1
|
Akutsu T, Endo A, Yamamoto R, Yamakawa K, Suzuki K, Hoshi H, Otomo Y, Morishita K. Veno-arterial extracorporeal membrane oxygenation uses in trauma: a retrospective analysis of the Japanese nationwide trauma registry. BMC Emerg Med 2024; 24:179. [PMID: 39379814 PMCID: PMC11462744 DOI: 10.1186/s12873-024-01096-6] [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: 07/11/2024] [Accepted: 09/23/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Extracorporeal membrane oxygenation (ECMO) can provide temporary circulatory support and vital organ oxygenation and is potentially useful as a bridge therapy in some trauma cases. We aimed to demonstrate the characteristics and outcomes of patients with trauma treated with veno-arterial ECMO (V-A ECMO) using data from a Japanese nationwide trauma registry. METHODS This retrospective descriptive study analyzed data from the Japan Trauma Data Bank between January 2019 and December 2021. Patients with severe trauma (injury severity score [ISS] ≥ 9) and treated using V-A ECMO were assessed. RESULTS Among the 72,439 patients with severe trauma, 51 received V-A ECMO. Sixteen patients (31.3%) survived until hospital discharge. On hospital arrival, six (37.5%) survivors and 15 (42.9%) non-survivors experienced cardiac arrest. The median ISS for the survivor and non-survivor group was 25 (range, 25-39) and 25 (range, 17-33), respectively. Thoracic trauma was the most common type of trauma in both groups. In the non-survivor group, open-chest cardiopulmonary resuscitation, aortic cross-clamping, and resuscitative endovascular balloon occlusion of the aorta were performed in 10 (28.6%), 5 (14.3%), and 4 (11.4%) patients, respectively. However, these procedures were not performed in the survivor group. Peripheral oxygen saturation tended to be lower in the survivor group both before and upon arrival at the hospital. CONCLUSIONS The results of this study suggest the potential benefit of V-A ECMO in some challenging trauma cases. Further studies are warranted to assess the indications for V-A ECMO in patients with trauma.
Collapse
Affiliation(s)
- Tomohiro Akutsu
- Department of Acute Critical Care Medicine, Tsuchiura Kyodo General Hospital, 4-1-1 Otsuno, Tsuchiura-shi, Ibaraki, Japan
| | - Akira Endo
- Department of Acute Critical Care Medicine, Tsuchiura Kyodo General Hospital, 4-1-1 Otsuno, Tsuchiura-shi, Ibaraki, Japan.
- Department of Acute Critical Care and Disaster Medicine, Tokyo Medical and Dental University Graduate School of Medicine and Dental Sciences, 1-5-45 Yushima, Bunkyo-ku, Tokyo, Japan.
| | - Ryo Yamamoto
- Department of Emergency and Critical Care Medicine, Keio University School of Medicine, 35 Shinanomachi, Hiroshima, Japan
| | - Kazuma Yamakawa
- Department of Emergency and Critical Care Medicine, Osaka Medical and Pharmaceutical University, 2-7 Daigakucho, Takatsuki-shi, Osaka, Japan
| | - Keisuke Suzuki
- Department of Acute Critical Care Medicine, Tsuchiura Kyodo General Hospital, 4-1-1 Otsuno, Tsuchiura-shi, Ibaraki, Japan
- Department of Acute Critical Care and Disaster Medicine, Tokyo Medical and Dental University Graduate School of Medicine and Dental Sciences, 1-5-45 Yushima, Bunkyo-ku, Tokyo, Japan
| | - Hiromasa Hoshi
- Department of Acute Critical Care Medicine, Tsuchiura Kyodo General Hospital, 4-1-1 Otsuno, Tsuchiura-shi, Ibaraki, Japan
| | - Yasuhiro Otomo
- National Hospital Organization Disaster Medical Center, 3256 Midoricho, Tachikawa-shi, Tokyo, Japan
| | - Koji Morishita
- Department of Acute Critical Care and Disaster Medicine, Tokyo Medical and Dental University Graduate School of Medicine and Dental Sciences, 1-5-45 Yushima, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
2
|
Kaatsch HL, Völlmecke MF, Becker BV, Dillinger D, Kubitscheck L, Wöhler A, Schaaf S, Piechotka J, Schreyer C, Schwab R, Overhoff D, Waldeck S. Improved Discriminability of Severe Lung Injury and Atelectasis in Thoracic Trauma at Low keV Virtual Monoenergetic Images from Photon-Counting Detector CT. Diagnostics (Basel) 2024; 14:2231. [PMID: 39410635 PMCID: PMC11475707 DOI: 10.3390/diagnostics14192231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/25/2024] [Accepted: 09/28/2024] [Indexed: 10/20/2024] Open
