1
|
Pequignot B, Lescroart M, Levy B, Kimounn A, Koszutski M. Electrical impedance tomography to set high pressure in time-controlled adaptive ventilation. J Crit Care 2025; 87:155033. [PMID: 39904168 DOI: 10.1016/j.jcrc.2025.155033] [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: 10/01/2024] [Accepted: 01/27/2025] [Indexed: 02/06/2025]
Abstract
INTRODUCTION TCAV (Time controlled adaptive ventilation), a combination of settings applied to the APRV (airway pressure release ventilation) mode, provides personalized ventilation tailored to the lung condition in ARDS (acute respiratory distress syndrome). The objective was to evaluate whether electrical impedance tomography (EIT) could serve as a tool for guiding Phigh level in TCAV for ARDS patients. METHODS Eleven patients with moderate or severe ARDS were enrolled in a prospective single-center study in 2023. Patients were monitored with EIT (PulmoVista 500). Phigh trial was conducted from 34 to 18 cmH2O, with 4-cmH2O Phigh decrements every 5 min. Driving pressure was maintained constant by adjusting Tlow. Best EIT-derived-Phigh was defined as the pressure at the crossing point between overdistension and collapse curves. RESULTS CRS was significantly higher at Phigh 18 cmH2O with 43 [32-50] mL/cmH2O than at Phigh 34 with 20 mL/cmH2O [14-24], p < 0.005. Highest Phigh levels caused significant overdistension in the anterior region and anterior compliance is significantly lower at Phigh 34 with 10 [6-11] mL/cmH2O than at Phigh 22 cmH2O with 18 [13-25] mL/cmH2O. Best EIT-derived Phigh were 18, 22, 26 cmH2O for four, five and two patients respectively. CONCLUSION EIT enabled detection of regional ventilation distribution on TCAV during a decremental Phigh trial and thus enabled the determination of a best EIT-derived-Phigh through an individualized approach, achieving best compromise between overdistension and collapse. The observed overdistention variability highlights the necessity of Phigh level personalization on TCAV.
Collapse
Affiliation(s)
- Benjamin Pequignot
- Service de Médecine Intensive et Réanimation, Hôpital Brabois, CHRU Nancy, 54500 Vandoeuvre les Nancy, France; Université de Lorraine, INSERM, DCAC, F-54000 Nancy, France.
| | - Mickael Lescroart
- Service de Médecine Intensive et Réanimation, Hôpital Brabois, CHRU Nancy, 54500 Vandoeuvre les Nancy, France; Université de Lorraine, INSERM, DCAC, F-54000 Nancy, France
| | - Bruno Levy
- Service de Médecine Intensive et Réanimation, Hôpital Brabois, CHRU Nancy, 54500 Vandoeuvre les Nancy, France; Université de Lorraine, INSERM, DCAC, F-54000 Nancy, France
| | - Antoine Kimounn
- Service de Médecine Intensive et Réanimation, Hôpital Brabois, CHRU Nancy, 54500 Vandoeuvre les Nancy, France; Université de Lorraine, INSERM, DCAC, F-54000 Nancy, France
| | - Matthieu Koszutski
- Service de Médecine Intensive et Réanimation, Hôpital Brabois, CHRU Nancy, 54500 Vandoeuvre les Nancy, France; Université de Lorraine, INSERM, DCAC, F-54000 Nancy, France
| |
Collapse
|
2
|
Shi Q, Liu H, Wang H, Tang L, Di Q, Wang D. MFGE8 regulates the EndoMT of HLMECs through the BMP signaling pathway and fibrosis in acute lung injury. Respir Res 2025; 26:142. [PMID: 40223052 PMCID: PMC11995649 DOI: 10.1186/s12931-025-03215-8] [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: 09/14/2024] [Accepted: 04/01/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND To investigate the effects and mechanisms of MFGE8 on LPS-induced endothelial-to-mesenchymal transition (EndoMT) and pulmonary fibrosis in human lung microvascular endothelial cells (HLMECs) and a mouse model of acute lung injury. METHODS Serum MFGE8 levels were compared between ARDS patients and controls. In vitro, HLMECs were treated with LPS, siRNA targeting MFGE8, and recombinant human MFGE8 (rhMFGE8).HLMEC morphology, invasion, migration, and EndoMT markers (CD31, ɑ-SMA) were evaluated. BMP/Smad1/5-Smad4 signaling and Snail expression were assessed via immunofluorescence, western blotting, and qRT-PCR. In vivo, rhMFGE8 effects on pulmonary fibrosis and EndoMT were analyzed in a mouse model of acute lung injury. RESULTS MFGE8 levels were significantly reduced in ARDS patients, with higher levels correlating to better survival. In vitro, rhMFGE8 improved HLMEC morphology, reduced invasion and migration, and attenuated LPS-induced EndoMT by increasing CD31 and decreasing α-SMA. MFGE8 knockdown increased BMP/Smad1/5-Smad4 signaling and Snail expression, while rhMFGE8 inhibited these effects. In vivo, rhMFGE8 ameliorated pulmonary fibrosis and EndoMT in mice. CONCLUSIONS MFGE8 regulates LPS-induced EndoMT in HLMECs via the BMP/Smad1/5-Smad4 pathway and protects against pulmonary fibrosis in acute lung injury, suggesting it as a therapeutic target for ALI and ARDS.
Collapse
Affiliation(s)
- Qingqiang Shi
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Huang Liu
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hanghang Wang
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Ling Tang
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Qi Di
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Daoxin Wang
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
3
|
Zhu D, Zhang J, Huang X, Wei N, Jiang J, Li J, Liu L, Liu Y, Zhou J, Jia J. Integrated network pharmacology and experimental validation to elucidate the mechanism of celastrol in mitigating sepsis-induced acute lung injury in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156678. [PMID: 40133025 DOI: 10.1016/j.phymed.2025.156678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/13/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND Sepsis is an acute, life-threatening condition that precipitates multiple organ failure, including acute lung injury (ALI), characterized by a complex pathophysiological process and elevated mortality rates. Celastrol, a pentacyclic triterpenoid quinone derived from traditional Chinese medicine, exhibits diverse pharmacological properties, including immunomodulatory, anti-inflammatory, anticancer, and antifibrotic effects, and has demonstrated favorable safety profiles in vivo. However, the precise mechanism by which CSL contributes to sepsis-induced ALI remains to be elucidated. PURPOSE The study aimed to explore the mechanisms by which celastrol mitigates sepsis-induced ALI using network pharmacology, followed by experimental validation of its regulatory effects on sepsis-induced ALI. METHODS Utilizing a network pharmacology analysis, the potential targets and pathways of celastrol were identified. To explore celastrol's therapeutic effects on ALI, a rat model of sepsis was induced via cecal ligation and puncture, followed by assessment through hematoxylin-eosin staining, Real-time quantitative polymerase chain reaction (RT-qPCR), and Western blotting. Further investigation involved evaluating celastrol's influence on LPS-stimulated A549 and Raw264.7 cells, employing RT-qPCR, Western blotting, and immunofluorescence techniques. RESULTS Network pharmacological analysis identified 10 core targets and 31 pathways relevant to sepsis-induced ALI, with STAT3, TLR4, HIF-1α, and NF-κB1 emerging as central targets. Animal experiments demonstrated that celastrol treatment significantly reduced lung tissue inflammation, as evidenced by immunohistochemistry, Western blot, and RT-qPCR results, in comparison to the cecal ligation and puncture group. Notably, the levels of IL-1β, TNF-α, HIF-1α, STAT3, and NF-κB1 proteins and mRNA in the celastrol treatment group were significantly reduced compared to those in the cecal ligation and puncture (CLP) group and the LPS-treated group. Additionally, Western blot and immunofluorescence analyses confirmed the activation of the NF-κB pathway in vitro. CONCLUSION This study indicates that celastrol significantly suppresses the expression of inflammatory factors in sepsis-induced ALI by inhibiting the NF-κB/HIF-1α pathway in both in vivo and in vitro models, highlighting its therapeutic potential for modulating inflammation. These findings provide valuable evidence for future clinical research and drug development.
Collapse
Affiliation(s)
- Danli Zhu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jinghan Zhang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xiaochun Huang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Na Wei
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jinxiu Jiang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jiayao Li
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Li Liu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Yulin Liu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jun Zhou
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China.
| | - Jing Jia
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China; Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China.
| |
Collapse
|
4
|
Feng Y, Tang M, Li H, Yao S, Li B. Mouse mesenchymal stem cell-derived exosomal miR-205-5p modulates LPS-induced macrophage polarization and alleviates lung injury by regulating the USP7/FOXM1 axis. Drug Deliv Transl Res 2025:10.1007/s13346-025-01813-z. [PMID: 40000557 DOI: 10.1007/s13346-025-01813-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2025] [Indexed: 02/27/2025]
Abstract
Exosomal microRNAs produced from mesenchymal stem cells (MSCs) are crucial in the management of acute lung injury (ALI). In this work, mMSCs separated from bone marrow were used to extract exosomes (MSC-Exos). MSC-Exos treatment attenuated pathological changes and scores, and edema in ALI mice. Also, MSC-Exos administration modulated the concentrations of inflammatory factors as well as the macrophage polarization both in vivo and in vitro. Upregulation of miR-205-5p in MSC-Exos regulated the macrophage polarization and the contents of inflammatory factors in animal and cell models. MiR-205-5p targeted USP7, and negatively modulated the expression of USP7. USP7 interacted with FOXM1, and reduced the ubiquitination degradation of FOXM1. MSC-derived exosomal miR-205-5p modulated ubiquitination of FOXM1 by targeting USP7. The ameliorative effect of MSC-Exos on the macrophage polarization and the inflammatory factors release was reversed with the overexpression of USP7 in animal and cell models. Collectively, MSC-derived exosomal miR-205-5p regulated lipopolysaccharide (LPS)-induced macrophage polarization and alleviated lung injury by the USP7/FOXM1 axis, which developed a potential target for the treatment of ALI.
Collapse
Affiliation(s)
- Yinglu Feng
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Min Tang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Haopeng Li
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Shanglong Yao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
| | - Bo Li
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Anesthesiology and Resuscitation, Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
| |
Collapse
|
5
|
Kingsley J, Kandil O, Satalin J, Bary AA, Coyle S, Nawar MS, Groom R, Farrag A, Shah J, Robedee BR, Darling E, Shawkat A, Chaudhuri D, Nieman GF, Aiash H. The use of protective mechanical ventilation during extracorporeal membrane oxygenation for the treatment of acute respiratory failure. Perfusion 2025; 40:69-82. [PMID: 38240747 DOI: 10.1177/02676591241227167] [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] [Indexed: 01/11/2025]
Abstract
Acute respiratory failure (ARF) strikes an estimated two million people in the United States each year, with care exceeding US$50 billion. The hallmark of ARF is a heterogeneous injury, with normal tissue intermingled with a large volume of low compliance and collapsed tissue. Mechanical ventilation is necessary to oxygenate and ventilate patients with ARF, but if set inappropriately, it can cause an unintended ventilator-induced lung injury (VILI). The mechanism of VILI is believed to be overdistension of the remaining normal tissue known as the 'baby' lung, causing volutrauma, repetitive collapse and reopening of lung tissue with each breath, causing atelectrauma, and inflammation secondary to this mechanical damage, causing biotrauma. To avoid VILI, extracorporeal membrane oxygenation (ECMO) can temporally replace the pulmonary function of gas exchange without requiring high tidal volumes (VT) or airway pressures. In theory, the lower VT and airway pressure will minimize all three VILI mechanisms, allowing the lung to 'rest' and heal in the collapsed state. The optimal method of mechanical ventilation for the patient on ECMO is unknown. The ARDSNetwork Acute Respiratory Management Approach (ARMA) is a Rest Lung Approach (RLA) that attempts to reduce the excessive stress and strain on the remaining normal lung tissue and buys time for the lung to heal in the collapsed state. Theoretically, excessive tissue stress and strain can also be avoided if the lung is fully open, as long as the alveolar re-collapse is prevented during expiration, an approach known as the Open Lung Approach (OLA). A third lung-protective strategy is the Stabilize Lung Approach (SLA), in which the lung is initially stabilized and gradually reopened over time. This review will analyze the physiologic efficacy and pathophysiologic potential of the above lung-protective approaches.
Collapse
Affiliation(s)
| | | | | | - Akram Abdel Bary
- Critical Care Department, Faculty of Medicine Cairo University, Cairo, Egypt
| | - Sierra Coyle
- SUNY Upstate Medical University, Syracuse, NY, USA
| | - Mahmoud Saad Nawar
- Critical Care Department, Faculty of Medicine Cairo University, Cairo, Egypt
| | - Robert Groom
- SUNY Upstate Medical University, Syracuse, NY, USA
| | - Amr Farrag
- Aswan Heart Centre, Magdi Yacoub Foundation, Aswan, Egypt
| | | | | | | | | | | | | | - Hani Aiash
- SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
6
|
Zhao M, Lin J, Wang X, Chen C, Li J, Yu J, Zhou T, Liang Y, Shen X, Shi R, Yang S, Zeng S, Deng Y, Duan X, Zhou L, Sun X, Wang Y, Shu Z. Multi-immunometabolomics mining: NP prevents hyperimmune in ALI by inhibiting Leucine/PI3K/Akt/mTOR signaling pathway. Free Radic Biol Med 2024; 225:302-315. [PMID: 39370053 DOI: 10.1016/j.freeradbiomed.2024.09.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
Acute lung injury (ALI) is currently a global health concern. Nicandra physalodes (L.) Gaertn. (NP) holds an important position in traditional Chinese medicine and nutrition. The potential protective mechanisms of NP against ALI remain unknown. The purpose of this study was to investigate the protective effects and molecular mechanisms of NP extract (NPE) on lipopolysaccharide (LPS)-induced ALI in mice. By utilizing network pharmacology to forecast the active ingredients in NP as well as possible signaling pathways. The composition of the NPE was analyzed using UPLC-Q-TOF-MS/MS. In addition, 1H-NMR immunometabolomics was employed to identify alterations in primary metabolic pathways and metabolites in the lung, serum, and fecal tissues. Finally, the protein and gene expression of key pathways were verified by IHC, IF, RT-qPCR, and ELISA. It was found that the main ingredients of NPE were revealed to be nicandrenone, withanolide A, and baicalin. NPE significantly improved lung injury, pulmonary edema, and inflammatory cell infiltration in mice with ALI. In addition, NPE improved autophagic activity and alleviated Th1 and Th17 cell-induced lung inflammation by suppressing the PI3K/Akt/mTOR signaling pathway. Importantly, immunometabolomic analysis of fecal, serum, and lung tissues revealed that NPE reversed ALI-induced leucine resistance by remodeling immunometabolism. We confirmed NPE prevents ALI by remodeling immunometabolism, regulating the Leucine/PI3K/Akt/mTOR signaling pathway, inhibiting Th1/Th17 cell differentiation, and providing a scientific immunological basis for the clinical application of NPE.
