccellenza Program 2023–2027” to the Dept. of Pathophysiology and Transplantation, University of Milan, from The Italian Ministry of Education and Research (MUR), Rome, Italy.
Collapse
Affiliation(s)
- Elena Spinelli
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Anna Damia
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Francesco Damarco
- Division of Thoracic Surgery and Lung Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Beatrice Gregori
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Federica Occhipinti
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Zara Busani
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Marco Leali
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Michele Battistin
- Center for Preclinical Research, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Caterina Lonati
- Center for Preclinical Research, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Zhanqi Zhao
- Furtwangen University, Institute of Technical Medicine, Villingen-Schwenningen, Germany
| | - Alessandra Maria Storaci
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gianluca Lopez
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Vaira
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Ferrero
- Division of Pathology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Biomedical Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Lorenzo Rosso
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Division of Thoracic Surgery and Lung Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Gatti
- Center for Preclinical Research, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Tommaso Mauri
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.
| |
Collapse
|
16
|
Ghazala Z, Ararat E, Carroll JL, Agarwal A. Volume-targeted ventilation as an invasive home ventilation strategy: A single-center experience. Pediatr Pulmonol 2024; 59:203-209. [PMID: 37861362 DOI: 10.1002/ppul.26732] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 10/06/2023] [Accepted: 10/12/2023] [Indexed: 10/21/2023]
Affiliation(s)
- Zena Ghazala
- Department of Pediatrics, Pediatric Pulmonary Division, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Erhan Ararat
- Department of Pediatrics, Pediatric Pulmonary Division, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - John L Carroll
- Department of Pediatrics, Pediatric Pulmonary Division, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Amit Agarwal
- Department of Pediatrics, Pediatric Pulmonary Division, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
17
|
Phoophiboon V, Owattanapanich N, Owattanapanich W, Schellenberg M. Effects of prone positioning on ARDS outcomes of trauma and surgical patients: a systematic review and meta-analysis. BMC Pulm Med 2023; 23:504. [PMID: 38093216 PMCID: PMC10716936 DOI: 10.1186/s12890-023-02805-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/05/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Prone position is an option for rescue therapy for acute respiratory distress syndrome. However, there are limited relevant data among trauma and surgical patients, who may be at increased risk for complications following position changes. This study aimed to identify the benefits and risks of proning in this patient subgroup. METHODS Follow the PRISMA 2020, MEDLINE and EMBASE database searches were conducted. Additional search of relevant primary literature and review articles was also performed. A random effects model was used to estimate the PF ratio, mortality rate, mechanical ventilator days, and intensive care unit length of stay using Review Manager 5.4.1 software. RESULTS Of 1,128 studies, 15 articles were included in this meta-analysis. The prone position significantly improved the PF ratio compared with the supine position (mean difference, 79.26; 95% CI, 53.38 to 105.13). The prone position group had a statistically significant mortality benefit (risk ratio [RR], 0.48; 95% CI, 0.35 to 0.67). Although there was no significant difference in the intensive care unit length of stay, the prone position significantly decreased mechanical ventilator days (-2.59; 95% CI, -4.21 to -0.97). On systematic review, minor complications were frequent, especially facial edema. There were no differences in local wound complications. CONCLUSIONS The prone position has comparable complications to the supine position. With its benefits of increased oxygenation and decreased mortality, the prone position can be considered for trauma and surgical patients. A prospective multicenter study is warranted.
Collapse
Affiliation(s)
- Vorakamol Phoophiboon
- Division of Critical Care Medicine, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Excellence Center for Critical Care Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Department of Critical Care Medicine, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, Canada
| | - Natthida Owattanapanich
- Division of Trauma Surgery, Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| | - Weerapat Owattanapanich
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Morgan Schellenberg
- Trauma and Surgical Critical Care, LAC+USC Medical Center, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
18
|
Buonanno P, Marra A, Iacovazzo C, Vargas M, Coviello A, Squillacioti F, Nappi S, de Siena AU, Servillo G. Impact of ventilation strategies on pulmonary and cardiovascular complications in patients undergoing general anaesthesia for elective surgery: a systematic review and meta-analysis. Br J Anaesth 2023; 131:1093-1101. [PMID: 37839932 PMCID: PMC10687618 DOI: 10.1016/j.bja.2023.09.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/10/2023] [Accepted: 09/11/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUND Many RCTs have evaluated the influence of intraoperative tidal volume (tV), PEEP, and driving pressure on the occurrence of postoperative pulmonary complications, cardiovascular complications, and mortality in adult patients. Our meta-analysis aimed to investigate the association between tV, PEEP, and driving pressure and the above-mentioned outcomes. METHODS We conducted a systematic review and meta-analysis of RCTs from inception to May 19, 2022. The primary outcome was the incidence of postoperative pulmonary complications; the secondary outcomes were intraoperative cardiovascular complications and 30-day mortality. Primary and secondary outcomes were evaluated stratifying patients in the following groups: (1) low tV (LV, tV 6-8 ml kg-1 and PEEP ≥5 cm H2O) vs high tV (HV, tV >8 ml kg-1 and PEEP=0 cm H2O); (2) higher PEEP (HP, ≥6 cm H2O) vs lower PEEP (LP, <6 cm H2O); and (3) driving pressure-guided PEEP (DP) vs fixed PEEP (FP). RESULTS We included 16 RCTs with a total sample size of 4993. The incidence of postoperative pulmonary complications was lower in patients treated with LV than with HV (OR=0.402, CI 0.280-0.577, P<0.001) and lower in DP than in FP group (OR=0.358, CI 0.187-0.684, P=0.002). Postoperative pulmonary complications did not differ between HP and LP groups; the incidence of intraoperative cardiovascular complications was higher in HP group (OR=1.385, CI 1.027-1.867, P=0.002). The 30-day mortality was not influenced by the ventilation strategy. CONCLUSIONS Optimal intraoperative mechanical ventilation is unclear; however, our meta-analysis showed that low tidal volume and driving pressure-guided PEEP strategies were associated with a reduction in postoperative pulmonary complications.
Collapse
Affiliation(s)
- Pasquale Buonanno
- Department of Neuroscience, Reproductive Science and Odontostomatological Science, University of Naples 'Federico II', Naples, Italy.
| | - Annachiara Marra
- Department of Neuroscience, Reproductive Science and Odontostomatological Science, University of Naples 'Federico II', Naples, Italy
| | - Carmine Iacovazzo
- Department of Neuroscience, Reproductive Science and Odontostomatological Science, University of Naples 'Federico II', Naples, Italy
| | - Maria Vargas
- Department of Neuroscience, Reproductive Science and Odontostomatological Science, University of Naples 'Federico II', Naples, Italy
| | - Antonio Coviello
- Department of Neuroscience, Reproductive Science and Odontostomatological Science, University of Naples 'Federico II', Naples, Italy
| | - Francesco Squillacioti
- Department of Neuroscience, Reproductive Science and Odontostomatological Science, University of Naples 'Federico II', Naples, Italy
| | - Serena Nappi
- Department of Neuroscience, Reproductive Science and Odontostomatological Science, University of Naples 'Federico II', Naples, Italy
| | - Andrea Uriel de Siena
- Department of Neuroscience, Reproductive Science and Odontostomatological Science, University of Naples 'Federico II', Naples, Italy
| | - Giuseppe Servillo
- Department of Neuroscience, Reproductive Science and Odontostomatological Science, University of Naples 'Federico II', Naples, Italy
| |
Collapse
|
19
|
Hardt K, Wappler F. Anesthesia for Morbidly Obese Patients. DEUTSCHES ARZTEBLATT INTERNATIONAL 2023; 120:779-785. [PMID: 37874129 PMCID: PMC10762842 DOI: 10.3238/arztebl.m2023.0216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 09/27/2023] [Accepted: 09/27/2023] [Indexed: 10/25/2023]
Abstract
BACKGROUND The prevalence of morbid obesity (BMI >35 kg/m2) has risen steadily in recent decades. With the corresponding rise in the number of bariatric operations, anesthesiologists deal with this patient group more commonly than before, particularly in specialized centers. METHODS This review is based on publications retrieved by a selective search in PubMed, including current guidelines and recommendations issued by specialist societies, as well as expert opinion. RESULTS In the anesthesiological care of morbidly obese patients, a preoperative assessment and risk stratification are just as important as the thoughtful selection of the anesthesia technique, the drugs used and their dosage, and perioperative management. A thorough understanding of the pathophysiological changes and comorbidities of morbid obesity and the associated risks is essential. The risk of pulmonary complications such as respiratory failure, hypoxia, and apnea is markedly higher in morbidly obese patients, especially those with obstructive sleep apnea. Short-acting, less lipophilic anesthetic drugs are particularly useful, as is multimodal pain therapy for the avoidance of high opiate doses. The indication for intensified postoperative monitoring depends on the patient's preexisting illnesses, the type of anesthesia, and the type of surgical procedure. Regional anesthetic techniques should be used if possible. CONCLUSION The perioperative care of morbidly obese patients presents special challenges. The anesthesiologist must be aware of potential comorbidities, specific risks, and pathophysiological changes in order to provide adequate care to this patient group.