Abstract
Objectives: To evaluate the value of virtual monoenergetic images (VMI) from photon-counting detector CT (PCD-CT) for discriminability of severe lung injury and atelectasis in polytraumatized patients. Materials & Methods: Contrast-enhanced PCD-CT examinations of 20 polytraumatized patients with severe thoracic trauma were included in this retrospective study. Spectral PCD-CT data were reconstructed using a noise-optimized virtual monoenergetic imaging (VMI) algorithm with calculated VMIs ranging from 40 to 120 keV at 10 keV increments. Injury-to-atelectasis contrast-to-noise ratio (CNR) was calculated and compared at each energy level based on CT number measurements in severely injured as well as atelectatic lung areas. Three radiologists assessed subjective discriminability, noise perception, and overall image quality. Results: CT values for atelectasis decreased as photon energy increased from 40 keV to 120 keV (mean Hounsfield units (HU): 69 at 40 keV; 342 at 120 keV), whereas CT values for severe lung injury remained near-constant from 40 keV to 120 keV (mean HU: 42 at 40 keV; 44 at 120 keV) with significant differences at each keV level (p < 0.001). The optimal injury-to-atelectasis CNR was observed at 40 keV in comparison with the remaining energy levels (p < 0.001) except for 50 keV (p > 0.05). In line with this, VMIs at 40 keV were rated best regarding subjective discriminability. VMIs at 60-70 keV, however, provided the highest subjective observer parameters regarding subjective image noise as well as image quality. Conclusions: Discriminability between severely injured and atelectatic lung areas after thoracic trauma can be substantially improved by virtual monoenergetic imaging from PCD-CT with superior contrast and visual discriminability at 40-50 keV.
Collapse
Affiliation(s)
- Hanns Leonhard Kaatsch
- Department of Radiology and Neuroradiology, Bundeswehr Central Hospital, 56072 Koblenz, Germany; (H.L.K.)
| | - Maximilian Franz Völlmecke
- Department of Plastic, Hand and Reconstructive Surgery, Burn Centre, Sarcoma Centre, BG University Hospital Bergmannsheil, 44789 Bochum, Germany
- Department of Trauma Surgery, Hand and Reconstructive Surgery, Bundeswehr Central Hospital, 56072 Koblenz, Germany
| | - Benjamin V. Becker
- Department of Radiology and Neuroradiology, Bundeswehr Central Hospital, 56072 Koblenz, Germany; (H.L.K.)
- Department of Neuroradiology, University Medical Center Mainz, 55131 Mainz, Germany
| | - Daniel Dillinger
- Department of Vascular Surgery and Endovascular Surgery, Bundeswehr Central Hospital, 56072 Koblenz, Germany
| | - Laura Kubitscheck
- Department of Radiology and Neuroradiology, Bundeswehr Central Hospital, 56072 Koblenz, Germany; (H.L.K.)
- Bundeswehr Institute of Radiobiology Affiliated to Ulm University, 80937 Munich, Germany
| | - Aliona Wöhler
- Department of General, Visceral and Thoracic Surgery, Bundeswehr Central Hospital, 56072 Koblenz, Germany
| | - Sebastian Schaaf
- Department of General, Visceral and Thoracic Surgery, Bundeswehr Central Hospital, 56072 Koblenz, Germany
| | - Joel Piechotka
- Department of Radiology and Neuroradiology, Bundeswehr Central Hospital, 56072 Koblenz, Germany; (H.L.K.)
| | - Christof Schreyer
- Department of General, Visceral and Thoracic Surgery, Bundeswehr Central Hospital, 56072 Koblenz, Germany
| | - Robert Schwab
- Department of General, Visceral and Thoracic Surgery, Bundeswehr Central Hospital, 56072 Koblenz, Germany
| | - Daniel Overhoff
- Department of Radiology and Neuroradiology, Bundeswehr Central Hospital, 56072 Koblenz, Germany; (H.L.K.)
- Department of Radiology and Nuclear Medicine, University Medical Center Mannheim, 68167 Mannheim, Germany
| | - Stephan Waldeck
- Department of Radiology and Neuroradiology, Bundeswehr Central Hospital, 56072 Koblenz, Germany; (H.L.K.)