Collapse
Affiliation(s)
- Mantong Zhao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jiazi Lin
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xiao Wang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Chengkai Chen
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jianhua Li
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jiamin Yu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Tong Zhou
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yefang Liang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xuejuan Shen
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Ruixiang Shi
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Simin Yang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Shuting Zeng
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yongan Deng
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Xiaodong Duan
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China; School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Lichang Zhou
- Ruyuan Yao Autonomous County Agricultural Technology Promotion Center, Shaoguan, 512700, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China.
| | - Yi Wang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Zunpeng Shu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, 519087, China.
| |
Collapse
|
7
|
Wang R, Ma S, Yang J, Luo K, Qian Q, Pan J, Liang K, Wang Y, Gao Y, Li M. Sodium Hydrosulfide Protects Rats from Hypobaric-Hypoxia-Induced Acute Lung Injury. Int J Mol Sci 2024; 25:10734. [PMID: 39409062 PMCID: PMC11477091 DOI: 10.3390/ijms251910734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
Hydrogen sulfide (H2S), as a key gas signaling molecule, plays an important role in regulating various diseases, with appropriate concentrations providing antioxidative, anti-inflammatory, and anti-apoptotic effects. The specific role of H2S in acute hypoxic injury remains to be clarified. This study focuses on the H2S donor sodium hydrosulfide (NaHS) and explores its protective effects and mechanisms against acute hypoxic lung injury. First, various mouse hypoxia models were established to evaluate H2S's protection in hypoxia tolerance. Next, a rat model of acute lung injury (ALI) induced by hypoxia at 6500 m above sea level for 72 h was created to assess H2S's protective effects and mechanisms. Evaluation metrics included blood gas analysis, blood routine indicators, lung water content, and lung tissue pathology. Additionally, LC-MS/MS and bioinformatic analyses were combined in performing quantitative proteomics on lung tissues from the normoxic control group, the hypoxia model group, and the hypoxia model group with NaHS treatment to preliminarily explore the protective mechanisms of H2S. Further, enzyme-linked immunosorbent assays (ELISA) were used to measure oxidative stress markers and inflammatory factors in rat lung tissues. Lastly, Western blot analysis was performed to detect Nrf2, HO-1, P-NF-κB, NF-κB, HIF-1α, Bcl-2, and Bax proteins in lung tissues. Results showed that H2S exhibited significant anti-hypoxic effects in various hypoxia models, effectively modulating blood gas and blood routine indicators in ALI rats, reducing pulmonary edema, improving lung tissue pathology, and alleviating oxidative stress, inflammatory responses, and apoptosis levels.
Collapse
Affiliation(s)
- Renjie Wang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (R.W.); (S.M.); (J.Y.); (K.L.); (K.L.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
| | - Shuhe Ma
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (R.W.); (S.M.); (J.Y.); (K.L.); (K.L.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
| | - Jun Yang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (R.W.); (S.M.); (J.Y.); (K.L.); (K.L.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
| | - Kai Luo
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (R.W.); (S.M.); (J.Y.); (K.L.); (K.L.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
| | - Qingyuan Qian
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
- College of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jinchao Pan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
- Faculty of Environment and Life, Beijing University of Technology, Beijing 100083, China
| | - Keke Liang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China; (R.W.); (S.M.); (J.Y.); (K.L.); (K.L.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
| | - Yihao Wang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
| | - Yue Gao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
- National Key Laboratory of Kidney Diseases, Beijing 100850, China
| | - Maoxing Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (Q.Q.); (J.P.); (Y.W.)
- National Key Laboratory of Kidney Diseases, Beijing 100850, China
| |
Collapse
|
8
|
Yang F, Chen M, Liu Y, Hu Y, Chen Y, Yu Y, Deng L. ANGPTL2 knockdown induces autophagy to relieve alveolar macrophage pyroptosis by reducing LILRB2-mediated inhibition of TREM2. J Cell Mol Med 2024; 28:e18280. [PMID: 38758159 PMCID: PMC11100552 DOI: 10.1111/jcmm.18280] [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: 09/24/2023] [Revised: 02/22/2024] [Accepted: 03/18/2024] [Indexed: 05/18/2024] Open
Abstract
Acute lung injury (ALI) is featured with a robust inflammatory response. Angiopoietin-like protein 2 (ANGPTL2), a pro-inflammatory protein, is complicated with various disorders. However, the role of ANGPTL2 in ALI remains to be further explored. The mice and MH-S cells were administrated with lipopolysaccharide (LPS) to evoke the lung injury in vivo and in vitro. The role and mechanism of ANGPTL was investigated by haematoxylin-eosin, measurement of wet/dry ratio, cell count, terminal deoxynucleotidyl transferase deoxyuridine triphosphate (dUTP) nick end labeling, reverse transcription quantitative polymerase chain reaction, immunofluorescence, enzyme-linked immunosorbent assay, detection of autophagic flux and western blot assays. The level of ANGPTL2 was upregulated in lung injury. Knockout of ANGPTL2 alleviated LPS-induced pathological symptoms, reduced pulmonary wet/dry weight ratio, the numbers of total cells and neutrophils in BALF, apoptosis rate and the release of pro-inflammatory mediators, and modulated polarization of alveolar macrophages in mice. Knockdown of ANGPTL2 downregulated the level of pyroptosis indicators, and elevated the level of autophagy in LPS-induced MH-S cells. Besides, downregulation of ANGPTL2 reversed the LPS-induced the expression of leukocyte immunoglobulin (Ig)-like receptor B2 (LILRB2) and triggering receptor expressed on myeloid cells 2 (TREM2), which was reversed by the overexpression of LILRB2. Importantly, knockdown of TREM2 reversed the levels of autophagy- and pyroptosis-involved proteins, and the contents of pro-inflammatory factors in LPS-induced MH-S cells transfected with si ANGPTL2, which was further inverted with the treatment of rapamycin. Therefore, ANGPTL2 silencing enhanced autophagy to alleviate alveolar macrophage pyroptosis via reducing LILRB2-mediated inhibition of TREM2.
Collapse
Affiliation(s)
- Fan Yang
- Department of Emergency MedicineThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Muhu Chen
- Department of Emergency MedicineThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Ying Liu
- Department of Emergency MedicineThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Yingchun Hu
- Department of Emergency MedicineThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Yangxi Chen
- Department of Emergency MedicineThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Youwei Yu
- Department of Emergency MedicineThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Lu Deng
- Department of Thyroid SurgeryThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
9
|
Lu X, Li G, Liu Y, Luo G, Ding S, Zhang T, Li N, Geng Q. The role of fatty acid metabolism in acute lung injury: a special focus on immunometabolism. Cell Mol Life Sci 2024; 81:120. [PMID: 38456906 PMCID: PMC10923746 DOI: 10.1007/s00018-024-05131-4] [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: 11/09/2023] [Revised: 01/06/2024] [Accepted: 01/17/2024] [Indexed: 03/09/2024]
Abstract
Reputable evidence from multiple studies suggests that excessive and uncontrolled inflammation plays an indispensable role in mediating, amplifying, and protracting acute lung injury (ALI). Traditionally, immunity and energy metabolism are regarded as separate functions regulated by distinct mechanisms, but recently, more and more evidence show that immunity and energy metabolism exhibit a strong interaction which has given rise to an emerging field of immunometabolism. Mammalian lungs are organs with active fatty acid metabolism, however, during ALI, inflammation and oxidative stress lead to a series metabolic reprogramming such as impaired fatty acid oxidation, increased expression of proteins involved in fatty acid uptake and transport, enhanced synthesis of fatty acids, and accumulation of lipid droplets. In addition, obesity represents a significant risk factor for ALI/ARDS. Thus, we have further elucidated the mechanisms of obesity exacerbating ALI from the perspective of fatty acid metabolism. To sum up, this paper presents a systematical review of the relationship between extensive fatty acid metabolic pathways and acute lung injury and summarizes recent advances in understanding the involvement of fatty acid metabolism-related pathways in ALI. We hold an optimistic believe that targeting fatty acid metabolism pathway is a promising lung protection strategy, but the specific regulatory mechanisms are way too complex, necessitating further extensive and in-depth investigations in future studies.
Collapse
Affiliation(s)
- Xiao Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Guorui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Guoqing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Tianyu Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China.
| |
Collapse
|
10
|
Camporota L, Rose L, Andrews PL, Nieman GF, Habashi NM. Airway pressure release ventilation for lung protection in acute respiratory distress syndrome: an alternative way to recruit the lungs. Curr Opin Crit Care 2024; 30:76-84. [PMID: 38085878 DOI: 10.1097/mcc.0000000000001123] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
PURPOSE OF REVIEW Airway pressure release ventilation (APRV) is a modality of ventilation in which high inspiratory continuous positive airway pressure (CPAP) alternates with brief releases. In this review, we will discuss the rationale for APRV as a lung protective strategy and then provide a practical introduction to initiating APRV using the time-controlled adaptive ventilation (TCAV) method. RECENT FINDINGS APRV using the TCAV method uses an extended inspiratory time and brief expiratory release to first stabilize and then gradually recruit collapsed lung (over hours/days), by progressively 'ratcheting' open a small volume of collapsed tissue with each breath. The brief expiratory release acts as a 'brake' preventing newly recruited units from re-collapsing, reversing the main drivers of ventilator-induced lung injury (VILI). The precise timing of each release is based on analysis of expiratory flow and is set to achieve termination of expiratory flow at 75% of the peak expiratory flow. Optimization of the release time reflects the changes in elastance and, therefore, is personalized (i.e. conforms to individual patient pathophysiology), and adaptive (i.e. responds to changes in elastance over time). SUMMARY APRV using the TCAV method is a paradigm shift in protective lung ventilation, which primarily aims to stabilize the lung and gradually reopen collapsed tissue to achieve lung homogeneity eliminating the main mechanistic drivers of VILI.
Collapse
Affiliation(s)
- Luigi Camporota
- Department of Critical Care, Guy's & St Thomas' NHS Foundation Trust
- Centre for Human & Applied Physiological Sciences, School of Basic & Medical Biosciences
| | - Louise Rose
- Department of Critical Care, Guy's & St Thomas' NHS Foundation Trust
- Florence Nightingale Faculty of Nursing, Midwifery, and Palliative Care, King's College London, London, UK
| | - Penny L Andrews
- Department of Critical Care, R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, Maryland
| | - Gary F Nieman
- Department of Surgery, Upstate Medical University, Syracuse, New York, USA
| | - Nader M Habashi
- Department of Critical Care, R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, Maryland
| |
Collapse
|
11
|
Wang GY, Xu X, Xiong DY, Deng L, Liu W, Huang XT. CPT1A as a potential therapeutic target for lipopolysaccharide-induced acute lung injury in mice. Sci Rep 2024; 14:1600. [PMID: 38238472 PMCID: PMC10796431 DOI: 10.1038/s41598-024-52042-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 01/12/2024] [Indexed: 01/22/2024] Open
Abstract
Acute lung injury (ALI) remains a high mortality rate with dramatic lung inflammation and alveolar epithelial cell death. Although fatty acid β-oxidation (FAO) impairment has been implicated in the pathogenesis of ALI, whether Carnitine palmitoyltransferase 1A (CPT1A), the rate-limiting enzyme for FAO, plays roles in lipopolysaccharide (LPS)-induced ALI remains unclear. Accordingly, we focused on exploring the effect of CPT1A in the context of ALI and the underlying mechanisms. We found that overexpression of CPT1A (AAV-CPT1A) effectively alleviated lung injury by reduction of lung wet-to-dry ratio, inflammatory cell infiltration, and protein levels in the BALF of ALI mice. Meanwhile, AAV-CPT1A significantly lessened histopathological changes and several cytokines' secretions. In contrast, blocking CPT1A with etomoxir augmented inflammatory responses and lung injury in ALI mice. Furthermore, we found that overexpression of CPT1A with lentivirus reduced the apoptosis rates of alveolar epithelial cells and the expression of apoptosis-related proteins induced by LPS in MLE12 cells, while etomoxir increased the apoptosis of MLE12 cells. Overexpression of CPT1A prevented the drop in bioenergetics, palmitate oxidation, and ATP levels. In conclusion, the results rendered CPT1A worthy of further development into a pharmaceutical drug for the treatment of ALI.
Collapse
Affiliation(s)
- Gui-Yun Wang
- Shandong Xiehe University, Jinan, 250109, Shandong, China
| | - Xia Xu
- Shandong Xiehe University, Jinan, 250109, Shandong, China
| | - Da-Yan Xiong
- Xiangya School of Nursing, Central South University, Changsha, 410013, Hunan, China
| | - Lang Deng
- Xiangya School of Nursing, Central South University, Changsha, 410013, Hunan, China
| | - Wei Liu
- Xiangya School of Nursing, Central South University, Changsha, 410013, Hunan, China
| | - Xiao-Ting Huang
- Xiangya School of Nursing, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
12
|
Al-Khalisy H, Nieman GF, Kollisch-Singule M, Andrews P, Camporota L, Shiber J, Manougian T, Satalin J, Blair S, Ghosh A, Herrmann J, Kaczka DW, Gaver DP, Bates JHT, Habashi NM. Time-Controlled Adaptive Ventilation (TCAV): a personalized strategy for lung protection. Respir Res 2024; 25:37. [PMID: 38238778 PMCID: PMC10797864 DOI: 10.1186/s12931-023-02615-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 11/25/2023] [Indexed: 01/22/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) alters the dynamics of lung inflation during mechanical ventilation. Repetitive alveolar collapse and expansion (RACE) predisposes the lung to ventilator-induced lung injury (VILI). Two broad approaches are currently used to minimize VILI: (1) low tidal volume (LVT) with low-moderate positive end-expiratory pressure (PEEP); and (2) open lung approach (OLA). The LVT approach attempts to protect already open lung tissue from overdistension, while simultaneously resting collapsed tissue by excluding it from the cycle of mechanical ventilation. By contrast, the OLA attempts to reinflate potentially recruitable lung, usually over a period of seconds to minutes using higher PEEP used to prevent progressive loss of end-expiratory lung volume (EELV) and RACE. However, even with these protective strategies, clinical studies have shown that ARDS-related mortality remains unacceptably high with a scarcity of effective interventions over the last two decades. One of the main limitations these varied interventions demonstrate to benefit is the observed clinical and pathologic heterogeneity in ARDS. We have developed an alternative ventilation strategy known as the Time Controlled Adaptive Ventilation (TCAV) method of applying the Airway Pressure Release Ventilation (APRV) mode, which takes advantage of the heterogeneous time- and pressure-dependent collapse and reopening of lung units. The TCAV method is a closed-loop system where the expiratory duration personalizes VT and EELV. Personalization of TCAV is informed and tuned with changes in respiratory system compliance (CRS) measured by the slope of the expiratory flow curve during passive exhalation. Two potentially beneficial features of TCAV are: (i) the expiratory duration is personalized to a given patient's lung physiology, which promotes alveolar stabilization by halting the progressive collapse of alveoli, thereby minimizing the time for the reopened lung to collapse again in the next expiration, and (ii) an extended inspiratory phase at a fixed inflation pressure after alveolar stabilization gradually reopens a small amount of tissue with each breath. Subsequently, densely collapsed regions are slowly ratcheted open over a period of hours, or even days. Thus, TCAV has the potential to minimize VILI, reducing ARDS-related morbidity and mortality.