Collapse
Affiliation(s)
- Katharina Hardt
- University Hospital of the Witten/Herdecke University – Cologne, Department of Anesthesiology and Operative Intensive Care Medicine, Cologne-Merheim Hospital
| | - Frank Wappler
- University Hospital of the Witten/Herdecke University – Cologne, Department of Anesthesiology and Operative Intensive Care Medicine, Cologne-Merheim Hospital
| |
Collapse
|
20
|
Shah N, Katira BH. Role of cardiopulmonary interactions in development of ventilator-induced lung injury-Experimental evidence and clinical Implications. Front Physiol 2023; 14:1228476. [PMID: 37534365 PMCID: PMC10391157 DOI: 10.3389/fphys.2023.1228476] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/07/2023] [Indexed: 08/04/2023] Open
Abstract
Ventilator-induced lung injury (VILI) impacts outcomes in ARDS and optimization of ventilatory strategies improves survival. Decades of research has identified various mechanisms of VILI, largely focusing on airspace forces of plateau pressure, tidal volume and driving pressure. Experimental evidence indicates the role of adverse cardiopulmonary interaction during mechanical ventilation, contributing to VILI genesis mostly by modulating pulmonary vascular dynamics. Under passive mechanical ventilation, high transpulmonary pressure increases afterload on right heart while high pleural pressure reduces the RV preload. Together, they can result in swings of pulmonary vascular flow and pressure. Altered vascular flow and pressure result in increased vascular shearing and wall tension, in turn causing direct microvascular injury accompanied with permeability to water, proteins and cells. Moreover, abrupt decreases in airway pressure, may result in sudden overperfusion of the lung and result in similar microvascular injury, especially when the endothelium is stretched or primed at high positive end-expiratory pressure. Microvascular injury is universal in VILI models and presumed in the diagnosis of ARDS; preventing such microvascular injury can reduce VILI and impact outcomes in ARDS. Consequently, developing cardiovascular targets to reduce macro and microvascular stressors in the pulmonary circulation can potentially reduce VILI. This paper reviews the role of cardiopulmonary interaction in VILI genesis.
Collapse
|
21
|
Schranc Á, Diaper J, Südy R, Peták F, Habre W, Albu G. Lung recruitment by continuous negative extra-thoracic pressure support following one-lung ventilation: an experimental study. Front Physiol 2023; 14:1160731. [PMID: 37256073 PMCID: PMC10225513 DOI: 10.3389/fphys.2023.1160731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/03/2023] [Indexed: 06/01/2023] Open
Abstract
Lung recruitment maneuvers following one-lung ventilation (OLV) increase the risk for the development of acute lung injury. The application of continuous negative extrathoracic pressure (CNEP) is gaining interest both in intubated and non-intubated patients. However, there is still a lack of knowledge on the ability of CNEP support to recruit whole lung atelectasis following OLV. We investigated the effects of CNEP following OLV on lung expansion, gas exchange, and hemodynamics. Ten pigs were anesthetized and mechanically ventilated with pressure-regulated volume control mode (PRVC; FiO2: 0.5, Fr: 30-35/min, VT: 7 mL/kg, PEEP: 5 cmH2O) for 1 hour, then baseline (BL) data for gas exchange (arterial partial pressure of oxygen, PaO2; and carbon dioxide, PaCO2), ventilation and hemodynamical parameters and lung aeration by electrical impedance tomography were recorded. Subsequently, an endobronchial blocker was inserted, and OLV was applied with a reduced VT of 5 mL/kg. Following a new set of measurements after 1 h of OLV, two-lung ventilation was re-established, combining PRVC (VT: 7 mL/kg) and CNEP (-15 cmH2O) without any hyperinflation maneuver and data collection was then repeated at 5 min and 1 h. Compared to OLV, significant increases in PaO2 (154.1 ± 13.3 vs. 173.8 ± 22.1) and decreases in PaCO2 (52.6 ± 11.7 vs. 40.3 ± 4.5 mmHg, p < 0.05 for both) were observed 5 minutes following initiation of CNEP, and these benefits in gas exchange remained after an hour of CNEP. Gradual improvements in lung aeration in the non-collapsed lung were also detected by electrical impedance tomography (p < 0.05) after 5 and 60 min of CNEP. Hemodynamics and ventilation parameters remained stable under CNEP. Application of CNEP in the presence of whole lung atelectasis proved to be efficient in improving gas exchange via recruiting the lung without excessive airway pressures. These benefits of combined CNEP and positive pressure ventilation may have particular value in relieving atelectasis in the postoperative period of surgical procedures requiring OLV.
Collapse
Affiliation(s)
- Álmos Schranc
- Unit for Anesthesiological Investigations, Department of Anesthesiology Pharmacology, Intensive Care and Emergency Medicine, University of Geneva, Geneva, Switzerland
| | - John Diaper
- Unit for Anesthesiological Investigations, Department of Anesthesiology Pharmacology, Intensive Care and Emergency Medicine, University of Geneva, Geneva, Switzerland
| | - Roberta Südy
- Unit for Anesthesiological Investigations, Department of Anesthesiology Pharmacology, Intensive Care and Emergency Medicine, University of Geneva, Geneva, Switzerland
| | - Ferenc Peták
- Department of Medical Physics and Informatics, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Walid Habre
- Unit for Anesthesiological Investigations, Department of Anesthesiology Pharmacology, Intensive Care and Emergency Medicine, University of Geneva, Geneva, Switzerland
| | - Gergely Albu
- Unit for Anesthesiological Investigations, Department of Anesthesiology Pharmacology, Intensive Care and Emergency Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
22
|
Santana PV, Cardenas LZ, de Albuquerque ALP. Diaphragm Ultrasound in Critically Ill Patients on Mechanical Ventilation—Evolving Concepts. Diagnostics (Basel) 2023; 13:diagnostics13061116. [PMID: 36980423 PMCID: PMC10046995 DOI: 10.3390/diagnostics13061116] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/11/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Mechanical ventilation (MV) is a life-saving respiratory support therapy, but MV can lead to diaphragm muscle injury (myotrauma) and induce diaphragmatic dysfunction (DD). DD is relevant because it is highly prevalent and associated with significant adverse outcomes, including prolonged ventilation, weaning failures, and mortality. The main mechanisms involved in the occurrence of myotrauma are associated with inadequate MV support in adapting to the patient’s respiratory effort (over- and under-assistance) and as a result of patient-ventilator asynchrony (PVA). The recognition of these mechanisms associated with myotrauma forced the development of myotrauma prevention strategies (MV with diaphragm protection), mainly based on titration of appropriate levels of inspiratory effort (to avoid over- and under-assistance) and to avoid PVA. Protecting the diaphragm during MV therefore requires the use of tools to monitor diaphragmatic effort and detect PVA. Diaphragm ultrasound is a non-invasive technique that can be used to monitor diaphragm function, to assess PVA, and potentially help to define diaphragmatic effort with protective ventilation. This review aims to provide clinicians with an overview of the relevance of DD and the main mechanisms underlying myotrauma, as well as the most current strategies aimed at minimizing the occurrence of myotrauma with special emphasis on the role of ultrasound in monitoring diaphragm function.
Collapse
Affiliation(s)
- Pauliane Vieira Santana
- Intensive Care Unit, AC Camargo Cancer Center, São Paulo 01509-011, Brazil
- Correspondence: (P.V.S.); (A.L.P.d.A.)
| | - Letícia Zumpano Cardenas
- Intensive Care Unit, Physical Therapy Department, AC Camargo Cancer Center, São Paulo 01509-011, Brazil
| | - Andre Luis Pereira de Albuquerque
- Pulmonary Division, Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-000, Brazil
- Sírio-Libanês Teaching and Research Institute, Hospital Sírio Libanês, São Paulo 01308-060, Brazil
- Correspondence: (P.V.S.); (A.L.P.d.A.)
| |
Collapse
|
23
|
Vallabh P, Ha M, Ahern K. Efficacy and Safety of Cisatracurium Compared to Vecuronium for Neuromuscular Blockade in Acute Respiratory Distress Syndrome. J Intensive Care Med 2023; 38:188-195. [PMID: 35821572 DOI: 10.1177/08850666221113504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE Previous studies analyzing neuromuscular blocking agents (NMBAs) in acute respiratory distress syndrome (ARDS) have evaluated the benefit of cisatracurium with conflicting results, and data evaluating other NMBAs remains limited. The objective of this study was to compare the efficacy and safety of cisatracurium to vecuronium in ARDS. MATERIALS AND METHODS A single-center, retrospective, propensity matched review of patients who received cisatracurium or vecuronium continuous infusions between October 1, 2017 and June 30, 2020 for ARDS was conducted. The primary endpoint was duration of mechanical ventilation. Secondary endpoints included change in PaO2/FiO2 ratio at 48 h, intensive care unit (ICU) and hospital mortality, and ICU and hospital length of stay (LOS). Safety endpoints included newly developed myopathy, presence of bradycardia or hypotension, and newly developed barotrauma or volutrauma. RESULTS Twenty-nine patients were included in each group. There was no statistically significant difference in the primary endpoint of ventilator days between cisatracurium and vecuronium groups (mean 15.9 vs. 20.5 days respectively; p = .2). No statistically significant differences were found in secondary endpoints of ICU mortality (51.7% vs. 51.7%) or length of stay (18.7 vs. 23.9 days, p = .19), hospital mortality (51.7% vs. 55.2%, p = .79) or length of stay (22 vs. 30.6 days, p = .08), or mean change in PaO2/FiO2 (29.8 vs. 36.6; p = .74). Statistically significant differences were not observed in safety endpoints of myopathy (37.9% vs. 37.9%), barotrauma or volutrauma (13.8% vs. 3.5%; p = .16), bradycardia (31% vs. 13.8%; p = .12), or hypotension (96.6% vs. 82.8%; p = .08). CONCLUSIONS No significant differences were seen in efficacy or safety endpoints between cisatracurium or vecuronium groups, suggesting that vecuronium may be a safe alternative agent for neuromuscular blockade in ARDS. Results of this analysis warrant confirmation in a larger, randomized study.
Collapse
Affiliation(s)
- Priya Vallabh
- Department of Pharmacy, 41528UMass Memorial Medical Center, Worcester, MA, USA
| | - Michael Ha
- Department of Pharmacy, 41528UMass Memorial Medical Center, Worcester, MA, USA
| | - Krystina Ahern
- Department of Pharmacy, 41528UMass Memorial Medical Center, Worcester, MA, USA
| |
Collapse
|
24
|
Significance of Pulmonary Endothelial Injury and the Role of Cyclooxygenase-2 and Prostanoid Signaling. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010117. [PMID: 36671689 PMCID: PMC9855370 DOI: 10.3390/bioengineering10010117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
The endothelium plays a key role in the dynamic balance of hemodynamic, humoral and inflammatory processes in the human body. Its central importance and the resulting therapeutic concepts are the subject of ongoing research efforts and form the basis for the treatment of numerous diseases. The pulmonary endothelium is an essential component for the gas exchange in humans. Pulmonary endothelial dysfunction has serious consequences for the oxygenation and the gas exchange in humans with the potential of consecutive multiple organ failure. Therefore, in this review, the dysfunction of the pulmonary endothel due to viral, bacterial, and fungal infections, ventilator-related injury, and aspiration is presented in a medical context. Selected aspects of the interaction of endothelial cells with primarily alveolar macrophages are reviewed in more detail. Elucidation of underlying causes and mechanisms of damage and repair may lead to new therapeutic approaches. Specific emphasis is placed on the processes leading to the induction of cyclooxygenase-2 and downstream prostanoid-based signaling pathways associated with this enzyme.
Collapse
|
25
|
Brandly JE, Midon M, Douglas HF, Hopster K. Flow-controlled expiration reduces positive end-expiratory pressure requirement in dorsally recumbent, anesthetized horses. Front Vet Sci 2023; 10:1135452. [PMID: 37124564 PMCID: PMC10140341 DOI: 10.3389/fvets.2023.1135452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/23/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction Equine peri-anesthetic mortality is higher than that for other commonly anesthetized veterinary species. Unique equine pulmonary pathophysiologic aspects are believed to contribute to this mortality due to impairment of gas exchange and subsequent hypoxemia. No consistently reliable solution for the treatment of peri-anesthetic gas exchange impairment is available. Flow-controlled expiration (FLEX) is a ventilatory mode that linearizes gas flow throughout the expiratory phase, reducing the rate of lung emptying and alveolar collapse. FLEX has been shown to improve gas exchange and pulmonary mechanics in anesthetized horses. This study further evaluated FLEX ventilation in anesthetized horses positioned in dorsal recumbency, hypothesizing that after alveolar recruitment, horses ventilated using FLEX would require a lower positive end-expiratory pressure (PEEP) to prevent alveolar closure than horses conventionally ventilated. Methods Twelve adult horses were used in this prospective, randomized study. Horses were assigned either to conventional volume-controlled ventilation (VCV) or to FLEX. Following induction of general anesthesia, horses were placed in dorsal recumbency mechanically ventilated for a total of approximately 6.5 hours. Thirty-minutes after starting ventilation with VCV or FLEX, a PEEP-titration alveolar recruitment maneuver was performed at the end of which the PEEP was reduced in decrements of 3 cmH2O until the alveolar closure pressure was determined. The PEEP was then increased to the previous level and maintained for additional three hours. During this time, the mean arterial blood pressure, pulmonary arterial pressure, central venous blood pressure, cardiac output (CO), dynamic respiratory system compliance and arterial blood gas values were measured. Results The alveolar closure pressure was significantly lower (6.5 ± 1.2 vs 11.0 ± 1.5 cmH2O) and significantly less PEEP was required to prevent alveolar closure (9.5 ± 1.2 vs 14.0 ± 1.5 cmH2O) for horses ventilated using FLEX compared with VCV. The CO was significantly higher in the horses ventilated with FLEX (37.5 ± 4 vs 30 ± 6 l/min). Discussion We concluded that FLEX ventilation was associated with a lower PEEP requirement due to a more homogenous distribution of ventilation in the lungs during expiration. This lower PEEP requirement led to more stable and improved cardiovascular conditions in horses ventilated with FLEX.
Collapse
|
26
|
Itagaki T, Akimoto Y, Nakano Y, Ueno Y, Ishihara M, Tane N, Tsunano Y, Oto J. Relationships between double cycling and inspiratory effort with diaphragm thickness during the early phase of mechanical ventilation: A prospective observational study. PLoS One 2022; 17:e0273173. [PMID: 35976965 PMCID: PMC9385032 DOI: 10.1371/journal.pone.0273173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 08/03/2022] [Indexed: 11/23/2022] Open
Abstract
Background Increased and decreased diaphragm thickness during mechanical ventilation is associated with poor outcomes. Some types of patient-ventilator asynchrony theoretically cause myotrauma of the diaphragm. However, the effects of double cycling on structural changes in the diaphragm have not been previously evaluated. Hence, this study aimed to investigate the relationship between double cycling during the early phase of mechanical ventilation and changes in diaphragm thickness, and the involvement of inspiratory effort in the occurrence of double cycling. Methods We evaluated adult patients receiving invasive mechanical ventilation for more than 48 h. The end-expiratory diaphragm thickness (Tdiee) was assessed via ultrasonography on days 1, 2, 3, 5 and 7 after the initiation of mechanical ventilation. Then, the maximum rate of change from day 1 (ΔTdiee%) was evaluated. Concurrently, we recorded esophageal pressure and airway pressure on days 1, 2 and 3 for 1 h during spontaneous breathing. Then, the waveforms were retrospectively analyzed to calculate the incidence of double cycling (double cycling index) and inspiratory esophageal pressure swing (ΔPes). Finally, the correlation between double cycling index as well as ΔPes and ΔTdiee% was investigated using linear regression models. Results In total, 19 patients with a median age of 69 (interquartile range: 65–78) years were enrolled in this study, and all received pressure assist-control ventilation. The Tdiee increased by more than 10% from baseline in nine patients, decreased by more than 10% in nine and remained unchanged in one. The double cycling indexes on days 1, 2 and 3 were 2.2%, 1.3% and 4.5%, respectively. There was a linear correlation between the double cycling index on day 3 and ΔTdiee% (R2 = 0.446, p = 0.002). The double cycling index was correlated with the ΔPes on days 2 (R2 = 0.319, p = 0.004) and 3 (R2 = 0.635, p < 0.001). Conclusions Double cycling on the third day of mechanical ventilation was associated with strong inspiratory efforts and, possibly, changes in diaphragm thickness.
Collapse
Affiliation(s)
- Taiga Itagaki
- Department of Emergency and Disaster Medicine, Tokushima University Hospital, Tokushima, Japan
- * E-mail:
| | - Yusuke Akimoto
- Department of Emergency and Critical Care Medicine, Tokushima University Hospital, Tokushima, Japan
| | - Yuki Nakano
- Department of Emergency and Critical Care Medicine, Tokushima University Hospital, Tokushima, Japan
| | - Yoshitoyo Ueno
- Department of Emergency and Critical Care Medicine, Tokushima University Hospital, Tokushima, Japan
| | - Manabu Ishihara
- Department of Emergency and Critical Care Medicine, Tokushima University Hospital, Tokushima, Japan
| | - Natsuki Tane
- Department of Emergency and Critical Care Medicine, Tokushima University Graduate School, Tokushima, Japan
| | - Yumiko Tsunano
- Department of Emergency and Critical Care Medicine, Tokushima University Graduate School, Tokushima, Japan
| | - Jun Oto
- Department of Emergency and Critical Care Medicine, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
27
|
Wang L, Li J, Zhu Y, Zha B. Low tidal volume ventilation alleviates ventilator-induced lung injury by regulating the NLRP3 inflammasome. Exp Lung Res 2022; 48:168-177. [PMID: 35916505 DOI: 10.1080/01902148.2022.2104409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
PURPOSE Low tidal volume ventilation (LTVV) is a well-known ventilation mode which can improve ventilator-induced lung injury (VILI). However, the mechanism of LTVV ameliorating VILI has not yet been elucidated. In this study, we aimed to reveal LTVV protected against VILI by inhibiting the activation of the NLRP3 inflammasome in bronchoalveolar lavage fluid (BALF) from humans and lungs from mice. MATERIALS AND METHODS Twenty-eight patients scheduled for video-assisted thoracoscopic esophagectomy were randomized to receive high-tidal-volume ventilation [Vt = 10 mL/kg without positive end-expiratory pressure (PEEP)] or LTVV (Vt = 5 mL/kg along with 5 cm of H2O PEEP) during one-lung ventilation. BALF was collected before and at the end of surgery. Male C57BL/6 mice received high-tidal-volume ventilation, LTVV or MCC950 (an inhibitor of NLRP3). The activation of the formation of NLRP3 inflammasome in BALF from patients and in lungs from mice were analyzed. RESULTS LTTV decreased the peak airway pressure (Ppeak), plateau airway pressure (Pplat) and driving pressure (ΔP) during one-lung ventilation. Additionally, LTVV not only inhibited pulmonary infiltration and inflammation caused by mechanical ventilation, but also suppressed the NLRP3 inflammasome activation in BALF from humans. In mice, ventilator-induced inflammatory response and pulmonary edema were suppressed by LTVV with an efficacy comparable to that of MCC950 treatment. Furthermore, LTVV, similar to MCC950, clearly decreased ventilator-induced NLRP3 inflammasome activation. CONCLUSION Our study showed that LTVV played a protective role in ventilator-induced lung injury by suppressing the activation of the NLRP3 inflammasome. TRIAL REGISTRATION This study was registered in The Chinese Clinical Trial Registry, ChiCTR1900026190 on 25 September 2019.