Collapse
Affiliation(s)
| | - Gary F Nieman
- SUNY Upstate Medical University, 750 E. Adams St., Syracuse, NY, 13210, USA
| | | | - Penny Andrews
- R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, MD, USA
| | - Luigi Camporota
- Health Centre for Human and Applied Physiological Sciences, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Joseph Shiber
- University of Florida College of Medicine, Jacksonville, FL, USA
| | | | - Joshua Satalin
- SUNY Upstate Medical University, 750 E. Adams St., Syracuse, NY, 13210, USA.
| | - Sarah Blair
- SUNY Upstate Medical University, 750 E. Adams St., Syracuse, NY, 13210, USA
| | - Auyon Ghosh
- SUNY Upstate Medical University, 750 E. Adams St., Syracuse, NY, 13210, USA
| | | | | | | | | | - Nader M Habashi
- R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, MD, USA
| |
Collapse
|
13
|
Ma H, Fujioka H, Halpern D, Bates JHT, Gaver DP. Full-lung simulations of mechanically ventilated lungs incorporating recruitment/derecruitment dynamics. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:1257710. [PMID: 38020240 PMCID: PMC10654632 DOI: 10.3389/fnetp.2023.1257710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023]
Abstract
This study developed and investigated a comprehensive multiscale computational model of a mechanically ventilated ARDS lung to elucidate the underlying mechanisms contributing to the development or prevention of VILI. This model is built upon a healthy lung model that incorporates realistic airway and alveolar geometry, tissue distensibility, and surfactant dynamics. Key features of the ARDS model include recruitment and derecruitment (RD) dynamics, alveolar tissue viscoelasticity, and surfactant deficiency. This model successfully reproduces realistic pressure-volume (PV) behavior, dynamic surface tension, and time-dependent descriptions of RD events as a function of the ventilation scenario. Simulations of Time-Controlled Adaptive Ventilation (TCAV) modes, with short and long durations of exhalation (T Low - and T Low +, respectively), reveal a higher incidence of RD for T Low + despite reduced surface tensions due to interfacial compression. This finding aligns with experimental evidence emphasizing the critical role of timing in protective ventilation strategies. Quantitative analysis of energy dissipation indicates that while alveolar recruitment contributes only a small fraction of total energy dissipation, its spatial concentration and brief duration may significantly contribute to VILI progression due to its focal nature and higher intensity. Leveraging the computational framework, the model may be extended to facilitate the development of personalized protective ventilation strategies to enhance patient outcomes. As such, this computational modeling approach offers valuable insights into the complex dynamics of VILI that may guide the optimization of ventilation strategies in ARDS management.
Collapse
Affiliation(s)
- Haoran Ma
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, United States
| | - Hideki Fujioka
- Center for Computational Science, Tulane University, New Orleans, LA, United States
| | - David Halpern
- Department of Mathematics, University of Alabama, Tuscaloosa, AL, United States
| | - Jason H. T. Bates
- Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Donald P. Gaver
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, United States
| |
Collapse
|
14
|
Zhang S, Hu D, Zhuo Y, Cui L, Li D, Zhang L, Yang L, Wang X. Protective effect of liriodendrin on IgG immune complex-induced acute lung injury via inhibiting SRC/STAT3/MAPK signaling pathway: a network pharmacology research. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3269-3283. [PMID: 37243760 DOI: 10.1007/s00210-023-02534-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/16/2023] [Indexed: 05/29/2023]
Abstract
The primary objectives of this research were to investigate the protective effects of liriodendrin against IgG immune complex (IgG-IC)-induced acute lung injury (ALI) and to elucidate the underlying mechanisms. This study employed a mouse and cell model of IgG-IC-induced acute lung injury. Lung tissue was stained with hematoxylin-eosin to observe pathological alterations and arterial blood gas analysis was tested. Inflammatory cytokines, including interleukin-6 (IL-6), interleukin-1β (IL-1β), and tumor necrosis factor-alpha (TNF-α), were measured using ELISA. The mRNA expression of inflammatory cytokines was assessed via RT-qPCR. Molecular docking and enrichment analysis were combined to identify the most potential signaling pathways modulated by liriodendrin, which were then verified using western blot analysis in IgG-IC-induced ALI models. We identified 253 shared targets between liriodendrin and IgG-IC-induced acute lung injury from the database. Through network pharmacology, enrichment analysis, and molecular docking, SRC was determined to be the most closely associated target of liriodendrin in IgG-IC-induced ALI. Pretreatment with liriodendrin notably reduced the increased cytokine secretion of IL-1β, IL-6, and TNF-α. Histopathological analysis of lung tissue demonstrated a protective effect of liriodendrin on IgG-IC-induced acute lung injury in mice. Arterial blood gas analysis showed liriodendrin ameliorated acidosis and hypoxemia efficiently. Further studies revealed that liriodendrin pretreatment substantially attenuated the elevated phosphorylation levels of SRC's downstream components (JNK, P38, and STAT3), suggesting that liriodendrin may protect against IgG-IC-induced ALI via the SRC/STAT3/MAPK pathway. Our findings indicate that liriodendrin protects against IgG-IC-induced acute lung injury by inhibiting the SRC/STAT3/MAPK signaling pathway, suggesting that liriodendrin may serve as a potential treatment for acute lung injury caused by IgG-IC.
Collapse
Affiliation(s)
- Sijia Zhang
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Dongsheng Hu
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Yuzhen Zhuo
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China
| | - Lingzhi Cui
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Dihua Li
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China
| | - Lanqiu Zhang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China
| | - Lei Yang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China.
| | - Ximo Wang
- Graduate School, Tianjin Medical University, Tianjin, China.
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China.
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin University, Tianjin, China.
| |
Collapse
|
15
|
Fawley JA, Tignanelli CJ, Werner NL, Kasotakis G, Mandell SP, Glass NE, Dries DJ, Costantini TW, Napolitano LM. American Association for the Surgery of Trauma/American College of Surgeons Committee on Trauma clinical protocol for management of acute respiratory distress syndrome and severe hypoxemia. J Trauma Acute Care Surg 2023; 95:592-602. [PMID: 37314843 PMCID: PMC10545067 DOI: 10.1097/ta.0000000000004046] [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: 03/12/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 06/15/2023]
Abstract
LEVEL OF EVIDENCE Therapeutic/Care Management: Level V.
Collapse
|
16
|
Nieman GF, Kaczka DW, Andrews PL, Ghosh A, Al-Khalisy H, Camporota L, Satalin J, Herrmann J, Habashi NM. First Stabilize and then Gradually Recruit: A Paradigm Shift in Protective Mechanical Ventilation for Acute Lung Injury. J Clin Med 2023; 12:4633. [PMID: 37510748 PMCID: PMC10380509 DOI: 10.3390/jcm12144633] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is associated with a heterogeneous pattern of injury throughout the lung parenchyma that alters regional alveolar opening and collapse time constants. Such heterogeneity leads to atelectasis and repetitive alveolar collapse and expansion (RACE). The net effect is a progressive loss of lung volume with secondary ventilator-induced lung injury (VILI). Previous concepts of ARDS pathophysiology envisioned a two-compartment system: a small amount of normally aerated lung tissue in the non-dependent regions (termed "baby lung"); and a collapsed and edematous tissue in dependent regions. Based on such compartmentalization, two protective ventilation strategies have been developed: (1) a "protective lung approach" (PLA), designed to reduce overdistension in the remaining aerated compartment using a low tidal volume; and (2) an "open lung approach" (OLA), which first attempts to open the collapsed lung tissue over a short time frame (seconds or minutes) with an initial recruitment maneuver, and then stabilize newly recruited tissue using titrated positive end-expiratory pressure (PEEP). A more recent understanding of ARDS pathophysiology identifies regional alveolar instability and collapse (i.e., hidden micro-atelectasis) in both lung compartments as a primary VILI mechanism. Based on this understanding, we propose an alternative strategy to ventilating the injured lung, which we term a "stabilize lung approach" (SLA). The SLA is designed to immediately stabilize the lung and reduce RACE while gradually reopening collapsed tissue over hours or days. At the core of SLA is time-controlled adaptive ventilation (TCAV), a method to adjust the parameters of the airway pressure release ventilation (APRV) modality. Since the acutely injured lung at any given airway pressure requires more time for alveolar recruitment and less time for alveolar collapse, SLA adjusts inspiratory and expiratory durations and inflation pressure levels. The TCAV method SLA reverses the open first and stabilize second OLA method by: (i) immediately stabilizing lung tissue using a very brief exhalation time (≤0.5 s), so that alveoli simply do not have sufficient time to collapse. The exhalation duration is personalized and adaptive to individual respiratory mechanical properties (i.e., elastic recoil); and (ii) gradually recruiting collapsed lung tissue using an inflate and brake ratchet combined with an extended inspiratory duration (4-6 s) method. Translational animal studies, clinical statistical analysis, and case reports support the use of TCAV as an efficacious lung protective strategy.
Collapse
Affiliation(s)
- Gary F. Nieman
- Department of Surgery, Upstate Medical University, Syracuse, NY 13210, USA;
| | - David W. Kaczka
- Departments of Anesthesia, Radiology and Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Penny L. Andrews
- Department of Medicine, R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, MD 21201, USA
| | - Auyon Ghosh
- Department of Medicine, Upstate Medical University, Syracuse, NY 13210, USA
| | - Hassan Al-Khalisy
- Brody School of Medicine, Department of Internal Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Luigi Camporota
- Department of Adult Critical Care, Guy’s and St Thomas’ NHS Foundation Trust, King’s Partners, St Thomas’ Hospital, London SE1 7EH, UK
| | - Joshua Satalin
- Department of Surgery, Upstate Medical University, Syracuse, NY 13210, USA;
| | - Jacob Herrmann
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Nader M. Habashi
- Department of Medicine, R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, MD 21201, USA
| |
Collapse
|
17
|
Shi K, Wang Y, Xiao Y, Tu J, Zhou Z, Cao G, Liu Y. Therapeutic effects and mechanism of Atractylodis rhizoma in acute lung injury: Investigation based on an Integrated approach. Front Pharmacol 2023; 14:1181951. [PMID: 37168993 PMCID: PMC10164760 DOI: 10.3389/fphar.2023.1181951] [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: 03/08/2023] [Accepted: 04/11/2023] [Indexed: 05/13/2023] Open
Abstract
Acute lung injury (ALI) is characterized by an excessive inflammatory response. Atractylodes lancea (Thunb.) DC. is a traditional chinese medicine with good anti-inflammatory activity that is commonly used clinically for the treatment of lung diseases in China; however, its mechanism of against ALI is unclear. We clarified the therapeutic effects of ethanol extract of Atractylodis rhizoma (EEAR) on lipopolysaccharide (LPS)-induced ALI by evaluation of hematoxylin-eosin (HE) stained sections, the lung wet/dry (W/D) ratio, and levels of inflammatory factors as indicators. We then characterized the chemical composition of EEAR by ultra-performance liquid chromatography and mass spectrometry (UPLC-MS) and screened the components and targets by network pharmacology to clarify the signaling pathways involved in the therapeutic effects of EEAR on ALI, and the results were validated by molecular docking simulation and Western blot (WB) analysis. Finally, we examined the metabolites in rat lung tissues by gas chromatography and mass spectrometry (GC-MS). The results showed that EEAR significantly reduced the W/D ratio, and tumor necrosis factor-α (TNF-α), interleukin-1 beta (IL-1β), interleukin-6 (IL-6) levels in the lungs of ALI model rats. Nineteen components of EEAR were identified and shown to act synergetically by regulating shared pathways such as the mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K)-protein kinase B (AKT) signaling pathways. Ferulic acid, 4-methylumbelliferone, acetylatractylodinol, atractylenolide I, and atractylenolide III were predicted to bind well to PI3K, AKT and MAPK1, respectively, with binding energies < -5 kcal/mol, although only atractylenolide II bound with high affinity to MAPK1. EEAR significantly inhibited the phosphorylation of PI3K, AKT, p38, and ERK1/2, thus reducing protein expression. EEAR significantly modulated the expression of metabolites such as D-Galactose, D-Glucose, serine and D-Mannose. These metabolites were mainly concentrated in the galactose and amino acid metabolism pathways. In conclusion, EEAR alleviates ALI by inhibiting activation of the PI3K-AKT and MAPK signaling pathways and regulating galactose metabolism, providing a new direction for the development of drugs to treat ALI.
Collapse
Affiliation(s)
- Kun Shi
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Yan Wang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Yangxin Xiao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Jiyuan Tu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Center for Hubei TCM Processing Technology Engineering, Wuhan, China
| | - Zhongshi Zhou
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Center for Hubei TCM Processing Technology Engineering, Wuhan, China
| | - Guosheng Cao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Center for Hubei TCM Processing Technology Engineering, Wuhan, China
| | - Yanju Liu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Center for Hubei TCM Processing Technology Engineering, Wuhan, China
| |
Collapse
|
18
|
Wang Z, Zhang L, Huang T, Yang R, Cheng H, Wang H, Yin H, Lyu J. Developing an explainable machine learning model to predict the mechanical ventilation duration of patients with ARDS in intensive care units. Heart Lung 2023; 58:74-81. [PMID: 36423504 PMCID: PMC9678346 DOI: 10.1016/j.hrtlng.2022.11.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/25/2022] [Accepted: 11/11/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is common in intensive care units with high mortality rate and mechanical ventilation (MV) is the most important related treatment. Early prediction of MV duration has benefit for patients risk stratification and care strategies support. OBJECTIVE To develop an explainable model for predicting mechanical ventilation (MV) duration in patients with ARDS using the machine learning (ML) approach. METHOD The number of 1,148, 1,697, and 29 ARDS patients admitted to intensive care units (ICU) in the MIMIC-IV, eICU-CRD, and AmsterdamUMCdb databases were included in the study. Features at MV initiation from the MIMIC-IV dataset were used to train prediction models based on seven supervised machine learning algorithms. After 5-fold cross-validation for hyperparameters tuning, the hyperparameters- optimized model of different algorithms was tested by external datasets extracted from eICU-CRD and Amsterdamumcdb. Finally, three descriptive machine learning explanation methods were conducted for the model explanation. RESULT The XGBoosting model showed the most stable and accurate performance among two testing datasets (RMSE= 5.57 and 5.46 days in eICU-CRD and AmsterdamUMCdb) and was selected as the optimal model. The model explanation based on SHAP, LIME, and DALEX results showed a consistent result, vasopressor, PH, and SOFA score had the highest effect on MV duration prediction. CONCLUSION ML models with features at MV initiation can accurate predict MV duration in patients with ARDS in ICUs. Among seven algorithms, XGB models showed the best performance (RMSE= 5.57 and 5.46 in two external datasets). LIME, SHAP, and Breakdown methods showed good performance as AXI methods.