Collapse
Affiliation(s)
- Lixia Wang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Jun Li
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Yan Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| | - Binshan Zha
- Department of Vascular and Thyroid Surgery, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
28
|
Chirico RN, de Matos NA, Castro TDF, Cândido LDS, Miranda AG, Costa GDP, Talvani A, Cangussú SD, Brochard L, Bezerra FS. The exogenous surfactant pre-treatment attenuates ventilator-induced lung injury in adult rats. Respir Physiol Neurobiol 2022; 302:103911. [PMID: 35430285 DOI: 10.1016/j.resp.2022.103911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 03/10/2022] [Accepted: 04/10/2022] [Indexed: 10/18/2022]
Abstract
Mechanical ventilation is an essential supportive therapy in the treatment of critical patients, and it aims to maintain adequate gas exchange; however, it can also contribute to inflammation and oxidative stress, thus leading to lung injury. We tested the hypothesis that exogenous surfactant administration will be protective against ventilator-induced lung injury in adult healthy Wistar rats both because of its anti-inflammatory properties as well as its role in preventing alveolar collapse at end-expiration. Thus, the effect of intranasal instillation of a bovine exogenous surfactant was tested in Wistar rats submitted to mechanical ventilation. The animals were divided into four groups: (1) CONTROL; (2) SURFACTANT; (3) Mechanical ventilation (MV); (4) MV with pre-treatment with surfactant (MVSURFACTANT). The MV and MVSURFACTANT were submitted to MV with high tidal volume (12 mL/kg) for 1 h. After the experimental protocol, all animals were euthanized and the arterial blood, bronchoalveolar lavage fluid and lungs were collected for biochemical, immunoenzymatic assay, arterial blood gases, and morphometric analyzes. The Wistar rats that received exogenous surfactant (Survanta®) by intranasal instillation before MV demonstrated reduced levels of leukocytes, inflammatory biomarkers such as CCL2, IL-1, IL-6 and TNF-α. Furthermore, it prevented oxidative damage by reducing lipid peroxidation and protein carbonylation as well as histological pattern changes of pulmonary parenchyma. Our data indicate that exogenous surfactant attenuated lung inflammation and redox imbalance induced by mechanical ventilation in healthy adult rats suggesting a preventive effect on ventilator-induced lung injury.
Collapse
Affiliation(s)
- Rafael Neto Chirico
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences, Center of Research in Biological Sciences, Federal University of Ouro Preto, 35400-000 Minas Gerais, Brazil
| | - Natália Alves de Matos
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences, Center of Research in Biological Sciences, Federal University of Ouro Preto, 35400-000 Minas Gerais, Brazil
| | - Thalles de Freitas Castro
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences, Center of Research in Biological Sciences, Federal University of Ouro Preto, 35400-000 Minas Gerais, Brazil
| | - Leandro da Silva Cândido
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences, Center of Research in Biological Sciences, Federal University of Ouro Preto, 35400-000 Minas Gerais, Brazil
| | - Amanda Gonçalves Miranda
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences, Center of Research in Biological Sciences, Federal University of Ouro Preto, 35400-000 Minas Gerais, Brazil
| | - Guilherme de Paula Costa
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences, Center of Research in Biological Sciences, Federal University of Ouro Preto, 35400-000 Minas Gerais, Brazil
| | - André Talvani
- Laboratory of Immunobiology of Inflammation (LABIIN), Department of Biological Sciences, Center of Research in Biological Sciences, Federal University of Ouro Preto, 35400-000 Minas Gerais, Brazil
| | - Sílvia Dantas Cangussú
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences, Center of Research in Biological Sciences, Federal University of Ouro Preto, 35400-000 Minas Gerais, Brazil
| | - Laurent Brochard
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto 416-360-4000, Ontario, Canada; Keenan Research Centre, Li KaShing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Frank Silva Bezerra
- Laboratory of Experimental Pathophysiology (LAFEx), Department of Biological Sciences, Center of Research in Biological Sciences, Federal University of Ouro Preto, 35400-000 Minas Gerais, Brazil; Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto 416-360-4000, Ontario, Canada; Keenan Research Centre, Li KaShing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
29
|
Idris AH. The Importance of Measuring Ventilation During Resuscitation. Resuscitation 2022; 177:41-42. [PMID: 35788022 DOI: 10.1016/j.resuscitation.2022.06.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 10/17/2022]
Affiliation(s)
- Ahamed H Idris
- Department of Emergency Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390-8579, United States.
| |
Collapse
|
30
|
Wang BC, Lion RP, Avesar M, Abdala J, Deming DD, Wilson CG. Comparison of Local and Systemic Inflammation During Invasive Versus Noninvasive Ventilation in Rats. J Interferon Cytokine Res 2022; 42:343-348. [PMID: 35704907 DOI: 10.1089/jir.2022.0040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The impact of noninvasive ventilation (NIV) on local and systemic inflammation is poorly characterized, particularly when compared with invasive mechanical ventilation (IMV). We sought to quantify the local and systemic inflammatory response of these 2 respiratory treatments in rats with lipopolysaccharide (LPS)-induced lung injury (LPS-injured) and healthy rats. Animals were subjected to 4 h of NIV or IMV treatments at noninjurious settings, or 4 h of control treatment in which healthy or LPS-injured animals remained spontaneously breathing under isoflurane anesthesia with no respiratory support. Cytokines were then quantified in the serum and lung tissue by multiplex enzyme-linked immunosorbent assay. Contrary to our hypothesis, there were no significant differences in cytokine levels in serum or lung when comparing the NIV- and IMV-treated groups; this was true in both LPS-injured and healthy rats. However, within the LPS-injured group, pulmonary levels of interleukin (IL)-1α, IL-6, and tumor necrosis factor α were significantly lower in the NIV-treated group than in control but not in the IMV-treated group compared with control. We conclude that NIV, unlike IMV, could attenuate local inflammation.
Collapse
Affiliation(s)
- Billy C Wang
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, OSF Children's Hospital of Illinois, Peoria, Illinois, USA
| | - Richard P Lion
- Department of Pediatrics, Texas Center for Pediatric and Congenital Heart Disease, UT Health Austin/Dell Children's Medical Center, University of Texas Dell Medical School, Austin, Texas, USA
| | - Michael Avesar
- Division of Pediatric Critical Care Medicine, Loma Linda University Children's Hospital, Loma Linda, California, USA
| | - Jonathan Abdala
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California, USA
| | - Douglas D Deming
- Division of Neonatology, Loma Linda University Children's Hospital, Loma Linda, California, USA
| | - Christopher G Wilson
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California, USA
| |
Collapse
|
31
|
Yang BY, Blackwood JE, Shin J, Guan S, Gao M, Jorgenson DB, Boehl JE, Sayre MR, Kudenchuk PJ, Rea TD, Kwok H, Johnson NJ. A pilot evaluation of respiratory mechanics during prehospital manual ventilation. Resuscitation 2022; 177:55-62. [PMID: 35690127 DOI: 10.1016/j.resuscitation.2022.06.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 05/27/2022] [Accepted: 06/01/2022] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Respiratory mechanics, such as tidal volume (VT) and inspiratory pressures, may affect outcome in hospitalized patients with respiratory failure. Little is known about respiratory mechanics in the prehospital setting. METHODS In this prospective, pilot investigation of patients receiving prehospital advanced airway placement, paramedics applied a device to measure respiratory mechanics. We evaluated tidal volume (VT) per predicted body weight (VTPBW) to determine the proportion of breaths within the lung-protective range of 4-10 mL/kg per PBW overall, according to ventilation bag volume (large versus small) and cardiac arrest status (active CPR, post-ROSC, non-arrest). RESULTS Over 16-months, 7371 post-intubation breaths were measured in 54 patients, 32 patients with cardiac arrest and 22 with other conditions. Paramedics ventilated 19 patients with a small bag and 35 patients with a large bag. Overall, mean VT was 435 mL (95% CI 403, 467); VTPBW was 7.0 mL/kg (95% CI 6.4, 7.6) with 75% within the lung-protective range. Mean VTPBW and peak pressure differed according to arrest status (absolute difference -0.36 mL/kg and 32 cmH2O for active CPR compared to post-ROSC), though not according to bag size. CONCLUSIONS We observed that measuring respiratory mechanics in the prehospital setting was feasible. Tidal volumes were generally delivered within a safe range. Respiratory mechanics varied most significantly with active CPR with lower VTPBW and higher peak pressures, though did not seem to be affected by bag size. Future work might examine the relationship between respiratory mechanics and outcomes, which may identify opportunities to improve clinical outcomes.
Collapse
Affiliation(s)
- Betty Y Yang
- Department of Emergency Medicine, University of Washington, Seattle, WA, United States.
| | - Jennifer E Blackwood
- Division of Emergency Medical Services, Public Health - Seattle & King County, Seattle, WA, United States
| | - Jenny Shin
- Division of Emergency Medical Services, Public Health - Seattle & King County, Seattle, WA, United States
| | - Sally Guan
- Division of Emergency Medical Services, Public Health - Seattle & King County, Seattle, WA, United States
| | - Mengqi Gao
- Philips Healthcare, Bothell, WA, United States
| | | | - James E Boehl
- Bellevue Fire Department, Bellevue, WA, United States
| | - Michael R Sayre
- Department of Emergency Medicine, University of Washington, Seattle, WA, United States
| | - Peter J Kudenchuk
- Division of Emergency Medical Services, Public Health - Seattle & King County, Seattle, WA, United States; Department of Medicine, Division of Cardiology, University of Washington, Seattle, WA, United States
| | - Thomas D Rea
- Division of Emergency Medical Services, Public Health - Seattle & King County, Seattle, WA, United States; Department of Medicine, Division of General Medicine, University of Washington, Seattle, WA, United States
| | - Heemun Kwok
- Department of Emergency Medicine, University of Washington, Seattle, WA, United States
| | - Nicholas J Johnson
- Department of Emergency Medicine, University of Washington, Seattle, WA, United States; Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
32
|
NISHASH: A reasonable cost-effective mechanical ventilator for COVID affected patients in Bangladesh. Heliyon 2022; 8:e09400. [PMID: 35578638 PMCID: PMC9093062 DOI: 10.1016/j.heliyon.2022.e09400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/19/2022] [Accepted: 05/05/2022] [Indexed: 11/23/2022] Open
Abstract
COVID-19 has elapsed all over the world with massive losses which indicate the lack of availability of medical equipment during the pandemic such as a ventilator. This is exemplified by the densely populated country Bangladesh who unable to maintain COVID-affected people because of the ventilator. Due to the higher price, unavailability, and manufacturing defection, most medical are unable to purchase this ventilator which causes terrible death for a respiratory problem. Of these cases, this paper represents a way to escape this problem and proposed a mechanical ventilator named “NISHASH” which will help to anticipate COVID affected people and higher price of the ventilator. Through the electromechanical instruments, a prototype lightweight easily moveable where preciously it automatically controls with digital feedback system ventilator which fulfills oxygen flow based on patient requirement are developed with different selection mode. The aim was to design and develop inexpensively automated easy to build to minimize the extreme shortage of the ventilator in Bangladesh. In this model of a mechanical ventilator, the cost is less than $90 where components are available all over the world.
Collapse
|
33
|
Miller AG, Tan HL, Smith BJ, Rotta AT, Lee JH. The Physiological Basis of High-Frequency Oscillatory Ventilation and Current Evidence in Adults and Children: A Narrative Review. Front Physiol 2022; 13:813478. [PMID: 35557962 PMCID: PMC9087180 DOI: 10.3389/fphys.2022.813478] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 04/08/2022] [Indexed: 12/12/2022] Open
Abstract
High-frequency oscillatory ventilation (HFOV) is a type of invasive mechanical ventilation that employs supra-physiologic respiratory rates and low tidal volumes (VT) that approximate the anatomic deadspace. During HFOV, mean airway pressure is set and gas is then displaced towards and away from the patient through a piston. Carbon dioxide (CO2) is cleared based on the power (amplitude) setting and frequency, with lower frequencies resulting in higher VT and CO2 clearance. Airway pressure amplitude is significantly attenuated throughout the respiratory system and mechanical strain and stress on the alveoli are theoretically minimized. HFOV has been purported as a form of lung protective ventilation that minimizes volutrauma, atelectrauma, and biotrauma. Following two large randomized controlled trials showing no benefit and harm, respectively, HFOV has largely been abandoned in adults with ARDS. A multi-center clinical trial in children is ongoing. This article aims to review the physiologic rationale for the use of HFOV in patients with acute respiratory failure, summarize relevant bench and animal models, and discuss the potential use of HFOV as a primary and rescue mode in adults and children with severe respiratory failure.
Collapse
Affiliation(s)
- Andrew G Miller
- Duke University Medical Center, Respiratory Care Services, Durham, NC, United States
| | - Herng Lee Tan
- KK Women's and Children's Hospital, Children's Intensive Care Unit, Singapore, Singapore
| | - Brian J Smith
- University of California, Davis, Respiratory Care Services, Sacramento, CA, United States
| | - Alexandre T Rotta
- Duke University Medical Center, Division of Pediatric Critical Care Medicine, Durham, NC, United States
| | - Jan Hau Lee
- KK Women's and Children's Hospital, Children's Intensive Care Unit, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
34
|
Cercos-Pita JL, Fardin L, Leclerc H, Maury B, Perchiazzi G, Bravin A, Bayat S. Lung tissue biomechanics imaged with synchrotron phase contrast microtomography in live rats. Sci Rep 2022; 12:5056. [PMID: 35322152 PMCID: PMC8942151 DOI: 10.1038/s41598-022-09052-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 03/09/2022] [Indexed: 12/19/2022] Open
Abstract
The magnitude and distribution of strain imposed on the peripheral airspaces by mechanical ventilation at the microscopic level and the consequent deformations are unknown despite their importance for understanding the mechanisms occurring at the onset of ventilator-induced lung injury. Here a 4-Dimensional (3D + time) image acquisition and processing technique is developed to assess pulmonary acinar biomechanics at microscopic resolution. Synchrotron radiation phase contrast CT with an isotropic voxel size of 6 µm3 is applied in live anesthetized rats under controlled mechanical ventilation. Video animations of regional acinar and vascular strain are acquired in vivo. Maps of strain distribution due to positive-pressure breaths and cardiovascular activity in lung acini and blood vessels are derived based on CT images. Regional strain within the lung peripheral airspaces takes average values of 0.09 ± 0.02. Fitting the expression S = kVn, to the changes in peripheral airspace area (S) and volume (V) during a positive pressure breath yields an exponent n = 0.82 ± 0.03, suggesting predominant alveolar expansion rather than ductal expansion or alveolar recruitment. We conclude that this methodology can be used to assess acinar conformational changes during positive pressure breaths in intact peripheral lung airspaces.
Collapse
Affiliation(s)
- Jose-Luis Cercos-Pita
- Hedenstierna Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Luca Fardin
- European Synchrotron Radiation Facility, Grenoble, France
| | - Hugo Leclerc
- Laboratoire de Mathématiques d'Orsay, Université Paris-Saclay, Orsay, France
| | - Bertrand Maury
- Département de Mathématiques Appliquées, Ecole Normale Supérieure, Université PSL, Paris, France
| | - Gaetano Perchiazzi
- Hedenstierna Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Alberto Bravin
- Physics Department, Milano Bicocca University, Milan, Italy
| | - Sam Bayat
- Synchrotron Radiation for Biomedicine STROBE Inserm UA07, Univ. Grenoble Alpes, Grenoble, France.
- Univ. Grenoble Alpes - Inserm UA07, Synchrotron Radiation for Biomedicine (STROBE) Laboratory, 2280 Rue de la Piscine, 38400, Grenoble, France.
| |
Collapse
|
35
|
Kawai M, Zhang E, Kabwe JC, Okada A, Maruyama J, Sawada H, Maruyama K. Lung damage created by high tidal volume ventilation in rats with monocrotaline-induced pulmonary hypertension. BMC Pulm Med 2022; 22:78. [PMID: 35247989 PMCID: PMC8897872 DOI: 10.1186/s12890-022-01867-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 02/17/2022] [Indexed: 11/10/2022] Open
Abstract
Background Rats with chronic hypoxia-induced non-inflammatory pulmonary hypertension (PH) are resistant to ventilator-induced lung injury. We investigated the effect of high tidal volume ventilation in another model of PH, monocrotaline (MCT)-induced PH, which is a type of inflammatory PH.
Methods PH was induced in rats by subcutaneous injection with 60 mg/kg MCT. Normal control rats, rats at 2 weeks after MCT injection (MCT2), and rats at 3 weeks after MCT injection (MCT3) were ventilated with low tidal volume (LV, 6 mL/kg) or high tidal volume (HV, 35 mL/kg) for 2 h with room air without positive end-expiratory pressure. Arterial oxygen pressure (PaO2) and Evans blue dye (EBD) extravasation were measured. Hypertensive pulmonary vascular remodeling was assessed morphometrically by the percentage of muscularized peripheral pulmonary arteries (%Muscularization) and the media wall thickness to external diameter ratio, namely percentage medial wall thickness (%MWT). To assess inflammation, lung IκB protein and cytokine mRNA expression levels were assessed. Results Baseline mean pulmonary arterial pressure was significantly higher in MCT rats (normal, 15.4 ± 0.5 mmHg; MCT2, 23.7 ± 0.9; and MCT3, 34.5 ± 1.5). After 2-h ventilation, PaO2 was significantly lower in the HV groups compared with the LV groups in normal and MCT2 rats, but not in MCT3 rats. Impairment of oxygenation with HV was less in MCT3 rats compared with normal and MCT2 rats. Among the HV groups, MCT3 rats showed significantly lower levels of EBD extravasation than normal and MCT2 rats. HV significantly downregulated IκB protein expression in normal and MCT3 rats and increased IL-6, MCP-1, CXCL-1 (MIP-1), and IL-10 mRNA levels in MCT3 rats. %Muscularization, %MWT, and the expression of lung elastin were significantly higher in MCT3 rats than in normal and MCT2 rats. Conclusion We found that HV-associated damage might be reduced in MCT-induced PH rats compared with normal rats. The results of this and earlier studies suggest that hypertensive pulmonary vascular structural changes might be protective against the occurrence of ventilator-induced lung injury, irrespective of the etiology of PH. Supplementary Information The online version contains supplementary material available at 10.1186/s12890-022-01867-6.
Collapse
|
36
|
Zhang Y, Zhang J, Fu Z. Molecular hydrogen is a potential protective agent in the management of acute lung injury. Mol Med 2022; 28:27. [PMID: 35240982 PMCID: PMC8892414 DOI: 10.1186/s10020-022-00455-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/14/2022] [Indexed: 11/21/2022] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome, which is a more severe form of ALI, are life-threatening clinical syndromes observed in critically ill patients. Treatment methods to alleviate the pathogenesis of ALI have improved to a great extent at present. Although the efficacy of these therapies is limited, their relevance has increased remarkably with the ongoing pandemic caused by the novel coronavirus disease 2019 (COVID-19), which causes severe respiratory distress syndrome. Several studies have demonstrated the preventive and therapeutic effects of molecular hydrogen in the various diseases. The biological effects of molecular hydrogen mainly involve anti-inflammation, antioxidation, and autophagy and cell death modulation. This review focuses on the potential therapeutic effects of molecular hydrogen on ALI and its underlying mechanisms and aims to provide a theoretical basis for the clinical treatment of ALI and COVID-19.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004 People’s Republic of China
| | - Jin Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004 People’s Republic of China
| | - Zhiling Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004 People’s Republic of China
| |
Collapse
|
37
|
Hopster K, Hurcombe SD, Simpson K, VanderBroek AR, Driessen B. Flow-controlled expiration improves respiratory mechanics, ventilation, and gas exchange in anesthetized horses. Am J Vet Res 2022; 83:393-398. [PMID: 35175934 DOI: 10.2460/ajvr.21.10.0158] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE Mechanical ventilation is usually achieved by active lung inflation during inspiration and passive lung emptying during expiration. By contrast, flow-controlled expiration (FLEX) ventilation actively reduces the rate of lung emptying by causing linear gas flow throughout the expiratory phase. Our aim was to evaluate the effects of FLEX on lung compliance and gas exchange in anesthetized horses in dorsal recumbency. ANIMALS 8 healthy horses. PROCEDURES All animals were anesthetized twice and either ventilated beginning with FLEX or conventional volume-controlled ventilation in a randomized, crossover design. Total anesthesia time was 3 hours, with the ventilatory mode being changed after 1.5 hours. During anesthesia, cardiac output (thermodilution), mean arterial blood pressures, central venous pressure, and pulmonary arterial pressure were recorded. Further, peak, plateau, and mean airway pressures and dynamic lung compliance (Cdyn) were measured. Arterial blood gases were analyzed every 15 minutes. Data were analyzed using ANOVA (P < 0.05). RESULTS FLEX ventilation resulted in significantly higher arterial oxygen partial pressures (521 vs 227 mm Hg) and Cdyn (564 vs 431 mL/cm H2O) values compared to volume-controlled ventilation. The peak and plateau airway pressure were lower, but mean airway pressure was significantly higher (4.8 vs 9.2 cm H2O) in FLEX ventilated horses. No difference for cardiovascular parameters were detected. CLINICAL RELEVANCE The results of this study showed a significant improvement of the Pao2 and Cdyn without compromising the cardiovascular system when horses were ventilated by use of FLEX compared to conventional ventilation.