Collapse
Affiliation(s)
- Zichen Wang
- Department of Intensive Care Unit, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China; Department of Public Health, University of California, Irvine, Irvine, California, United State
| | - Luming Zhang
- Department of Intensive Care Unit, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China; Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Tao Huang
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Rui Yang
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China; School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Hongtao Cheng
- School of Nursing, Jinan University, Guangzhou, China
| | - Hao Wang
- Department of Statistics, Iowa State University, Ames, Iowa, Unite States
| | - Haiyan Yin
- Department of Intensive Care Unit, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, Guangdong, China.
| |
Collapse
|
19
|
Yamano S, Takeda T, Goto Y, Hirai S, Furukawa Y, Kikuchi Y, Misumi K, Suzuki M, Takanobu K, Senoh H, Saito M, Kondo H, Kobashi Y, Okamoto K, Kishimoto T, Umeda Y. Mechanisms of pulmonary disease in F344 rats after workplace-relevant inhalation exposure to cross-linked water-soluble acrylic acid polymers. Respir Res 2023; 24:47. [PMID: 36782232 PMCID: PMC9926550 DOI: 10.1186/s12931-023-02355-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 02/01/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Recently in Japan, six workers at a chemical plant that manufactures resins developed interstitial lung diseases after being involved in loading and packing cross-linked water-soluble acrylic acid polymers (CWAAPs). The present study focused on assessing lung damage in rats caused by workplace-relevant inhalation exposure to CWAAP and investigated the molecular and cellular mechanisms involved in lung lesion development. METHODS Using a whole-body inhalation exposure system, male F344 rats were exposed once to 40 or 100 mg/m3 of CWAAP-A for 4 h or to 15 or 40 mg/m3 of CWAAP-A for 4 h per day once per week for 2 months (9 exposures). In a separate set of experiments, male F344 rats were administered 1 mg/kg CWAAP-A or CWAAP-B by intratracheal instillation once every 2 weeks for 2 months (5 doses). Lung tissues, mediastinal lymph nodes, and bronchoalveolar lavage fluid were collected and subjected to biological and histopathological analyses. RESULTS A single 4-h exposure to CWAAP-A caused alveolar injury, and repeated exposures resulted in regenerative changes in the alveolar epithelium with activation of TGFβ signaling. During the recovery period after the last exposure, some alveolar lesions were partially healed, but other lesions developed into alveolitis with fibrous thickening of the alveolar septum. Rats administered CWAAP-A by intratracheal instillation developed qualitatively similar pulmonary pathology as rats exposed to CWAAP-A by inhalation. At 2 weeks after intratracheal instillation, rats administered CWAAP-B appeared to have a slightly higher degree of lung lesions compared to rats administered CWAAP-A, however, there was no difference in pulmonary lesions in the CWAAP-A and CWAAP-B exposed rats examined 18 weeks after administration of these materials. CONCLUSIONS The present study reports our findings on the cellular and molecular mechanisms of pulmonary disease in rats after workplace-relevant inhalation exposure to CWAAP-A. This study also demonstrates that the lung pathogenesis of rats exposed to CWAAP-A by systemic inhalation was qualitatively similar to that of rats administered CWAAP-A by intratracheal instillation.
Collapse
Affiliation(s)
- Shotaro Yamano
- Japan Bioassay Research Center, Japan Organization of Occupational Health and Safety, Hadano, Kanagawa, 257-0015, Japan.
| | - Tomoki Takeda
- Japan Bioassay Research Center, Japan Organization of Occupational Health and Safety, Hadano, Kanagawa, 257-0015, Japan.
| | - Yuko Goto
- Japan Bioassay Research Center, Japan Organization of Occupational Health and Safety, Hadano, Kanagawa, 257-0015, Japan
| | - Shigeyuki Hirai
- Japan Bioassay Research Center, Japan Organization of Occupational Health and Safety, Hadano, Kanagawa, 257-0015, Japan
| | - Yusuke Furukawa
- Japan Bioassay Research Center, Japan Organization of Occupational Health and Safety, Hadano, Kanagawa, 257-0015, Japan
| | - Yoshinori Kikuchi
- Japan Bioassay Research Center, Japan Organization of Occupational Health and Safety, Hadano, Kanagawa, 257-0015, Japan
| | - Kyohei Misumi
- Japan Bioassay Research Center, Japan Organization of Occupational Health and Safety, Hadano, Kanagawa, 257-0015, Japan
| | - Masaaki Suzuki
- Japan Bioassay Research Center, Japan Organization of Occupational Health and Safety, Hadano, Kanagawa, 257-0015, Japan
| | - Kenji Takanobu
- Japan Bioassay Research Center, Japan Organization of Occupational Health and Safety, Hadano, Kanagawa, 257-0015, Japan
| | - Hideki Senoh
- Japan Bioassay Research Center, Japan Organization of Occupational Health and Safety, Hadano, Kanagawa, 257-0015, Japan
| | - Misae Saito
- Japan Bioassay Research Center, Japan Organization of Occupational Health and Safety, Hadano, Kanagawa, 257-0015, Japan
| | - Hitomi Kondo
- Japan Bioassay Research Center, Japan Organization of Occupational Health and Safety, Hadano, Kanagawa, 257-0015, Japan
| | - Yoichiro Kobashi
- Department of Pathology, Tenri Hospital, Tenri, Nara, 632-8552, Japan
| | - Kenzo Okamoto
- Department of Pathology, Hokkaido Chuo Rosai Hospital, Japan Organization of Occupational Health and Safety, Iwamizawa, Hokkaido, 068-0004, Japan
| | - Takumi Kishimoto
- Director of Research and Training Center for Asbestos-Related Diseases, Okayama, Okayama, 702-8055, Japan
| | - Yumi Umeda
- Japan Bioassay Research Center, Japan Organization of Occupational Health and Safety, Hadano, Kanagawa, 257-0015, Japan
| |
Collapse
|
20
|
Roshdy A, Elsayed AS, Saleh AS. Airway Pressure Release Ventilation for Acute Respiratory Failure Due to Coronavirus Disease 2019: A Systematic Review and Meta-Analysis. J Intensive Care Med 2023; 38:160-168. [PMID: 35733377 DOI: 10.1177/08850666221109779] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Objective: To explore the evidence surrounding the use of Airway Pressure Release Ventilation (APRV) in patients with coronavirus disease 2019 (COVID-19). Methods: A Systematic electronic search of PUBMED, EMBASE, and the WHO COVID-19 database. We also searched the grey literature via Google and preprint servers (medRxive and research square). Eligible studies included randomised controlled trials and observational studies comparing APRV to conventional mechanical ventilation (CMV) in adults with acute hypoxemic respiratory failure due to COVID-19 and reporting at least one of the following outcomes; in-hospital mortality, ventilator free days (VFDs), ICU length of stay (LOS), changes in gas exchange parameters, and barotrauma. Two authors independently screened and selected articles for inclusion and extracted data in a pre-specified form. Results: Of 181 articles screened, seven studies (one randomised controlled trial, two cohort studies, and four before-after studies) were included comprising 354 patients. APRV was initiated at a mean of 1.2-13 days after intubation. APRV wasn't associated with improved mortality compared to CMV (relative risk [RR], 1.20; 95% CI 0.70-2.05; I2, 61%) neither better VFDs (ratio of means [RoM], 0.80; 95% CI, 0.52-1.24; I2, 0%) nor ICU LOS (RoM, 1.10; 95% CI, 0.79-1.51; I2, 57%). Compared to CMV, APRV was associated with a 33% increase in PaO2/FiO2 ratio (RoM, 1.33; 95% CI, 1.21-1.48; I2, 29%) and a 9% decrease in PaCO2 (RoM, 1.09; 95% CI, 1.02-1.15; I2, 0%). There was no significant increased risk of barotrauma compared to CMV (RR, 1.55; 95% CI, 0.60-4.00; I2, 0%). Conclusions: In adult patients with COVID-19 requiring mechanical ventilation, APRV is associated with improved gas exchange but not mortality nor VFDs when compared with CMV. The results were limited by high uncertainty given the low quality of the available studies and limited number of patients. Adequately powered and well-designed clinical trials to define the role of APRV in COVID-19 patients are still needed. Registration: PROSPERO; CRD42021291234.
Collapse
Affiliation(s)
- Ashraf Roshdy
- Critical Care Medicine Department, Faculty of Medicine, 54562Alexandria University, Alexandria, Egypt.,Intensive Care Unit, 156506William Harvey Hospital, East Kent Hospitals University NHS Foundation Trust, Kent, UK
| | - Ahmad Samy Elsayed
- Intensive Care Unit, 37841King Fahd Military Medical Complex, Dhahran, Saudi Arabia
| | | |
Collapse
|
21
|
He W, Dong H, Wu C, Zhong Y, Li J. The role of NLRP3 inflammasome in sepsis: A potential therapeutic target. Int Immunopharmacol 2023; 115:109697. [PMID: 37724951 DOI: 10.1016/j.intimp.2023.109697] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/28/2022] [Accepted: 01/03/2023] [Indexed: 01/22/2023]
Abstract
Sepsis is the host immune imbalance following infection and leads to organ dysfunction, with highly complicated pathophysiology. To date, sepsis still lacks effective therapies with high mortality rates. Recently, numerous studies have highlighted the potential of NLRP3 inflammasome as a therapeutic target during sepsis. NLRP3 inflammasome is a protein complex that could induce the activation of caspase-1 and the following release of pro-inflammatory cytokines such as IL-1β and IL-18. It was demonstrated that NLRP3 inflammasome was involved in the development and progression of sepsis. In contrast, inhibition of NLRP3 inflammasome activation could mitigate the inflammatory response, protect organ function, and improve outcomes and mortality. This paper illustrated the activation pathways of the NLRP3 inflammasome and its possible molecular mechanisms in the pathophysiology of sepsis. Meanwhile, the beneficial effects of inhibiting NLRP3 activation in sepsis-related organ damage were also presented. In addition, the diverse role of NLRP3 inflammasome in bacterial clearance was addressed. Of note, several herbal extracts targeting NLRP3 inflammasome in the treatment of sepsis were emphasized. We hope that this paper could provide a basis for further drug research targeting NLRP3 inflammasome.
Collapse
Affiliation(s)
- Wenfang He
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Haiyun Dong
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chenfang Wu
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yanjun Zhong
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jinxiu Li
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
22
|
Wu X, Wu L, Wu Y, Chen W, Chen J, Gong L, Yu J. Heme oxygenase-1 ameliorates endotoxin-induced acute lung injury by modulating macrophage polarization via inhibiting TXNIP/NLRP3 inflammasome activation. Free Radic Biol Med 2023; 194:12-22. [PMID: 36436727 DOI: 10.1016/j.freeradbiomed.2022.11.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
Acute lung injury (ALI) remains a global public health issue without specific and effective treatment options available in the clinic. Alveolar macrophage polarization is involved in the initiation, development and progression of ALI; however, the underlying mechanism remains poorly understood. Heme oxygenase-1 (HO-1) acts as an antioxidant in pulmonary inflammation and has been demonstrated to be linked with the severity and prognosis of ALI. In this study, the therapeutic effects of HO-1 were examined, along with the mechanisms involved, mainly focusing on alveolar macrophage polarization. HO-1 depletion induced higher iNOS and CD86 (M1 phenotype) expression but was significantly decreased in Arg-1 and CD206 (M2 phenotype) expression in BALF alveolar macrophages after equivalent LPS stimulation. We also found that HO-1 deletion distinctly accelerated the expression of inflammasome-associated components NLRP3, ASC and caspase-1 in vivo and in vivo and in vitro. Moreover, on the basis of LPS for MH-S cells, levels of TXNIP, NLRP3, ASC and caspase-1 were increased and HO-1 depletion exacerbated these changes, whereas double depletion of HO-1 and TXNIP partially mitigated these elevations. Also, HO-1 knockdown induced more M1 phenotype and less M2 phenotype compared with LPS alone, whereas double silence of HO-1 and TXNIP partially changed the polarization state. Taken together, we demonstrated that HO-1 could modulate macrophage polarization via TXNIP/NLRP3 signaling pathway, which could be a potential therapeutic target for ALI treatment.
Collapse
Affiliation(s)
- Xiaoyang Wu
- School of Medicine, Nankai University, Tianjin, China
| | - Lili Wu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Ya Wu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Wei Chen
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Jinkun Chen
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Lirong Gong
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China.
| | - Jianbo Yu
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, School of Medicine, Nankai University, Tianjin, China.
| |
Collapse
|
23
|
Shi K, Xiao Y, Dong Y, Wang D, Xie Y, Tu J, Xu K, Zhou Z, Cao G, Liu Y. Protective Effects of Atractylodis lancea Rhizoma on Lipopolysaccharide-Induced Acute Lung Injury via TLR4/NF-κB and Keap1/Nrf2 Signaling Pathways In Vitro and In Vivo. Int J Mol Sci 2022; 23:16134. [PMID: 36555773 PMCID: PMC9781712 DOI: 10.3390/ijms232416134] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Acute lung injury (ALI) is a syndrome caused by an excessive inflammatory response characterized by intractable hypoxemia both inside and outside the lung, for which effective therapeutic drugs are lacking. Atractylodis rhizoma, a traditional Chinese medicine, has excellent anti-inflammatory and antiviral properties in addition to protecting the integrity of the cellular barrier. However, few studies of Atractylodis rhizoma for the treatment of ALI have been published, and its mechanism of action remains unclear. In the present study, the chemical composition of the ethanolic extract of Atractylodis rhizoma (EEAR) was initially clarified by high performance liquid chromatography (HPLC), after which it was studied in vivo using a lipopolysaccharide (LPS)-induced ALI rat model. Treatment with EEAR significantly reduced the lung wet/dry (W/D) ratio, neutrophil infiltration, and malondialdehyde (MDA) and myeloperoxidase (MPO) formation, and enhanced superoxide dismutase (SOD) and glutathione (GSH) depletion in rats with ALI, thereby improving lung barrier function and effectively reducing lung injury. In addition, EEAR significantly reduced histopathological changes, decreased the expression of inflammatory factors (such as tumor necrosis factor-α (TNF-α), interleukin-1 beta (IL-1β), inducible nitric oxide synthase (INOS), and cyclooxygenase-2 (COX-2)), and inhibited the activation of the NF-κB signaling pathway, thus reducing inflammation. In addition, EEAR was found to also reduce oxidative stress in ALI by upregulating the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream proteins heme oxygenase-1 (HO-1) and NADPH quinone acceptor oxidoreductase 1 (NQO-1). EEAR also reduced LPS-induced inflammatory factor expression in THP-1 cells in vitro by inhibition of the NF-κB signaling pathway, and reduced damage from lipopolysaccharide (LPS)-induced oxidative stress in THP-1 cells by promoting the expression of Nrf2 and its downstream targets HO-1 and NQO-1, the molecular mechanism of which was consistent with in vivo observations. Therefore, we conclude that EEAR attenuates oxidative stress and inflammatory responses via TLR4/NF-κB and Keap1/Nrf2 signaling pathways to alleviate LPS-induced ALI, suggesting that Atractylodis rhizoma is a potential drug candidate for the treatment of ALI.
Collapse
Affiliation(s)
- Kun Shi
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Yangxin Xiao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Yan Dong
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Dongpeng Wang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Ying Xie
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Jiyuan Tu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Center for Hubei TCM Processing Technology Engineering, Wuhan 430065, China
| | - Kang Xu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Center for Hubei TCM Processing Technology Engineering, Wuhan 430065, China
| | - Zhongshi Zhou
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Center for Hubei TCM Processing Technology Engineering, Wuhan 430065, China
| | - Guosheng Cao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Center for Hubei TCM Processing Technology Engineering, Wuhan 430065, China
| | - Yanju Liu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
- Center for Hubei TCM Processing Technology Engineering, Wuhan 430065, China
| |
Collapse
|
24
|
Krygier A, Szmajda-Krygier D, Świechowski R, Pietrzak J, Wosiak A, Wodziński D, Balcerczak E. Molecular Pathogenesis of Fibrosis, Thrombosis and Surfactant Dysfunction in the Lungs of Severe COVID-19 Patients. Biomolecules 2022; 12:1845. [PMID: 36551272 PMCID: PMC9776352 DOI: 10.3390/biom12121845] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
The global scope and scale of the SARS-CoV-2 pandemic led to huge amounts of important data from clinical observations and experimental analyses being collected, in particular, regarding the long-term impact of COVID-19 on lung tissue. Visible changes in lung tissue mainly relate to the destruction of the alveolar architecture, dense cellularity, and pulmonary fibrosis with myofibroblast proliferation and collagen deposition. These changes are the result of infection, mainly with virus variants from the first pandemic waves (Alpha to Delta). In addition, proper regulation of immune responses to pathogenic viral stimuli is critical for the control of and recovery from tissue/organ damage, including in the lungs. We can distinguish three main processes in the lungs during SARS-CoV-2 infection: damage or deficiency of the pulmonary surfactant, coagulation processes, and fibrosis. Understanding the molecular basis of these processes is extremely important in the context of elucidating all pathologies occurring after virus entry. In the present review, data on the abovementioned three biochemical processes that lead to pathological changes are gathered together and discussed. Systematization of the knowledge is necessary to explore the three key pathways in lung tissue after SARS-CoV-2 virus infection as a result of a prolonged and intense inflammatory process in the context of pulmonary fibrosis, hemostatic disorders, and disturbances in the structure and/or metabolism of the surfactant. Despite the fact that the new Omicron variant does not affect the lungs as much as the previous variants, we cannot ignore the fact that other new mutations and emerging variants will not cause serious damage to the lung tissue. In the future, this review will be helpful to stratify the risk of serious complications in patients, to improve COVID-19 treatment outcomes, and to select those who may develop complications before clinical manifestation.
Collapse
Affiliation(s)
| | - Dagmara Szmajda-Krygier
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
| | | | | | | | | | | |
Collapse
|
25
|
Nieman G, Cereda M, Camporota L, Habashi NM. Editorial: Protecting the acutely injured lung: Physiologic, mechanical, inflammatory, and translational perspectives. Front Physiol 2022; 13:1009294. [PMID: 36148299 PMCID: PMC9486833 DOI: 10.3389/fphys.2022.1009294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 08/11/2022] [Indexed: 11/25/2022] Open
Affiliation(s)
- Gary Nieman
- Department of Suregy, Upstate Medical University, Syracuse, NY, United States
| | - Maurizio Cereda
- Department of Anesthesia, Critical Care and Pain Medicine, Harvard Medical School, Boston, MA, United States
| | - Luigi Camporota
- Department of Adult Critical Care, Guy’s and St Thomas’ NHS Foundation Trust, Health Centre for Human and Applied Physiological Sciences, London, United Kingdom
| | - Nader M. Habashi
- 1R Adams Cowley Shock Trauma Center, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
26
|
Kim HY, Kwon S, Um W, Shin S, Kim CH, Park JH, Kim BS. Functional Extracellular Vesicles for Regenerative Medicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106569. [PMID: 35322545 DOI: 10.1002/smll.202106569] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/22/2022] [Indexed: 06/14/2023]
Abstract
The unique biological characteristics and promising clinical potential of extracellular vesicles (EVs) have galvanized EV applications for regenerative medicine. Recognized as important mediators of intercellular communication, naturally secreted EVs have the potential, as innate biotherapeutics, to promote tissue regeneration. Although EVs have emerged as novel therapeutic agents, challenges related to the clinical transition have led to further functionalization. In recent years, various engineering approaches such as preconditioning, drug loading, and surface modification have been developed to potentiate the therapeutic outcomes of EVs. Also, limitations of natural EVs have been addressed by the development of artificial EVs that offer advantages in terms of production yield and isolation methodologies. In this review, an updated overview of current techniques is provided for the functionalization of natural EVs and recent advances in artificial EVs, particularly in the scope of regenerative medicine.
Collapse
Affiliation(s)
- Han Young Kim
- Department of Biomedical-Chemical Engineering, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Seunglee Kwon
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Wooram Um
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Sol Shin
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Republic of Korea
| | - Chan Ho Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jae Hyung Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Republic of Korea
- Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Interdisciplinary Program of Bioengineering, Institute of Chemical Processes, Institute of Engineering Research, BioMAX, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
27
|
Xiang ML, Zhao YL, Liu YY, Yan XJ, Chen S, Luo XD. The phytochemical constituents and protective effect of Fritillaria hupehensis on acute lung injury. Fitoterapia 2022; 162:105283. [PMID: 36007807 DOI: 10.1016/j.fitote.2022.105283] [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] [Received: 06/07/2022] [Revised: 08/16/2022] [Accepted: 08/16/2022] [Indexed: 12/21/2022]
Abstract
Acute lung injury (ALI), a severe respiratory disorder, frequently develops into acute respiratory distress syndrome (ARDS) without timely treatment and scores highly in terms of morbidity and mortality rates. Fritillaria hupehensis is a famous traditional Chinese medicine with antitussive, expectorant and anti-asthmatic effect. Here, the effects of F. hupehensis extracts on lipopolysaccharide (LPS)-induced ALI mice were evaluated for the first time. We showed ethyl acetate fraction (EAF) significantly reduced the leukocytes and neutrophils of bronchoalveolar lavage fluid (BALF) and the lung index as well as pro-inflammatory cytokines (TNF-α and IL-6) of lung homogenates but increasing the anti-inflammatory cytokines (IL-4 and IL-10). Additionally, the alleviation of EAF treatment on lung injury was verified through histopathological observations. Subsequent phytochemical investigation on bioactive fraction led to isolation of 17 compounds including two new, in which compounds 2, 5 and 6 exhibited better anti-inflammatory effect on LPS-induced 16 human airway epithelial (16HBE) cells model by inhibiting the production of CRP and PCT. Furthermore, compound 2 suppressed the LPS-induced upregulation of proteins containing p-p65, COX-2, Caspase-1 and IL-18. In summary, F. hupehensis alleviating LPS-induced ALI in mice may be associated with the anti-inflammatory activity of steroidal alkaloids by suppressing the NF-κB-regulated pro-inflammatory proteins.
Collapse
Affiliation(s)
- Mei-Ling Xiang
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province Yunnan Characteristic Plant Extraction Laboratory, School of Chemical Science and Technology, Yunnan University, Kunmina 650500, PR China
| | - Yun-Li Zhao
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province Yunnan Characteristic Plant Extraction Laboratory, School of Chemical Science and Technology, Yunnan University, Kunmina 650500, PR China
| | - Yang-Yang Liu
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province Yunnan Characteristic Plant Extraction Laboratory, School of Chemical Science and Technology, Yunnan University, Kunmina 650500, PR China
| | - Xiao-Jun Yan
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province Yunnan Characteristic Plant Extraction Laboratory, School of Chemical Science and Technology, Yunnan University, Kunmina 650500, PR China
| | - Song Chen
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province Yunnan Characteristic Plant Extraction Laboratory, School of Chemical Science and Technology, Yunnan University, Kunmina 650500, PR China
| | - Xiao-Dong Luo
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province Yunnan Characteristic Plant Extraction Laboratory, School of Chemical Science and Technology, Yunnan University, Kunmina 650500, PR China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, PR China.
| |
Collapse
|
28
|
Nieman G, Kollisch-Singule M, Ramcharran H, Satalin J, Blair S, Gatto LA, Andrews P, Ghosh A, Kaczka DW, Gaver D, Bates J, Habashi NM. Unshrinking the baby lung to calm the VILI vortex. Crit Care 2022; 26:242. [PMID: 35934707 PMCID: PMC9357329 DOI: 10.1186/s13054-022-04105-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/12/2022] [Indexed: 02/07/2023] Open
Abstract
A hallmark of ARDS is progressive shrinking of the ‘baby lung,’ now referred to as the ventilator-induced lung injury (VILI) ‘vortex.’ Reducing the risk of the VILI vortex is the goal of current ventilation strategies; unfortunately, this goal has not been achieved nor has mortality been reduced. However, the temporal aspects of a mechanical breath have not been considered. A brief expiration prevents alveolar collapse, and an extended inspiration can recruit the atelectatic lung over hours. Time-controlled adaptive ventilation (TCAV) is a novel ventilator approach to achieve these goals, since it considers many of the temporal aspects of dynamic lung mechanics.
Collapse
Affiliation(s)
- Gary Nieman
- Department of Surgery, SUNY Upstate Medical Center, SUNY Upstate, 750 East Adams St., Syracuse, NY, 13210, USA
| | - Michaela Kollisch-Singule
- Department of Surgery, SUNY Upstate Medical Center, SUNY Upstate, 750 East Adams St., Syracuse, NY, 13210, USA
| | - Harry Ramcharran
- Department of Surgery, SUNY Upstate Medical Center, SUNY Upstate, 750 East Adams St., Syracuse, NY, 13210, USA
| | - Joshua Satalin
- Department of Surgery, SUNY Upstate Medical Center, SUNY Upstate, 750 East Adams St., Syracuse, NY, 13210, USA.
| | - Sarah Blair
- Department of Surgery, SUNY Upstate Medical Center, SUNY Upstate, 750 East Adams St., Syracuse, NY, 13210, USA
| | - Louis A Gatto
- Department of Surgery, SUNY Upstate Medical Center, SUNY Upstate, 750 East Adams St., Syracuse, NY, 13210, USA
| | - Penny Andrews
- Department of Medicine, University of Maryland, Baltimore, MD, USA
| | - Auyon Ghosh
- Department of Surgery, SUNY Upstate Medical Center, SUNY Upstate, 750 East Adams St., Syracuse, NY, 13210, USA
| | - David W Kaczka
- Departments of Anesthesia, Biomedical Engineering, and Radiology, University of Iowa, Iowa City, IA, USA
| | - Donald Gaver
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, USA
| | - Jason Bates
- Department of Medicine, University of Vermont, Burlington, VT, USA
| | - Nader M Habashi
- Department of Medicine, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
29
|
Lescroart M, Pequignot B, Bitker L, Pina H, Tran N, Hébert JL, Richard JC, Lévy B, Koszutski M. Time-Controlled Adaptive Ventilation Does Not Induce Hemodynamic Impairment in a Swine ARDS Model. Front Med (Lausanne) 2022; 9:883950. [PMID: 35655856 PMCID: PMC9152423 DOI: 10.3389/fmed.2022.883950] [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: 02/25/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Background The current standard of care during severe acute respiratory distress syndrome (ARDS) is based on low tidal volume (VT) ventilation, at 6 mL/kg of predicted body weight. The time-controlled adaptive ventilation (TCAV) is an alternative strategy, based on specific settings of the airway pressure release ventilation (APRV) mode. Briefly, TCAV reduces lung injury, including: (1) an improvement in alveolar recruitment and homogeneity; (2) reduction in alveolar and alveolar duct micro-strain and stress-risers. TCAV can result in higher intra-thoracic pressures and thus impair hemodynamics resulting from heart-lung interactions. The objective of our study was to compare hemodynamics between TCAV and conventional protective ventilation in a porcine ARDS model. Methods In 10 pigs (63–73 kg), lung injury was induced by repeated bronchial saline lavages followed by 2 h of injurious ventilation. The animals were then randomized into two groups: (1) Conventional protective ventilation with a VT of 6 mL/kg and PEEP adjusted to a plateau pressure set between 28 and 30 cmH2O; (2) TCAV group with P-high set between 27 and 29 cmH2O, P-low at 0 cmH2O, T-low adjusted to terminate at 75% of the expiratory flow peak, and T-high at 3–4 s, with I:E > 6:1. Results Both lung elastance and PaO2:FiO2 were consistent with severe ARDS after 2 h of injurious mechanical ventilation. There was no significant difference in systemic arterial blood pressure, pulmonary blood pressure or cardiac output between Conventional protective ventilation and TCAV. Levels of total PEEP were significantly higher in the TCAV group (p < 0.05). Driving pressure and lung elastance were significantly lower in the TCAV group (p < 0.05). Conclusion No hemodynamic adverse events were observed in the TCAV group compared as to the standard protective ventilation group in this swine ARDS model, and TCAV appeared to be beneficial to the respiratory system.
Collapse
Affiliation(s)
- Mickael Lescroart
- CHRU Nancy, Service de Médecine Intensive et Réanimation, Hôpital Brabois, Vandœuvre-lès-Nancy, France.,INSERM U 1116, Groupe Choc, Équipe 2, Faculté de Médecine, Vandœuvre-lès-Nancy, France.,Université de Lorraine, Faculté de Médecine, Nancy, France
| | - Benjamin Pequignot
- CHRU Nancy, Service de Médecine Intensive et Réanimation, Hôpital Brabois, Vandœuvre-lès-Nancy, France.,INSERM U 1116, Groupe Choc, Équipe 2, Faculté de Médecine, Vandœuvre-lès-Nancy, France.,Université de Lorraine, Faculté de Médecine, Nancy, France
| | - Laurent Bitker
- Service de Médecine Intensive - Réanimation, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France.,Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Héloïse Pina
- CHRU de Nancy, Département D'Anatomie Pathologique, Laboratoires de Biologie Médicale et de Biopathologie, Hôpital Brabois, Vandœuvre-lès-Nancy, France
| | - N'Guyen Tran
- Université de Lorraine, Faculté de Médecine, Nancy, France.,Ecole de Chirurgie, Faculté de Médecine, Université de Lorraine, Nancy, France
| | - Jean-Louis Hébert
- Université Paris XI, Institut de Cardiologie, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Jean-Christophe Richard
- Service de Médecine Intensive - Réanimation, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France.,Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Bruno Lévy
- CHRU Nancy, Service de Médecine Intensive et Réanimation, Hôpital Brabois, Vandœuvre-lès-Nancy, France.,INSERM U 1116, Groupe Choc, Équipe 2, Faculté de Médecine, Vandœuvre-lès-Nancy, France.,Université de Lorraine, Faculté de Médecine, Nancy, France
| | - Matthieu Koszutski
- CHRU Nancy, Service de Médecine Intensive et Réanimation, Hôpital Brabois, Vandœuvre-lès-Nancy, France.,Université de Lorraine, Faculté de Médecine, Nancy, France
| |
Collapse
|
30
|
Rola P, Daxon B. Airway Pressure Release Ventilation With Time-Controlled Adaptive Ventilation (TCAV™) in COVID-19: A Community Hospital's Experience. Front Physiol 2022; 13:787231. [PMID: 35480043 PMCID: PMC9037240 DOI: 10.3389/fphys.2022.787231] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/27/2022] [Indexed: 12/30/2022] Open
Abstract
Santa Cabrini Ospedale, a community hospital in Montreal, Canada, used the airway pressure release ventilation following a time-controlled adaptive ventilation (APRV-TCAV™) approach for several patients in the first wave of the coronavirus disease 2019 (COVID-19) outbreak in the spring of 2021. Based on favorable patient responses, it became the primary mode of invasive mechanical ventilation—from initiation through extubation—during the second and third waves of COVID-19. In this article, we describe our success with APRV-TCAV™ over more conventional modes and protocols and look at three cases that aptly demonstrate our experience. We then outline several risks with our approach and the lessons learned from our experience. While we generally saw improvement in patients’ clinical course with APRV-TCAV™, there are inherent risks with this approach that others must prepare for if they attempt to implement it in their practice.