Collapse
|
38
|
Liao X, Zhang W, Dai H, Jing R, Ye M, Ge W, Pei S, Pan L. Neutrophil-Derived IL-17 Promotes Ventilator-Induced Lung Injury via p38 MAPK/MCP-1 Pathway Activation. Front Immunol 2022; 12:768813. [PMID: 34975857 PMCID: PMC8714799 DOI: 10.3389/fimmu.2021.768813] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/29/2021] [Indexed: 12/28/2022] Open
Abstract
Ventilator-induced lung injury (VILI) is one of the most common complications of mechanical ventilation and can severely affect health. VILI appears to involve excessive inflammatory responses, but its pathogenesis has not yet been clarified. Since interleukin-17 (IL-17) plays a critical role in the immune system and the development of infectious and inflammatory diseases, we investigated here whether it plays a role in VILI. In a mouse model of VILI, mechanical ventilation with high tidal volume promoted the accumulation of lung neutrophils, leading to increased IL-17 levels in the lung, which in turn upregulated macrophage chemoattractant protein-1 via p38 mitogen-activated protein kinase. Depletion of neutrophils decreases the production IL-17 in mice and inhibition of IL-17 significantly reduced HTV-induced lung injury and inflammatory response. These results were confirmed in vitro using RAW264.7 macrophage cultures. Our results suggest that IL-17 plays a pro-inflammatory role in VILI and could serve as a new target for its treatment.
Collapse
Affiliation(s)
- Xiaoting Liao
- Department of Anesthesiology, Guangxi Key Laboratory of Basic Research on Perioperative Organ Function Injury & Control, and Guangxi Medical Engineering Research Center of Tissue Injury and Repair, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Weikang Zhang
- Department of Anesthesiology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Huijun Dai
- Department of Anesthesiology, Guangxi Key Laboratory of Basic Research on Perioperative Organ Function Injury & Control, and Guangxi Medical Engineering Research Center of Tissue Injury and Repair, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Ren Jing
- Department of Anesthesiology, Guangxi Key Laboratory of Basic Research on Perioperative Organ Function Injury & Control, and Guangxi Medical Engineering Research Center of Tissue Injury and Repair, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Mengling Ye
- Department of Anesthesiology, Guangxi Key Laboratory of Basic Research on Perioperative Organ Function Injury & Control, and Guangxi Medical Engineering Research Center of Tissue Injury and Repair, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Wanyun Ge
- Department of Anesthesiology, Guangxi Key Laboratory of Basic Research on Perioperative Organ Function Injury & Control, and Guangxi Medical Engineering Research Center of Tissue Injury and Repair, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Shenglin Pei
- Department of Anesthesiology, Guangxi Key Laboratory of Basic Research on Perioperative Organ Function Injury & Control, and Guangxi Medical Engineering Research Center of Tissue Injury and Repair, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Linghui Pan
- Department of Anesthesiology, Guangxi Key Laboratory of Basic Research on Perioperative Organ Function Injury & Control, and Guangxi Medical Engineering Research Center of Tissue Injury and Repair, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
39
|
Rudolph MW, Slager S, Burgerhof JGM, van Woensel JB, Alffenaar JWC, Wösten - van Asperen RM, de Hoog M, IJland MM, Kneyber MCJ. Paediatric Acute Respiratory Distress Syndrome Neuromuscular Blockade study (PAN-study): a phase IV randomised controlled trial of early neuromuscular blockade in moderate-to-severe paediatric acute respiratory distress syndrome. Trials 2022; 23:96. [PMID: 35101098 PMCID: PMC8802263 DOI: 10.1186/s13063-021-05927-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/08/2021] [Indexed: 11/11/2022] Open
Abstract
Background Paediatric acute respiratory distress syndrome (PARDS) is a manifestation of severe, life-threatening lung injury necessitating mechanical ventilation with mortality rates ranging up to 40–50%. Neuromuscular blockade agents (NMBAs) may be considered to prevent patient self-inflicted lung injury in PARDS patients, but two trials in adults with severe ARDS yielded conflicting results. To date, randomised controlled trials (RCT) examining the effectiveness and efficacy of NMBAs for PARDS are lacking. We hypothesise that using NMBAs for 48 h in paediatric patients younger than 5 years of age with early moderate-to-severe PARDS will lead to at least a 20% reduction in cumulative respiratory morbidity score 12 months after discharge from the paediatric intensive care unit (PICU). Methods This is a phase IV, multicentre, randomised, double-blind, placebo-controlled trial performed in level-3 PICUs in the Netherlands. Eligible for inclusion are children younger than 5 years of age requiring invasive mechanical ventilation with positive end-expiratory pressure (PEEP) ≥ 5 cm H2O for moderate-to-severe PARDS occurring within the first 96 h of PICU admission. Patients are randomised to continuous infusion of rocuronium bromide or placebo for 48 h. The primary endpoint is the cumulative respiratory morbidity score 12 months after PICU discharge, adjusted for confounding by age, gestational age, family history of asthma and/or allergy, season in which questionnaire was filled out, day-care and parental smoking. Secondary outcomes include respiratory mechanics, oxygenation and ventilation metrics, pulmonary and systemic inflammation markers, prevalence of critical illness polyneuropathy and myopathy and metrics for patient outcome including ventilator free days at day 28, length of PICU and hospital stay, and mortality Discussion This is the first paediatric trial evaluating the effects of muscular paralysis in moderate-to-severe PARDS. The proposed study addresses a huge research gap identified by the Paediatric Acute Lung Injury Consensus Collaborative by evaluating practical needs regarding the treatment of PARDS. Paediatric critical care practitioners are inclined to use interventions such as NMBAs in the most critically ill. This liberal use must be weighed against potential side effects. The proposed study will provide much needed scientific support in the decision-making to start NMBAs in moderate-to-severe PARDS. Trial registration ClinicalTrials.govNCT02902055. Registered on September 15, 2016.
Collapse
|
40
|
Itagaki T. Diaphragm-protective mechanical ventilation in acute respiratory failure. THE JOURNAL OF MEDICAL INVESTIGATION 2022; 69:165-172. [DOI: 10.2152/jmi.69.165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
41
|
Tukanova KH, Chidambaram S, Guidozzi N, Hanna GB, McGregor AH, Markar SR. Physiotherapy Regimens in Esophagectomy and Gastrectomy: a Systematic Review and Meta-Analysis. Ann Surg Oncol 2021; 29:3148-3167. [PMID: 34961901 PMCID: PMC8990957 DOI: 10.1245/s10434-021-11122-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/11/2021] [Indexed: 12/30/2022]
Abstract
Background Esophageal and gastric cancer surgery are associated with considerable morbidity, specifically postoperative pulmonary complications (PPCs), potentially accentuated by underlying challenges with malnutrition and cachexia affecting respiratory muscle mass. Physiotherapy regimens aim to increase the respiratory muscle strength and may prevent postoperative morbidity. Objective The aim of this study was to assess the impact of physiotherapy regimens in patients treated with esophagectomy or gastrectomy. Methods An electronic database search was performed in the MEDLINE, EMBASE, CENTRAL, CINAHL and Pedro databases. A meta-analysis was performed to assess the impact of physiotherapy on the functional capacity, incidence of PPCs and postoperative morbidity, in-hospital mortality rate, length of hospital stay (LOS) and health-related quality of life (HRQoL). Results Seven randomized controlled trials (RCTs) and seven cohort studies assessing prehabilitation totaling 960 patients, and five RCTs and five cohort studies assessing peri- or postoperative physiotherapy with 703 total patients, were included. Prehabilitation resulted in a lower incidence of postoperative pneumonia and morbidity (Clavien–Dindo score ≥ II). No difference was observed in functional exercise capacity and in-hospital mortality following prehabilitation. Meanwhile, peri- or postoperative rehabilitation resulted in a lower incidence of pneumonia, shorter LOS, and better HRQoL scores for dyspnea and physical functioning, while no differences were found for the QoL summary score, global health status, fatigue, and pain scores. Conclusion This meta-analysis suggests that implementing an exercise intervention may be beneficial in both the preoperative and peri- or postoperative periods. Further investigation is needed to understand the mechanism through which exercise interventions improve clinical outcomes and which patient subgroup will gain the maximal benefit. Supplementary Information The online version contains supplementary material available at 10.1245/s10434-021-11122-7.