Collapse
Affiliation(s)
- Philippe Rola
- Intensive Care Unit, Santa Cabrini Hospital, Montreal, QC, Canada
| | | |
Collapse
|
31
|
Stabilization of the floating sternocostal segment of the chest with multiple bilateral fractures of the ribs and the manubrium of the sternum. ACTA BIOMEDICA SCIENTIFICA 2022. [DOI: 10.29413/abs.2022-7.1.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Among patients with floating rib fractures without pneumo- and hemopneumothorax or after their elimination, the most severe disorders of ventilation and circulation occur in patients with multiple bilateral rib fractures and a fracture of the sternum manubrium with the formation of a floating sternocostal segment of the chest. At the same time, the suction aero- and hemodynamic function of the chest is disturbed, there is pressure on the heart and large vessels. As a result, the efficiency of external respiration progressively decreases, the respiratory muscles are exhausted, which requires an urgent transfer to artificial ventilation of the lungs. The article presents a clinical case of successful treatment of such a chest injury using the author’s technique (Patent No. 2621871 of the Russian Federation). The extrathoracic silicone reinforced splint has two horizontal branches that go around the mammary glands. The splint is attached to the floating sternocostal segment with ligatures passed behind the sternum and laterally – to stable sections of the ribs along the posterior axillary line on both sides. The tire reliably holds the sternocostal segment from paradoxical movements. The tire is removed after 3 weeks. By this time, fibrous calluses are formed in places of fractures of bones and cartilage, and the swelling of the chest wall subsides. Superficial bedsores in the places of fixation of the splint are epithelialized under the scab within 7–8 days. The patient was examined a year later, her condition was satisfactory, she had no complaints, there was no chest deformity. The technique is less traumatic, it is indicated for patients with polytrauma and in other cases.
Collapse
|
32
|
LTB4 Promotes Acute Lung Injury via Upregulating the PLCε-1/TLR4/NF-κB Pathway in One-Lung Ventilation. DISEASE MARKERS 2022; 2022:1839341. [PMID: 35059042 PMCID: PMC8766192 DOI: 10.1155/2022/1839341] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/23/2021] [Indexed: 11/21/2022]
Abstract
Background Mechanical ventilation (MV) can provoke acute lung injury (ALI) by increasing inflammation activation and disrupting the barrier in lung tissues even causing death. However, the inflammation-related molecules and pathways in MV-induced ALI remain largely unknown. Hence, the purposes of this study are to examine the role and mechanism of a novel inflammation-related molecule, leukotriene B4 (LTB4), in ALI. Methods The functions of LTB4 in one-lung ventilation (OLV) model were detected by the loss-of-function experiments. H&E staining was used to examine the pathologic changes of lung tissues. Functionally, PLCε-1 knockdown and Toll-like receptor 4 (TLR4)/NF-κB pathway inhibitor were used to detect the regulatory effects of LTB4 on the phospholipase Cε (PLCε-1)/TLR4/nuclear factor-kappa B (NF-κB) pathway. The levels of genes and proteins were determined by RT-qPCR and western blotting assay. The levels of inflammation cytokines and chemokines were measured by ELISA. Results Here, we found LTA4H, leukotriene B (4) receptor 1 (BLT1), LTB4, and PLCε-1 upregulated in OLV rats and associated with inflammatory activation and lung permeability changes of lung tissues. Inhibition of LTB4 alleviated the OLV-induced ALI by inhibiting inflammatory activation and lung permeability changes of lung tissues. For mechanism analyses, LTB4 promoted OLV-induced ALI by activating the PLCε-1/TLR4/NF-κB pathway. Conclusion LTB4 induced ALI in OLV rats by activating the PLCε-1/TLR4/NF-κB pathway. Our findings might supply a new potential therapeutic for OLV-induced ALI.
Collapse
|
33
|
Beretta E, Romanò F, Sancini G, Grotberg JB, Nieman GF, Miserocchi G. Pulmonary Interstitial Matrix and Lung Fluid Balance From Normal to the Acutely Injured Lung. Front Physiol 2021; 12:781874. [PMID: 34987415 PMCID: PMC8720972 DOI: 10.3389/fphys.2021.781874] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/02/2021] [Indexed: 01/17/2023] Open
Abstract
This review analyses the mechanisms by which lung fluid balance is strictly controlled in the air-blood barrier (ABB). Relatively large trans-endothelial and trans-epithelial Starling pressure gradients result in a minimal flow across the ABB thanks to low microvascular permeability aided by the macromolecular structure of the interstitial matrix. These edema safety factors are lost when the integrity of the interstitial matrix is damaged. The result is that small Starling pressure gradients, acting on a progressively expanding alveolar barrier with high permeability, generate a high transvascular flow that causes alveolar flooding in minutes. We modeled the trans-endothelial and trans-epithelial Starling pressure gradients under control conditions, as well as under increasing alveolar pressure (Palv) conditions of up to 25 cmH2O. We referred to the wet-to-dry weight (W/D) ratio, a specific index of lung water balance, to be correlated with the functional state of the interstitial structure. W/D averages ∼5 in control and might increase by up to ∼9 in severe edema, corresponding to ∼70% loss in the integrity of the native matrix. Factors buffering edemagenic conditions include: (i) an interstitial capacity for fluid accumulation located in the thick portion of ABB, (ii) the increase in interstitial pressure due to water binding by hyaluronan (the "safety factor" opposing the filtration gradient), and (iii) increased lymphatic flow. Inflammatory factors causing lung tissue damage include those of bacterial/viral and those of sterile nature. Production of reactive oxygen species (ROS) during hypoxia or hyperoxia, or excessive parenchymal stress/strain [lung overdistension caused by patient self-induced lung injury (P-SILI)] can all cause excessive inflammation. We discuss the heterogeneity of intrapulmonary distribution of W/D ratios. A W/D ∼6.5 has been identified as being critical for the transition to severe edema formation. Increasing Palv for W/D > 6.5, both trans-endothelial and trans-epithelial gradients favor filtration leading to alveolar flooding. Neither CT scan nor ultrasound can identify this initial level of lung fluid balance perturbation. A suggestion is put forward to identify a non-invasive tool to detect the earliest stages of perturbation of lung fluid balance before the condition becomes life-threatening.
Collapse
Affiliation(s)
- Egidio Beretta
- Department of Medicine and Surgery, School of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Monza, Italy
| | - Francesco Romanò
- Univ. Lille, CNRS, ONERA, Arts et Métiers, Centrale Lille, FRE 2017-LMFL-Laboratoire de Mécanique des Fluides de Lille – Kampé de Fériet, Lille, France
| | - Giulio Sancini
- Department of Medicine and Surgery, School of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Monza, Italy
| | - James B. Grotberg
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Gary F. Nieman
- Department of Surgery, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Giuseppe Miserocchi
- Department of Medicine and Surgery, School of Medicine and Surgery, Università degli Studi di Milano-Bicocca, Monza, Italy
| |
Collapse
|
34
|
Zhang L, Wang Z, Xu F, Ren Y, Wang H, Han D, Lyu J, Yin H. The Role of Glucocorticoids in the Treatment of ARDS: A Multicenter Retrospective Study Based on the eICU Collaborative Research Database. Front Med (Lausanne) 2021; 8:678260. [PMID: 34381796 PMCID: PMC8350484 DOI: 10.3389/fmed.2021.678260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 07/05/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Acute respiratory distress syndrome (ARDS) is a common cause of respiratory failure in patients in intensive care unit (ICU). The therapeutic value of glucocorticoids (GCs) in the prognosis of ARDS remains controversial. The aim of this research is studying the impacts of GCs treatment on ARDS patients in ICU. Methods: We retrospectively studied 2,167 ARDS patients whose data were collected from the public eICU Collaborative Research Database, among which 254 patients who received glucocorticoid (GCs) treatment were 1:1 matched by propensity matching analysis (PSM). The primary outcome was ICU mortality. Every oxygenation index (PaO2/FiO2) measurement before death or ICU discharge was recorded. A joint model (JM) which combined longitudinal sub-model (mixed-effect model) and time-to-event sub-model (Cox regression model) by trajectory functions of PaO2/FiO2 was conducted to determine the effects of GCs treatment on both ICU mortality and PaO2/FiO2 level and further PaO2/FiO2's effect on event status. The marginal structural cox model (MSCM) adjusted the overall PaO2/FiO2 of patients to further validate the results. Results: The result of the survival sub-model showed that GCs treatment was significantly associated with reduced ICU mortality in ARDS patients [HR (95% CI) = 0.642 (0.453, 0.912)], demonstrating that GCs treatment was a protective factor of ICU mortality. In the longitudinal sub-model, GCs treatment was not correlated to the PaO2/FiO2. After adjusted by the JM, the HR of GCs treatment was 0.602 while GCs was still not significantly related to PaO2/FiO2 level. The JM-induced association showed that higher PaO2/FiO2 was a significant protective factor of mortality in ARDS patients and the HR was 0.991 which demonstrated that one level increase of PaO2/FiO2 level decreased 0.9% risk of ICU mortality. MSCM results also show that GCs can improve the prognosis of patients. Conclusion: Rational use of GCs can reduce the ICU mortality of ARDS patients in ICU. In addition to the use of GCs treatment, clinicians should also focus on the shifting trend of PaO2/FiO2 level to provide better conditions for patients' survival.
Collapse
Affiliation(s)
- Luming Zhang
- Intensive Care Unit, The First Affiliated Hospital of Jinan University, Guangzhou, China.,Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zichen Wang
- Department of Public Health, University of California, Irvine, Irvine, CA, United States
| | - Fengshuo Xu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, China.,School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yinlong Ren
- Intensive Care Unit, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Hao Wang
- Department of Statistics, Iowa State University, Ames, IA, United States
| | - Didi Han
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, China.,School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Haiyan Yin
- Intensive Care Unit, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
35
|
Li J, Xue L, Wu Y, Yang Q, Liu D, Yu C, Peng J. STAT3-activated lncRNA XIST accelerates the inflammatory response and apoptosis of LPS-induced acute lung injury. J Cell Mol Med 2021; 25:6550-6557. [PMID: 34114724 PMCID: PMC8278113 DOI: 10.1111/jcmm.16653] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 03/22/2021] [Accepted: 04/28/2021] [Indexed: 12/19/2022] Open
Abstract
Acute lung injury (ALI) is a severe lung respiratory failure characterized by high morbidity and mortality. Novel findings demonstrated the critical roles of long non-coding RNA (lncRNA) in ALI. Here, we tried to investigate the roles and potential mechanism of lncRNA X-inactive specific transcript (XIST) in ALI. Results illustrated that lncRNA XIST was up-regulated in the lipopolysaccharide (LPS)-induced ALI mice models and pulmonary endothelial cells. Biofunctional assays unveiled that knockdown of XIST repressed the inflammatory response and apoptosis in LPS-induced endothelial cells. Mechanistically, XIST acted as the miR-146a-5p sponge to positively regulate STAT3. Moreover, STAT3 combined the promoter region of XIST to accelerate the transcription, constituting the positive feedback loop of XIST/miR-146a-5p/STAT3 in ALI. Collectively, these findings suggested that XIST knockdown attenuates the LPS-induced ALI, providing a potential therapeutic target.
Collapse
Affiliation(s)
- Jun Li
- Department of Thoracic SurgeryThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| | - Lei Xue
- Department of Thoracic SurgeryThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| | - Yunfei Wu
- Department of Thoracic SurgeryThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Qiang Yang
- Department of Thoracic SurgeryThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| | - Degang Liu
- Department of Thoracic SurgeryThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| | - Changhui Yu
- Department of Respiratory and Critical Care MedicineChronic Airways Diseases LaboratoryNanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Jiangzhou Peng
- Department of Thoracic SurgeryThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouChina
| |
Collapse
|
36
|
Wang S, Li Z, Wang X, Zhang S, Gao P, Shi Z. The Role of Pulmonary Surfactants in the Treatment of Acute Respiratory Distress Syndrome in COVID-19. Front Pharmacol 2021; 12:698905. [PMID: 34267664 PMCID: PMC8276044 DOI: 10.3389/fphar.2021.698905] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/17/2021] [Indexed: 12/30/2022] Open
Abstract
Lung alveolar type-II (AT-II) cells produce pulmonary surfactant (PS), consisting of proteins and lipids. The lipids in PS are primarily responsible for reducing the air-fluid surface tension inside the alveoli of the lungs and to prevent atelectasis. The proteins are of two types: hydrophilic and hydrophobic. Hydrophilic surfactants are primarily responsible for opsonisation, thereby protecting the lungs from microbial and environmental contaminants. Hydrophobic surfactants are primarily responsible for respiratory function. Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) enters the lungs through ACE-2 receptors on lungs and replicates in AT-II cells leading to the etiology of Coronavirus disease - 2019 (COVID-19). The SARS-CoV-2 virus damages the AT-II cells and results in decreased production of PS. The clinical symptoms of acute respiratory distress syndrome (ARDS) in COVID-19 patients are like those of neonatal respiratory distress syndrome (NRDS). The PS treatment is first-line treatment option for NRDS and found to be well tolerated in ARDS patients with inconclusive efficacy. Over the past 70°years, a lot of research is underway to produce natural/synthetic PS and developing systems for delivering PS directly to the lungs, in addition to finding the association between PS levels and respiratory illnesses. In the present COVID-19 pandemic situation, the scientific community all over the world is searching for the effective therapeutic options to improve the clinical outcomes. With a strong scientific and evidence-based background on role of PS in lung homeostasis and infection, few clinical trials were initiated to evaluate the functions of PS in COVID-19. Here, we connect the data on PS with reference to pulmonary physiology and infection with its possible therapeutic benefit in COVID-19 patients.