Collapse
Affiliation(s)
- Karina H Tukanova
- Department of Surgery and Cancer, Imperial College London, London, UK
| | | | - Nadia Guidozzi
- Department of Surgery, University of the Witwatersrand, Johannesburg, South Africa
| | - George B Hanna
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Alison H McGregor
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Sheraz R Markar
- Department of Surgery and Cancer, Imperial College London, London, UK. .,Nuffield Department of Surgery, University of Oxford, Oxford, UK. .,Department of Molecular Medicine and Surgery, Karolinska Institutet, Solna, Sweden. .,Division of Surgery, Department of Surgery and Cancer, St Mary's Hospital, London, UK.
| |
Collapse
|
42
|
Wu SW, Peng CK, Wu SY, Wang Y, Yang SS, Tang SE, Huang KL. Obesity Attenuates Ventilator-Induced Lung Injury by Modulating the STAT3-SOCS3 Pathway. Front Immunol 2021; 12:720844. [PMID: 34489970 PMCID: PMC8417798 DOI: 10.3389/fimmu.2021.720844] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
Background Ventilator-induced lung injury (VILI) is characterized by vascular barrier dysfunction and suppression of alveolar fluid clearance (AFC). Obesity itself leads to chronic inflammation, which may initiate an injurious cascade to the lungs and simultaneously induce a protective feedback. In this study, we investigated the protective mechanism of obesity on VILI in a mouse model. Methods The VILI model was set up via 6-h mechanical ventilation with a high tidal volume. Parameters including lung injury score, STAT3/NFκB pathway, and AFC were assessed. Mice with diet-induced obesity were obtained by allowing free access to a high-fat diet since the age of 3 weeks. After a 9-week diet intervention, these mice were sacrificed at the age of 12 weeks. The manipulation of SOCS3 protein was achieved by siRNA knockdown and pharmaceutical stimulation using hesperetin. WNK4 knockin and knockout obese mice were used to clarify the pathway of AFC modulation. Results Obesity itself attenuated VILI. Knockdown of SOCS3 in obese mice offset the protection against VILI afforded by obesity. Hesperetin stimulated SOCS3 upregulation in nonobese mice and provided protection against VILI. In obese mice, the WNK4 axis was upregulated at the baseline, but was significantly attenuated after VILI compared with nonobese mice. At the baseline, the manipulation of SOCS3 by siRNA and hesperetin also led to the corresponding alteration of WNK4, albeit to a lesser extent. After VILI, WNK4 expression correlated with STAT3/NFκB activation, regardless of SOCS3 status. Obese mice carrying WNK4 knockout had VILI with a severity similar to that of wild-type obese mice. The severity of VILI in WNK4-knockin obese mice was counteracted by obesity, similar to that of wild-type nonobese mice only. Conclusions Obesity protects lungs from VILI by upregulating SOCS3, thus suppressing the STAT3/NFκB inflammatory pathway and enhancing WNK4-related AFC. However, WNK4 activation is mainly from direct NFκB downstreaming, and less from SOCS3 upregulation. Moreover, JAK2-STAT3/NFκB signaling predominates the pathogenesis of VILI. Nevertheless, the interaction between SOCS3 and WNK4 in modulating VILI in obesity warrants further investigation.
Collapse
Affiliation(s)
- Shih-Wei Wu
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Tri-Service General Hospital, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chung-Kan Peng
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Tri-Service General Hospital, Taipei, Taiwan
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Yu Wu
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Yu Wang
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Sung-Sen Yang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Division of Nephrology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shih-En Tang
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Tri-Service General Hospital, Taipei, Taiwan
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Tri-Service General Hospital, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
43
|
Effect of Driving Pressure Change During Extracorporeal Membrane Oxygenation in Adults With Acute Respiratory Distress Syndrome: A Randomized Crossover Physiologic Study. Crit Care Med 2021; 48:1771-1778. [PMID: 33044283 DOI: 10.1097/ccm.0000000000004637] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Venovenous extracorporeal membrane oxygenation is an effective intervention to improve gas exchange in patients with severe acute respiratory distress syndrome. However, the mortality of patients with severe acute respiratory distress syndrome supported with venovenous extracorporeal membrane oxygenation remains high, and this may be due in part to a lack of standardized mechanical ventilation strategies aimed at further minimizing ventilator-induced lung injury. We tested whether a continuous positive airway pressure ventilation strategy mitigates ventilator-induced lung injury in patients with severe acute respiratory distress syndrome on venovenous extracorporeal membrane oxygenation, compared with current ventilation practice that employs tidal ventilation with limited driving pressure. We used plasma biomarkers as a surrogate outcome for ventilator-induced lung injury. DESIGN Randomized crossover physiologic study. SETTING Single-center ICU. PATIENTS Ten patients with severe acute respiratory distress syndrome supported on venovenous extracorporeal membrane oxygenation. INTERVENTIONS The study included four phases. After receiving pressure-controlled ventilation with driving pressure of 10 cm H2O for 1 hour (phase 1), patients were randomly assigned to receive first either pressure-controlled ventilation 20 cm H2O for 2 hours (phase 2) or continuous positive airway pressure for 2 hours (phase 3), and then crossover to the other phase for 2 hours; during phase 4 ventilation settings returned to baseline (pressure-controlled ventilation 10 cm H2O) for 4 hours. MEASUREMENTS AND MAIN RESULTS There was a linear relationship between the change in driving pressure and the plasma concentration of interleukin-6, soluble receptor for advanced glycation end products, interleukin-1ra, tumor necrosis factor alpha, surfactant protein D, and interleukin-10. CONCLUSIONS Ventilator-induced lung injury may occur in acute respiratory distress syndrome patients on venovenous extracorporeal membrane oxygenation despite the delivery of volume- and pressure-limited mechanical ventilation. Reducing driving pressure to zero may provide more protective mechanical ventilation in acute respiratory distress syndrome patients supported with venovenous extracorporeal membrane oxygenation. However, the risks versus benefits of such an approach need to be confirmed in studies that are designed to test patient centered outcomes.
Collapse
|
44
|
Kosyreva A, Dzhalilova D, Lokhonina A, Vishnyakova P, Fatkhudinov T. The Role of Macrophages in the Pathogenesis of SARS-CoV-2-Associated Acute Respiratory Distress Syndrome. Front Immunol 2021; 12:682871. [PMID: 34040616 PMCID: PMC8141811 DOI: 10.3389/fimmu.2021.682871] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 04/22/2021] [Indexed: 12/11/2022] Open
Abstract
Macrophages are cells that mediate both innate and adaptive immunity reactions, playing a major role in both physiological and pathological processes. Systemic SARS-CoV-2-associated complications include acute respiratory distress syndrome (ARDS), disseminated intravascular coagulation syndrome, edema, and pneumonia. These are predominantly effects of massive macrophage activation that collectively can be defined as macrophage activation syndrome. In this review we focus on the role of macrophages in COVID-19, as pathogenesis of the new coronavirus infection, especially in cases complicated by ARDS, largely depends on macrophage phenotypes and functionalities. We describe participation of monocytes, monocyte-derived and resident lung macrophages in SARS-CoV-2-associated ARDS and discuss possible utility of cell therapies for its treatment, notably the use of reprogrammed macrophages with stable pro- or anti-inflammatory phenotypes.
Collapse
Affiliation(s)
- Anna Kosyreva
- Department of Neuromorphology, Science Research Institute of Human Morphology, Moscow, Russia
- Histology Department, Peoples Friendship University of Russia (RUDN University), Moscow, Russia
| | - Dzhuliia Dzhalilova
- Department of Immunomorphology of Inflammation, Science Research Institute of Human Morphology, Moscow, Russia
| | - Anastasia Lokhonina
- Histology Department, Peoples Friendship University of Russia (RUDN University), Moscow, Russia
- Department of Regenerative Medicine, National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Polina Vishnyakova
- Histology Department, Peoples Friendship University of Russia (RUDN University), Moscow, Russia
- Department of Regenerative Medicine, National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Timur Fatkhudinov
- Histology Department, Peoples Friendship University of Russia (RUDN University), Moscow, Russia
- Department of Growth and Development, Science Research Institute of Human Morphology, Moscow, Russia
| |
Collapse
|
45
|
Orizondo RA, Omecinski KS, May AG, Dhamotharan V, Frankowski BJ, Burgreen GW, Ye SH, Kocyildirim E, Sanchez PG, D’Cunha J, Wagner WR, Federspiel WJ. Month-long Respiratory Support by a Wearable Pumping Artificial Lung in an Ovine Model. Transplantation 2021; 105:999-1007. [PMID: 33031226 PMCID: PMC8024407 DOI: 10.1097/tp.0000000000003481] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND A wearable artificial lung could improve lung transplantation outcomes by easing implementation of physical rehabilitation during long-term pretransplant respiratory support. The Modular Extracorporeal Lung Assist System (ModELAS) is a compact pumping artificial lung currently under development. This study evaluated the long-term in vivo performance of the ModELAS during venovenous support in awake sheep. Feedback from early trials and computational fluid dynamic analysis guided device design optimization along the way. METHODS The ModELAS was connected to healthy sheep via a dual-lumen cannula in the jugular vein. Sheep were housed in a fixed-tether pen while wearing the device in a holster during support. Targeted blood flow rate and support duration were 2-2.5 L/min and 28-30 days, respectively. Anticoagulation was maintained via systemic heparin. Device pumping and gas exchange performance and hematologic indicators of sheep physiology were measured throughout support. RESULTS Computational fluid dynamic-guided design modifications successfully decreased pump thrombogenicity from initial designs. For the optimized design, 4 of 5 trials advancing past early perioperative and cannula-related complications lasted the full month of support. Blood flow rate and CO2 removal in these trials were 2.1 ± 0.3 L/min and 139 ± 15 mL/min, respectively, and were stable during support. One trial ended after 22 days of support due to intradevice thrombosis. Support was well tolerated by the sheep with no signs of hemolysis or device-related organ impairment. CONCLUSIONS These results demonstrate the ability of the ModELAS to provide safe month-long support without consistent deterioration of pumping or gas exchange capabilities.