Collapse
Affiliation(s)
- Shengguang Wang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhen Li
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinyu Wang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shiming Zhang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Peng Gao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zuorong Shi
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
37
|
Janssen M, Meeder JHJ, Seghers L, den Uil CA. Time controlled adaptive ventilation™ as conservative treatment of destroyed lung: an alternative to lung transplantation. BMC Pulm Med 2021; 21:176. [PMID: 34022829 PMCID: PMC8140588 DOI: 10.1186/s12890-021-01545-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/16/2021] [Indexed: 02/06/2023] Open
Abstract
Background Acute respiratory distress syndrome (ARDS) often requires controlled ventilation, yielding high mechanical power and possibly further injury. Veno-venous extracorporeal membrane oxygenation (VV-ECMO) can be used as a bridge to recovery, however, if this fails the end result is destroyed lung parenchyma. This condition is fatal and the only remaining alternative is lung transplantation. In the case study presented in this paper, lung transplantation was not an option given the critically ill state and the presence of HLA antibodies. Airway pressure release ventilation (APRV) may be valuable in ARDS, but APRV settings recommended in various patient and clinical studies are inconsistent. The Time Controlled Adaptive Ventilation (TCAV™) method is the most studied technique to set and adjust the APRV mode and uses an extended continuous positive airway pressure (CPAP) Phase in combination with a very brief Release Phase. In addition, the TCAV™ method settings are personalized and adaptive based on changes in lung pathophysiology. We used the TCAV™ method in a case of severe ARDS, which enabled us to open, stabilize and slowly heal the severely damaged lung parenchyma. Case presentation A 43-year-old woman presented with Staphylococcus Aureus necrotizing pneumonia. Progressive respiratory failure necessitated invasive mechanical ventilation and VV-ECMO. Mechanical ventilation (MV) was ultimately discontinued because lung protective settings resulted in trivial tidal volumes. She was referred to our academic transplant center for bilateral lung transplantation after the remaining infection had been cleared. We initiated the TCAV™ method in order to stabilize the lung parenchyma and to promote tissue recovery. This strategy was challenged by the presence of a large bronchopleural fistula, however, APRV enabled weaning from VV-ECMO and mechanical ventilation. After two months, following nearly complete surgical closure of the remaining bronchopleural fistulas, the patient was readmitted to ICU where she had early postoperative complications. Since other ventilation modes resulted in significant atelectasis and hypercapnia, APRV was restarted. The patient was then again weaned from MV. Conclusions The TCAV™ method can be useful to wean challenging patients with severe ARDS and might contribute to lung recovery. In this particular case, a lung transplantation was circumvented.
Collapse
Affiliation(s)
- Malou Janssen
- Department of Intensive Care Medicine, Erasmus MC, University Medical Center, Dr Molewaterplein 40, Room Rg 626, 3015 GD, Rotterdam, The Netherlands.
| | - J Han J Meeder
- Department of Intensive Care Medicine, Erasmus MC, University Medical Center, Dr Molewaterplein 40, Room Rg 626, 3015 GD, Rotterdam, The Netherlands
| | - Leonard Seghers
- Department of Pulmonary Medicine, Transplant Center, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Corstiaan A den Uil
- Department of Intensive Care Medicine, Erasmus MC, University Medical Center, Dr Molewaterplein 40, Room Rg 626, 3015 GD, Rotterdam, The Netherlands.,Department of Cardiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.,Department of Intensive Care Medicine, Maasstad Hospital, Rotterdam, The Netherlands
| |
Collapse
|
38
|
Nikitopoulou I, Ninou I, Manitsopoulos N, Dimopoulou I, Orfanos SE, Aidinis V, Kotanidou A. A role for bronchial epithelial autotaxin in ventilator-induced lung injury. Intensive Care Med Exp 2021; 9:12. [PMID: 33778909 PMCID: PMC8005331 DOI: 10.1186/s40635-021-00379-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/22/2021] [Indexed: 01/13/2023] Open
Abstract
Background The pathophysiology of acute respiratory distress syndrome (ARDS) may eventually result in heterogeneous lung collapse and edema-flooded airways, predisposing the lung to progressive tissue damage known as ventilator-induced lung injury (VILI). Autotaxin (ATX; ENPP2), the enzyme largely responsible for extracellular lysophosphatidic acid (LPA) production, has been suggested to play a pathogenic role in, among others, pulmonary inflammation and fibrosis. Methods C57BL/6 mice were subjected to low and high tidal volume mechanical ventilation using a small animal ventilator: respiratory mechanics were evaluated, and plasma and bronchoalveolar lavage fluid (BALF) samples were obtained. Total protein concentration was determined, and lung histopathology was further performed Results Injurious ventilation resulted in increased BALF levels of ATX. Genetic deletion of ATX from bronchial epithelial cells attenuated VILI-induced pulmonary edema. Conclusion ATX participates in VILI pathogenesis.
Collapse
Affiliation(s)
- Ioanna Nikitopoulou
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, National and Kapodistrian University of Athens Medical School, Evangelismos Hospital, 45, Ipsilantou Street, Athens, Greece
| | - Ioanna Ninou
- Institute of Immunology, Biomedical Sciences Research Center Alexander Fleming, Athens, Greece
| | - Nikolaos Manitsopoulos
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, National and Kapodistrian University of Athens Medical School, Evangelismos Hospital, 45, Ipsilantou Street, Athens, Greece
| | - Ioanna Dimopoulou
- 1st Department of Critical Care Medicine & Pulmonary Services, National and Kapodistrian University of Athens Medical School, Evangelismos Hospital, 45, Ipsilantou Street, Athens, Greece
| | - Stylianos E Orfanos
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, National and Kapodistrian University of Athens Medical School, Evangelismos Hospital, 45, Ipsilantou Street, Athens, Greece.,2nd Department of Critical Care, National and Kapodistrian University of Athens Medical School, Attikon" Hospital, Athens, Greece
| | - Vassilis Aidinis
- Institute of Immunology, Biomedical Sciences Research Center Alexander Fleming, Athens, Greece
| | - Anastasia Kotanidou
- 1st Department of Critical Care Medicine & Pulmonary Services, GP Livanos and M Simou Laboratories, National and Kapodistrian University of Athens Medical School, Evangelismos Hospital, 45, Ipsilantou Street, Athens, Greece. .,1st Department of Critical Care Medicine & Pulmonary Services, National and Kapodistrian University of Athens Medical School, Evangelismos Hospital, 45, Ipsilantou Street, Athens, Greece.
| |
Collapse
|
39
|
Habashi NM, Camporota L, Gatto LA, Nieman G. Functional pathophysiology of SARS-CoV-2-induced acute lung injury and clinical implications. J Appl Physiol (1985) 2021; 130:877-891. [PMID: 33444117 PMCID: PMC7984238 DOI: 10.1152/japplphysiol.00742.2020] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 02/08/2023] Open
Abstract
The worldwide pandemic caused by the SARS-CoV-2 virus has resulted in over 84,407,000 cases, with over 1,800,000 deaths when this paper was submitted, with comorbidities such as gender, race, age, body mass, diabetes, and hypertension greatly exacerbating mortality. This review will analyze the rapidly increasing knowledge of COVID-19-induced lung pathophysiology. Although controversial, the acute respiratory distress syndrome (ARDS) associated with COVID-19 (CARDS) seems to present as two distinct phenotypes: type L and type H. The "L" refers to low elastance, ventilation/perfusion ratio, lung weight, and recruitability, and the "H" refers to high pulmonary elastance, shunt, edema, and recruitability. However, the LUNG-SAFE (Large Observational Study to Understand the Global Impact of Severe Acute Respiratory Failure) and ESICM (European Society of Intensive Care Medicine) Trials Groups have shown that ∼13% of the mechanically ventilated non-COVID-19 ARDS patients have the type-L phenotype. Other studies have shown that CARDS and ARDS respiratory mechanics overlap and that standard ventilation strategies apply to these patients. The mechanisms causing alterations in pulmonary perfusion could be caused by some combination of 1) renin-angiotensin system dysregulation, 2) thrombosis caused by loss of endothelial barrier, 3) endothelial dysfunction causing loss of hypoxic pulmonary vasoconstriction perfusion control, and 4) hyperperfusion of collapsed lung tissue that has been directly measured and supported by a computational model. A flowchart has been constructed highlighting the need for personalized and adaptive ventilation strategies, such as the time-controlled adaptive ventilation method, to set and adjust the airway pressure release ventilation mode, which recently was shown to be effective at improving oxygenation and reducing inspiratory fraction of oxygen, vasopressors, and sedation in patients with COVID-19.
Collapse
Affiliation(s)
- Nader M Habashi
- R Adams Cowley Shock Trauma Center, University of Maryland, Baltimore, Maryland
| | - Luigi Camporota
- Department of Adult Critical Care, Guy's and St Thomas' NHS Foundation Trust, King's Health Partners, St Thomas' Hospital, London, United Kingdom
| | - Louis A Gatto
- Department of Surgery, Upstate Medical University, Syracuse, New York
| | - Gary Nieman
- Department of Surgery, Upstate Medical University, Syracuse, New York
| |
Collapse
|
40
|
Gaver DP, Nieman GF, Gatto LA, Cereda M, Habashi NM, Bates JHT. The POOR Get POORer: A Hypothesis for the Pathogenesis of Ventilator-induced Lung Injury. Am J Respir Crit Care Med 2020; 202:1081-1087. [PMID: 33054329 DOI: 10.1164/rccm.202002-0453cp] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Protective ventilation strategies for the injured lung currently revolve around the use of low Vt, ostensibly to avoid volutrauma, together with positive end-expiratory pressure to increase the fraction of open lung and reduce atelectrauma. Protective ventilation is currently applied in a one-size-fits-all manner, and although this practical approach has reduced acute respiratory distress syndrome deaths, mortality is still high and improvements are at a standstill. Furthermore, how to minimize ventilator-induced lung injury (VILI) for any given lung remains controversial and poorly understood. Here we present a hypothesis of VILI pathogenesis that potentially serves as a basis upon which minimally injurious ventilation strategies might be developed. This hypothesis is based on evidence demonstrating that VILI begins in isolated lung regions manifesting a Permeability-Originated Obstruction Response (POOR) in which alveolar leak leads to surfactant dysfunction and increases local tissue stresses. VILI progresses topographically outward from these regions in a POOR-get-POORer fashion unless steps are taken to interrupt it. We propose that interrupting the POOR-get-POORer progression of lung injury relies on two principles: 1) open the lung to minimize the presence of heterogeneity-induced stress concentrators that are focused around the regions of atelectasis, and 2) ventilate in a patient-dependent manner that minimizes the number of lung units that close during each expiration so that they are not forced to rerecruit during the subsequent inspiration. These principles appear to be borne out in both patient and animal studies in which expiration is terminated before derecruitment of lung units has enough time to occur.
Collapse
Affiliation(s)
- Donald P Gaver
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana
| | - Gary F Nieman
- Department of Surgery, SUNY Upstate Medical University, Syracuse, New York
| | - Louis A Gatto
- Department of Surgery, SUNY Upstate Medical University, Syracuse, New York
| | - Maurizio Cereda
- Department of Anesthesiology and Critical Care and.,Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nader M Habashi
- R Adams Cowley Shock Trauma Center, University of Maryland, Baltimore, Maryland; and
| | - Jason H T Bates
- Department of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
41
|
Schousboe P, Wiese L, Heiring C, Verder H, Poorisrisak P, Verder P, Nielsen HB. Assessment of pulmonary surfactant in COVID-19 patients. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:552. [PMID: 32894160 PMCID: PMC7475719 DOI: 10.1186/s13054-020-03268-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 08/27/2020] [Indexed: 12/03/2022]
Affiliation(s)
- Peter Schousboe
- Department of Pediatrics, Holbaek University Hospital, Smedelundsgade 60, 4300, Holbaek, Denmark
| | - Lothar Wiese
- Department of Infectious Diseases, Zealand University Hospital Roskilde, Sygehusvej 10, 4000, Roskilde, Denmark
| | - Christian Heiring
- Department of Neonatology, Neonatal and Pediatric Intensive Care Unit 5021 Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Verder
- Department of Pediatrics, Holbaek University Hospital, Smedelundsgade 60, 4300, Holbaek, Denmark
| | - Porntiva Poorisrisak
- Department of Neonatology, Neonatal and Pediatric Intensive Care Unit 5021 Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Povl Verder
- Department of Pediatrics, Holbaek University Hospital, Smedelundsgade 60, 4300, Holbaek, Denmark
| | - Henning Bay Nielsen
- Department of Anesthesia and Intensive Care, Zealand University Hospital Roskilde, Sygehusvej 10, 4000, Roskilde, Denmark.
| |
Collapse
|
42
|
Sehlmeyer K, Ruwisch J, Roldan N, Lopez-Rodriguez E. Alveolar Dynamics and Beyond - The Importance of Surfactant Protein C and Cholesterol in Lung Homeostasis and Fibrosis. Front Physiol 2020; 11:386. [PMID: 32431623 PMCID: PMC7213507 DOI: 10.3389/fphys.2020.00386] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 03/30/2020] [Indexed: 12/13/2022] Open
Abstract
Surfactant protein C (SP-C) is an important player in enhancing the interfacial adsorption of lung surfactant lipid films to the alveolar air-liquid interface. Doing so, surface tension drops down enough to stabilize alveoli and the lung, reducing the work of breathing. In addition, it has been shown that SP-C counteracts the deleterious effect of high amounts of cholesterol in the surfactant lipid films. On its side, cholesterol is a well-known modulator of the biophysical properties of biological membranes and it has been proven that it activates the inflammasome pathways in the lung. Even though the molecular mechanism is not known, there are evidences suggesting that these two molecules may interplay with each other in order to keep the proper function of the lung. This review focuses in the role of SP-C and cholesterol in the development of lung fibrosis and the potential pathways in which impairment of both molecules leads to aberrant lung repair, and therefore impaired alveolar dynamics. From molecular to cellular mechanisms to evidences in animal models and human diseases. The evidences revised here highlight a potential SP-C/cholesterol axis as target for the treatment of lung fibrosis.
Collapse
Affiliation(s)
- Kirsten Sehlmeyer
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Member of the German Centre for Lung Research, Hanover, Germany
| | - Jannik Ruwisch
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Member of the German Centre for Lung Research, Hanover, Germany
| | - Nuria Roldan
- Alveolix AG and ARTORG Center, University of Bern, Bern, Switzerland
| | - Elena Lopez-Rodriguez
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Member of the German Centre for Lung Research, Hanover, Germany
- Institute of Functional Anatomy, Charité – Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
43
|
In Brief. Curr Probl Surg 2020. [DOI: 10.1016/j.cpsurg.2020.100778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
44
|
Guérin C, Terzi N, Galerneau LM, Mezidi M, Yonis H, Baboi L, Kreitmann L, Turbil E, Cour M, Argaud L, Louis B. Lung and chest wall mechanics in patients with acute respiratory distress syndrome, expiratory flow limitation, and airway closure. J Appl Physiol (1985) 2020; 128:1594-1603. [PMID: 32352339 DOI: 10.1152/japplphysiol.00059.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tidal expiratory flow limitation (EFL), which may herald airway closure (AC), is a mechanism of loss of aeration in ARDS. In this prospective, short-term, two-center study, we measured static and dynamic chest wall (Est,cw and Edyn,cw) and lung (Est,L and Edyn,L) elastance with esophageal pressure, EFL, and AC at 5 cmH2O positive end-expiratory pressure (PEEP) in intubated, sedated, and paralyzed ARDS patients. For EFL determination, we used the atmospheric method and a new device allowing comparison of tidal flow during expiration to PEEP and to atmosphere. AC was validated when airway opening pressure (AOP) assessed from volume-pressure curve was found greater than PEEP by at least 1 cmH2O. EFL was defined whenever flow did not increase between exhalation to PEEP and to atmosphere over all or part of expiration. Elastance values were expressed as percentage of normal predicted values (%N). Among the 25 patients included, eight had EFL (32%) and 13 AOP (52%). Between patients with and without EFL Edyn,cw [median (1st to 3rd quartiles)] was 70 (16-127) and 102 (70-142) %N (P = 0.32) and Edyn,L338 (332-763) and 224 (160-275) %N (P < 0.001). The corresponding values for Est,cw and Est,L were 70 (56-88) and 85 (64-103) %N (P = 0.35) and 248 (206-348) and 170 (144-195) (P = 0.02), respectively. Dynamic EL had an area receiver operating characteristic curve of 0.88 [95% confidence intervals 0.83-0.92] for EFL and 0.74[0.68-0.79] for AOP. Higher Edyn,L was accurate to predict EFL in ARDS patients; AC can occur independently of EFL, and both should be assessed concurrently in ARDS patients.NEW & NOTEWORTHY Expiratory flow limitation (EFL) and airway closure (AC) were observed in 32% and 52%, respectively, of 25 patients with ARDS investigated during mechanical ventilation in supine position with a positive end-expiratory pressure of 5 cmH2O. The performance of dynamic lung elastance to detect expiratory flow limitation was good and better than that to detect airway closure. The vast majority of patients with EFL also had AC; however, AC can occur in the absence of EFL.