Collapse
Affiliation(s)
- Ryan A. Orizondo
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
- Department of Medicine, University of Pittsburgh
- Department of Bioengineering, University of Pittsburgh
| | - Katelin S. Omecinski
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
- Department of Bioengineering, University of Pittsburgh
| | - Alexandra G. May
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
- Department of Chemical and Petroleum Engineering, University of Pittsburgh
| | - Vishaal Dhamotharan
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
- Department of Bioengineering, University of Pittsburgh
| | | | - Greg W. Burgreen
- Computational Fluid Dynamics Group, Center for Advanced Vehicular Systems, Mississippi State University
| | - Sang-Ho Ye
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
- Department of Surgery, University of Pittsburgh
| | - Ergin Kocyildirim
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
- Department of Cardiothoracic Surgery, Children’s Hospital of Pittsburgh
| | - Pablo G. Sanchez
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center
| | - Jonathan D’Cunha
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center
| | - William R. Wagner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
- Department of Bioengineering, University of Pittsburgh
- Department of Chemical and Petroleum Engineering, University of Pittsburgh
- Department of Surgery, University of Pittsburgh
| | - William J. Federspiel
- McGowan Institute for Regenerative Medicine, University of Pittsburgh
- Department of Bioengineering, University of Pittsburgh
- Department of Chemical and Petroleum Engineering, University of Pittsburgh
- Department of Critical Care Medicine, University of Pittsburgh Medical Center
- Clinical and Translational Science Institute, University of Pittsburgh
| |
Collapse
|
46
|
Nguyen TK, Mai DH, Le AN, Nguyen QH, Nguyen CT, Vu TA. A review of intraoperative lung-protective mechanical ventilation strategy. TRENDS IN ANAESTHESIA AND CRITICAL CARE 2021. [DOI: 10.1016/j.tacc.2020.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
47
|
Liu H, Jin J, Huang D. Strategic ventilation reduces non-ventilated contralateral lung injury induced by one-lung ventilation in rabbits. ARQ BRAS MED VET ZOO 2021. [DOI: 10.1590/1678-4162-12198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
ABSTRACT One lung ventilation (OLV) often results in trauma to the unventilated contralateral lung. This study aims to evaluate the effects of different OLV regimens on the injury of the unventilated contralateral lung to identify the best conditions for OLV. Forty rabbits were divided into five groups: a sham group, OLV group I (fraction of inspired oxygen (FIO2) 1.0, tidal volume (VT) 8mL/kg, respiratory rate (R) 40 breaths/min and inspiratory/expiratory ratio (I:E) 1:2), OLV group II (FIO2=1.0, VT 8mL/kg, R 40 breaths/min, I:E 1:2, and positive end-expiratory pressure (PEEP) 5 cm H2O), OLV group III (FIO2 1.0, VT 6mL/kg, R 40 breaths/min, I:E 1:2 and PEEP 5 cm H2O) and OLV group IV (FIO2 0.8, VT 6mL/kg, R 40 breaths/min, I:E 1:2 and PEEP 5 cm H2O). Animals from all OLV groups received two-lung ventilation (TLV) to establish a baseline, followed by one of the indicated OLV regimens. The rabbits in the sham group were intubated through trachea and ventilated with fresh air. Arterial blood gas samples were collected, lung injury parameters were evaluated, and the concentrations of TNF-α and IL-8 in bronchoalveolar lavage fluid (BALF) and pulmonary surfactant protein A (SPA) in the unventilated lung were also measured. In OLV group I, the unventilated left lung had higher TNF-α, IL-8 and lung injury score but lower SPA than the ventilated right lung. In OLV groups I to III, the concentrations of TNF-α, IL-8 and lung injury score in the left lung decreased but SPA increased. No differences in these parameters between OLV groups III and IV were observed. Strategic ventilation designed for OLV groups III and IV reduced OLV-induced injury of the non-ventilated contralateral lung in rabbits.
Collapse
Affiliation(s)
- H.J. Liu
- Shanghai University of Medicine & Health Sciences, China
| | - J. Jin
- Shanghai University of Medicine & Health Sciences, China
| | | |
Collapse
|
48
|
Knecht RS, Bucher CH, Van Linthout S, Tschöpe C, Schmidt-Bleek K, Duda GN. Mechanobiological Principles Influence the Immune Response in Regeneration: Implications for Bone Healing. Front Bioeng Biotechnol 2021; 9:614508. [PMID: 33644014 PMCID: PMC7907627 DOI: 10.3389/fbioe.2021.614508] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 01/22/2021] [Indexed: 12/13/2022] Open
Abstract
A misdirected or imbalanced local immune composition is often one of the reasons for unsuccessful regeneration resulting in scarring or fibrosis. Successful healing requires a balanced initiation and a timely down-regulation of the inflammation for the re-establishment of a biologically and mechanically homeostasis. While biomaterial-based approaches to control local immune responses are emerging as potential new treatment options, the extent to which biophysical material properties themselves play a role in modulating a local immune niche response has so far been considered only occasionally. The communication loop between extracellular matrix, non-hematopoietic cells, and immune cells seems to be specifically sensitive to mechanical cues and appears to play a role in the initiation and promotion of a local inflammatory setting. In this review, we focus on the crosstalk between ECM and its mechanical triggers and how they impact immune cells and non-hematopoietic cells and their crosstalk during tissue regeneration. We realized that especially mechanosensitive receptors such as TRPV4 and PIEZO1 and the mechanosensitive transcription factor YAP/TAZ are essential to regeneration in various organ settings. This indicates novel opportunities for therapeutic approaches to improve tissue regeneration, based on the immune-mechanical principles found in bone but also lung, heart, and skin.
Collapse
Affiliation(s)
- Raphael S Knecht
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christian H Bucher
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Carsten Tschöpe
- Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany.,Department of Cardiology, Charite'-Universitätsmedizin Berlin, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Georg N Duda
- Julius Wolff Institute and Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
49
|
Li J, Wang K, Huang B, Li R, Wang X, Zhang H, Tang H, Chen X. The receptor for advanced glycation end products mediates dysfunction of airway epithelial barrier in a lipopolysaccharides-induced murine acute lung injury model. Int Immunopharmacol 2021; 93:107419. [PMID: 33548580 DOI: 10.1016/j.intimp.2021.107419] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 01/10/2021] [Accepted: 01/18/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Airway epithelial cells (AECs) act as the first barrier protecting against invasion of environment agents and maintain integrity of lung structure and function. Dysfunction of airway epithelial barrier has been shown to be involved in ALI/ARDS pathogenesis. Yet, the exact mechanism is still obscure. Our study evaluated whether the receptor for advanced glycation end products (RAGE) mediates impaired airway epithelial barrier in LPS-induced murine ALI model. METHODS Male BALB/c mice were subjected to intratracheal instillation of LPS to generate an ALI model. Inhibitors of RAGE, FPS-ZM1 and Azeliragon were respectively given to the mice through intraperitoneal injection. Bronchoalveolar lavage fluid (BALF) and lung tissues were collected for further analysis. RESULTS LPS exposure led to markedly increased expression of RAGE and its ligands HMGB1, HSP70, S100b. Treatment of FPS-ZM1 or Azeliragon not only effectively descended the expression of RAGE and its ligands but also attenuated LPS-induced neutrophil-predominant airway inflammation and injury, decreased levels of IL-6, IL-1β and TNF-α in BALF, alleviated increased alveolar-capillary permeability and pulmonary edema. LPS stimulation significantly impaired the integrity of airway epithelium, paralleled with dislocation of adheren junction (AJ) protein E-cadherin at cell-cell contacts and down-expression of both AJ and tight junction (TJ) proteins Claudin-2 and occludin, all of which were dramatically rescued by RAGE inhibition. CONCLUSION RAGE signaling mediates airway epithelial barrier dysfunction in a LPS-induced ALI murine model.
Collapse
Affiliation(s)
- Jiahui Li
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China
| | - Kai Wang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China
| | - Bo Huang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China
| | - Rui Li
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China
| | - Xilong Wang
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China
| | - Hailing Zhang
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China
| | - Haixiong Tang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China.
| | - Xin Chen
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, Guangdong 510280, China.
| |
Collapse
|
50
|
Abstract
Acute Respiratory Distress Syndrome (ARDS) is defined as the rapid onset of non-cardiogenic pulmonary edema resulting in respiratory failure and hypoxemia. Efforts over the past 25 years, such as those of the ARDS and Prevention and Early Treatment of Acute Lung Injury (PETAL) Networks, have demonstrated a praiseworthy collaboration to further optimize the management of ARDS. However, improvements have been only moderate and ARDS remains a leading cause of mortality in the perioperative and critical care setting. Recently, the significant morbidity and mortality of ARDS have been emphasized by its high incidence in Coronavirus Disease 2019 (COVID-19) patients. A major hurdle to reducing ARDS mortality is that current treatment is limited to preventive measures – such as the use of lung-protective ventilation. Therapeutic approaches targeting the underlying inflammatory lung disease are areas of intensive research, but have not been clinically implemented. Nevertheless, basic science and clinical research efforts that are aimed at identifying novel treatment approaches and further improving outcomes for ARDS are ongoing. Here, we review evidence-based management approaches for ARDS, while highlighting those being investigated or heavily utilized in ARDS associated with COVID-19. Acute Respiratory Distress Syndrome remains a condition that carries a high mortality. Evidence-based clinical management and emerging concepts for new therapies for COVID-19 are reviewed.
Collapse
Affiliation(s)
- George W. Williams
- Department of Anesthesiology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Nathaniel K. Berg
- Department of Anesthesiology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Alexander Reskallah
- Department of Anesthesiology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Xiaoyi Yuan
- Department of Anesthesiology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Holger K. Eltzschig
- Department of Anesthesiology, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| |
Collapse
|