Collapse
Affiliation(s)
- Claude Guérin
- Medecine Intensive-Réanimation, Groupement Hospitalier Centre, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France.,Université de Lyon, Lyon, France.,Institut Mondor de Recherches Biomédicales INSERM 955 CNRS ERL 7000, Créteil, France
| | - Nicolas Terzi
- Medecine Intensive-Réanimation, CHU Grenoble-Alpes, Grenoble, France.,Université de Grenoble-Alpes, Grenoble, France
| | - Louis-Marie Galerneau
- Medecine Intensive-Réanimation, CHU Grenoble-Alpes, Grenoble, France.,Université de Grenoble-Alpes, Grenoble, France
| | - Mehdi Mezidi
- Université de Lyon, Lyon, France.,Médecine Intensive-Réanimation, Groupement Hospitalier Nord, Hospices Civils de Lyon, Lyon, France
| | - Hodane Yonis
- Médecine Intensive-Réanimation, Groupement Hospitalier Nord, Hospices Civils de Lyon, Lyon, France
| | - Loredana Baboi
- Médecine Intensive-Réanimation, Groupement Hospitalier Nord, Hospices Civils de Lyon, Lyon, France
| | - Louis Kreitmann
- Medecine Intensive-Réanimation, Groupement Hospitalier Centre, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France.,Université de Lyon, Lyon, France
| | - Emanuele Turbil
- Department of Anesthesia and Critical Care, University of Sassari, Sassari, Italy
| | - Martin Cour
- Medecine Intensive-Réanimation, Groupement Hospitalier Centre, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France.,Université de Lyon, Lyon, France
| | - Laurent Argaud
- Medecine Intensive-Réanimation, Groupement Hospitalier Centre, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France.,Université de Lyon, Lyon, France
| | - Bruno Louis
- Institut Mondor de Recherches Biomédicales INSERM 955 CNRS ERL 7000, Créteil, France
| |
Collapse
|
45
|
Kollisch-Singule M, Satalin J, Blair SJ, Andrews PL, Gatto LA, Nieman GF, Habashi NM. Mechanical Ventilation Lessons Learned From Alveolar Micromechanics. Front Physiol 2020; 11:233. [PMID: 32265735 PMCID: PMC7105828 DOI: 10.3389/fphys.2020.00233] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/28/2020] [Indexed: 01/05/2023] Open
Abstract
Morbidity and mortality associated with lung injury remains disappointingly unchanged over the last two decades, in part due to the current reliance on lung macro-parameters set on the ventilator instead of considering the micro-environment and the response of the alveoli and alveolar ducts to ventilator adjustments. The response of alveoli and alveolar ducts to mechanical ventilation modes cannot be predicted with current bedside methods of assessment including lung compliance, oxygenation, and pressure-volume curves. Alveolar tidal volumes (Vt) are less determined by the Vt set on the mechanical ventilator and more dependent on the number of recruited alveoli available to accommodate that Vt and their heterogeneous mechanical properties, such that high lung Vt can lead to a low alveolar Vt and low Vt can lead to high alveolar Vt. The degree of alveolar heterogeneity that exists cannot be predicted based on lung calculations that average the individual alveolar Vt and compliance. Finally, the importance of time in promoting alveolar stability, specifically the inspiratory and expiratory times set on the ventilator, are currently under-appreciated. In order to improve outcomes related to lung injury, the respiratory physiology of the individual patient, specifically at the level of the alveolus, must be targeted. With experimental data, this review highlights some of the known mechanical ventilation adjustments that are helpful or harmful at the level of the alveolus.
Collapse
Affiliation(s)
| | - Joshua Satalin
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Sarah J. Blair
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Penny L. Andrews
- Department of Critical Care, R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, MD, United States
| | - Louis A. Gatto
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
- Department of Biological Sciences, SUNY Cortland, Cortland, NY, United States
| | - Gary F. Nieman
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Nader M. Habashi
- Department of Critical Care, R Adams Cowley Shock Trauma Center, University of Maryland Medical Center, Baltimore, MD, United States
| |
Collapse
|
46
|
Nieman GF, Al-Khalisy H, Kollisch-Singule M, Satalin J, Blair S, Trikha G, Andrews P, Madden M, Gatto LA, Habashi NM. A Physiologically Informed Strategy to Effectively Open, Stabilize, and Protect the Acutely Injured Lung. Front Physiol 2020; 11:227. [PMID: 32265734 PMCID: PMC7096584 DOI: 10.3389/fphys.2020.00227] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 02/27/2020] [Indexed: 12/16/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) causes a heterogeneous lung injury and remains a serious medical problem, with one of the only treatments being supportive care in the form of mechanical ventilation. It is very difficult, however, to mechanically ventilate the heterogeneously damaged lung without causing secondary ventilator-induced lung injury (VILI). The acutely injured lung becomes time and pressure dependent, meaning that it takes more time and pressure to open the lung, and it recollapses more quickly and at higher pressure. Current protective ventilation strategies, ARDSnet low tidal volume (LVt) and the open lung approach (OLA), have been unsuccessful at further reducing ARDS mortality. We postulate that this is because the LVt strategy is constrained to ventilating a lung with a heterogeneous mix of normal and focalized injured tissue, and the OLA, although designed to fully open and stabilize the lung, is often unsuccessful at doing so. In this review we analyzed the pathophysiology of ARDS that renders the lung susceptible to VILI. We also analyzed the alterations in alveolar and alveolar duct mechanics that occur in the acutely injured lung and discussed how these alterations are a key mechanism driving VILI. Our analysis suggests that the time component of each mechanical breath, at both inspiration and expiration, is critical to normalize alveolar mechanics and protect the lung from VILI. Animal studies and a meta-analysis have suggested that the time-controlled adaptive ventilation (TCAV) method, using the airway pressure release ventilation mode, eliminates the constraints of ventilating a lung with heterogeneous injury, since it is highly effective at opening and stabilizing the time- and pressure-dependent lung. In animal studies it has been shown that by “casting open” the acutely injured lung with TCAV we can (1) reestablish normal expiratory lung volume as assessed by direct observation of subpleural alveoli; (2) return normal parenchymal microanatomical structural support, known as alveolar interdependence and parenchymal tethering, as assessed by morphometric analysis of lung histology; (3) facilitate regeneration of normal surfactant function measured as increases in surfactant proteins A and B; and (4) significantly increase lung compliance, which reduces the pathologic impact of driving pressure and mechanical power at any given tidal volume.
Collapse
Affiliation(s)
- Gary F Nieman
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Hassan Al-Khalisy
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States.,Department of Medicine, SUNY Upstate Medical University, Syracuse, NY, United States
| | | | - Joshua Satalin
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Sarah Blair
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Girish Trikha
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States.,Department of Medicine, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Penny Andrews
- Department of Trauma Critical Care Medicine, R Adams Cowley Shock Trauma Center, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Maria Madden
- Department of Trauma Critical Care Medicine, R Adams Cowley Shock Trauma Center, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Louis A Gatto
- Department of Surgery, SUNY Upstate Medical University, Syracuse, NY, United States.,Department of Biological Sciences, SUNY Cortland, Cortland, NY, United States
| | - Nader M Habashi
- Department of Trauma Critical Care Medicine, R Adams Cowley Shock Trauma Center, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
47
|
Mowery NT, Terzian WTH, Nelson AC. Acute lung injury. Curr Probl Surg 2020; 57:100777. [PMID: 32505224 DOI: 10.1016/j.cpsurg.2020.100777] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 02/24/2020] [Indexed: 01/04/2023]
Affiliation(s)
- Nathan T Mowery
- Associate Professor of Surgery, Wake Forest Medical Center, Winston-Salem, NC.
| | | | - Adam C Nelson
- Acute Care Surgery Fellow, Wake Forest Medical Center, Winston-Salem, NC
| |
Collapse
|
48
|
Fredericks AS, Bunker MP, Gliga LA, Ebeling CG, Ringqvist JR, Heravi H, Manley J, Valladares J, Romito BT. Airway Pressure Release Ventilation: A Review of the Evidence, Theoretical Benefits, and Alternative Titration Strategies. CLINICAL MEDICINE INSIGHTS-CIRCULATORY RESPIRATORY AND PULMONARY MEDICINE 2020; 14:1179548420903297. [PMID: 32076372 PMCID: PMC7003159 DOI: 10.1177/1179548420903297] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 01/08/2020] [Indexed: 11/15/2022]
Abstract
Objective: To review the theoretical benefits of airway pressure release ventilation (APRV), summarize the evidence for its use in clinical practice, and discuss different titration strategies. Data Source: Published randomized controlled trials in humans, observational human studies, animal studies, review articles, ventilator textbooks, and editorials. Data Summary: Airway pressure release ventilation optimizes alveolar recruitment, reduces airway pressures, allows for spontaneous breathing, and offers many hemodynamic benefits. Despite these physiologic advantages, there are inconsistent data to support the use of APRV over other modes of ventilation. There is considerable heterogeneity in the application of APRV among providers and a shortage of information describing initiation and titration strategies. To date, no direct comparison studies of APRV strategies have been performed. This review describes 2 common management approaches that bedside providers can use to optimally tailor APRV to their patients. Conclusion: Airway pressure release ventilation remains a form of mechanical ventilation primarily used for refractory hypoxemia. It offers unique physiological advantages over other ventilatory modes, and providers must be familiar with different titration methods. Given its inconsistent outcome data and heterogeneous use in practice, future trials should directly compare APRV strategies to determine the optimal management approach.
Collapse
Affiliation(s)
- Andrew S Fredericks
- Department of Anesthesiology and Pain Management, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Matthew P Bunker
- Department of Anesthesiology and Pain Management, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Louise A Gliga
- Department of Anesthesiology and Pain Management, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Callie G Ebeling
- Department of Anesthesiology and Pain Management, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jenny Rb Ringqvist
- Department of Anesthesiology and Pain Management, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hooman Heravi
- Department of Anesthesiology and Pain Management, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - James Manley
- Department of Respiratory Care, Parkland Memorial Hospital, Dallas, TX, USA.,The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jason Valladares
- Department of Anesthesiology and Pain Management, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bryan T Romito
- Department of Anesthesiology and Pain Management, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
49
|
Li L, Huang Q, Wang DC, Ingbar DH, Wang X. Acute lung injury in patients with COVID-19 infection. Clin Transl Med 2020; 10:20-27. [PMID: 32508022 PMCID: PMC7240840 DOI: 10.1002/ctm2.16] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 03/14/2020] [Indexed: 01/10/2023] Open
Abstract
During the 2020 Spring Festival in China, the outbreak of a novel coronavirus, named COVID-19 by WHO, brought on a worldwide panic. According to the clinical data of infected patients, radiologic evidence of lung edema is common and deserves clinical attention. Lung edema is a manifestation of acute lung injury (ALI) and may progress to hypoxemia and potentially acute respiratory distress syndrome (ARDS). Patients diagnosed with ARDS have poorer prognosis and potentially higher mortality. Although no effective treatment is formally approved for COVID-19 infection, support of ventilation with oxygen therapy and sometimes mechanical ventilation is often required. Treatment with systemic and/or local glucocorticoids might be helpful to alleviate the pulmonary inflammation and edema, which may decrease the development and/or consequences of ARDS. In this article, we focus on the lung edema and ALI of patients with this widely transmitted COVID-19 infection in order to provide clinical indications and potential therapeutic targets for clinicians and researchers.
Collapse
Affiliation(s)
- Liyang Li
- Zhongshan Hospital Institute of Clinical ScienceShanghai Medical SchoolFudan UniversityShanghaiChina
| | - Qihong Huang
- Zhongshan Hospital Institute of Clinical ScienceShanghai Medical SchoolFudan UniversityShanghaiChina
| | - Diane C. Wang
- Department of EmergencySunshine Coast University HospitalBirtinyaQueenslandAustralia
| | - David H. Ingbar
- Pulmonary, Allergy, Critical Care & Sleep Division, Center for Lung Science and HealthUniversity of MinnesotaMinnesotaUSA
| | - Xiangdong Wang
- Zhongshan Hospital Institute of Clinical ScienceShanghai Medical SchoolFudan UniversityShanghaiChina
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan HospitalShanghai Medical SchoolFudan UniversityShanghaiChina
- Shanghai Engineering Research Center of AI Technology for Cardiopulmonary DiseasesShanghaiChina
- Shanghai Institute of Clinical BioinformaticsShanghaiChina
| |
Collapse
|
50
|
Viola H, Chang J, Grunwell JR, Hecker L, Tirouvanziam R, Grotberg JB, Takayama S. Microphysiological systems modeling acute respiratory distress syndrome that capture mechanical force-induced injury-inflammation-repair. APL Bioeng 2019; 3:041503. [PMID: 31768486 PMCID: PMC6874511 DOI: 10.1063/1.5111549] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 11/08/2019] [Indexed: 12/14/2022] Open
Abstract
Complex in vitro models of the tissue microenvironment, termed microphysiological systems, have enormous potential to transform the process of discovering drugs and disease mechanisms. Such a paradigm shift is urgently needed in acute respiratory distress syndrome (ARDS), an acute lung condition with no successful therapies and a 40% mortality rate. Here, we consider how microphysiological systems could improve understanding of biological mechanisms driving ARDS and ultimately improve the success of therapies in clinical trials. We first discuss how microphysiological systems could explain the biological mechanisms underlying the segregation of ARDS patients into two clinically distinct phenotypes. Then, we contend that ARDS-mimetic microphysiological systems should recapitulate three critical aspects of the distal airway microenvironment, namely, mechanical force, inflammation, and fibrosis, and we review models that incorporate each of these aspects. Finally, we recognize the substantial challenges associated with combining inflammation, fibrosis, and/or mechanical force in microphysiological systems. Nevertheless, complex in vitro models are a novel paradigm for studying ARDS, and they could ultimately improve patient care.
Collapse
Affiliation(s)
| | - Jonathan Chang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, Georgia 30332, USA
| | - Jocelyn R. Grunwell
- Department of Pediatrics, Division of Critical Care Medicine, Children's Healthcare of Atlanta at Egleston, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Louise Hecker
- Division of Pulmonary, Allergy and Critical Care and Sleep Medicine, University of Arizona, Tucson, Arizona 85724, USA and Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona 85723, USA
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia 30322, USA and Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, Georgia 30322, USA
| | - James B. Grotberg
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | |
Collapse
|