- Department of Neuroradiology, University Medical Center Mainz, 55131 Mainz, Germany
| |
Collapse
|
3
|
Qu Y, Wang Z, Dong L, Zhang D, Shang F, Li A, Gao Y, Bai Q, Liu D, Xie X, Ming L. Natural small molecules synergize mesenchymal stem cells for injury repair in vital organs: a comprehensive review. Stem Cell Res Ther 2024; 15:243. [PMID: 39113141 PMCID: PMC11304890 DOI: 10.1186/s13287-024-03856-4] [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: 06/13/2024] [Accepted: 07/17/2024] [Indexed: 08/10/2024] Open
Abstract
Mesenchymal stem cells (MSCs) therapy is a highly researched treatment that has the potential to promote immunomodulation and anti-inflammatory, anti-apoptotic, and antimicrobial activities. It is thought that it can enhance internal organ function, reverse tissue remodeling, and achieve significant organ repair and regeneration. However, the limited infusion, survival, and engraftment of transplanted MSCs diminish the effectiveness of MSCs-based therapy. Consequently, various preconditioning methods have emerged as strategies for enhancing the therapeutic effects of MSCs and achieving better clinical outcomes. In particular, the use of natural small molecule compounds (NSMs) as a pretreatment strategy is discussed in this narrative review, with a focus on their roles in regulating MSCs for injury repair in vital internal organs. Additionally, the discussion focuses on the future directions and challenges of transforming mesenchymal stem cell research into clinical applications.
Collapse
Affiliation(s)
- Yanling Qu
- Shaanxi Zhonghong, Institute of Regenerative Medicine, Xi'an, 710003, Shaanxi Province, China
| | - Zhe Wang
- Shaanxi Zhonghong, Institute of Regenerative Medicine, Xi'an, 710003, Shaanxi Province, China
| | - Lingjuan Dong
- Shaanxi Zhonghong, Institute of Regenerative Medicine, Xi'an, 710003, Shaanxi Province, China
| | - Dan Zhang
- Shaanxi Zhonghong, Institute of Regenerative Medicine, Xi'an, 710003, Shaanxi Province, China
| | - Fengqing Shang
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510000, China
| | - Afeng Li
- Shaanxi Zhonghong, Institute of Regenerative Medicine, Xi'an, 710003, Shaanxi Province, China
| | - Yanni Gao
- Shaanxi Zhonghong, Institute of Regenerative Medicine, Xi'an, 710003, Shaanxi Province, China
| | - Qinhua Bai
- Shaanxi Zhonghong, Institute of Regenerative Medicine, Xi'an, 710003, Shaanxi Province, China
| | - Dan Liu
- Shaanxi Zhonghong, Institute of Regenerative Medicine, Xi'an, 710003, Shaanxi Province, China
| | - Xiaodong Xie
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu Province, China.
| | - Leiguo Ming
- Shaanxi Zhonghong, Institute of Regenerative Medicine, Xi'an, 710003, Shaanxi Province, China.
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
4
|
Yin Z, Zhang J, Qin J, Guo L, Guo Q, Kang W, Ma C, Chen L. Anti-inflammatory properties of polysaccharides from edible fungi on health-promotion: a review. Front Pharmacol 2024; 15:1447677. [PMID: 39130633 PMCID: PMC11310034 DOI: 10.3389/fphar.2024.1447677] [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: 06/12/2024] [Accepted: 07/12/2024] [Indexed: 08/13/2024] Open
Abstract
Edible fungus polysaccharides have garnered significant attention from scholars due to their safety and potential anti-inflammatory activity. However, comprehensive summaries of their anti-inflammatory properties are still rare. This paper provides a detailed overview of the anti-inflammatory effects and mechanisms of these polysaccharides, as well as their impact on inflammation-related diseases. Additionally, the relationship between their structure and anti-inflammatory activity is discussed. It is believed that this review will greatly enhance the understanding of the application of edible fungus polysaccharides in anti-inflammatory treatments, thereby significantly promoting the development and utilization of edible fungi.
Collapse
Affiliation(s)
- Zhenhua Yin
- Henan Comprehensive Utilization of Edible and Medicinal Plant Resources Engineering Technology Research Center, Huanghe Science and Technology College, Zhengzhou, China
- National R and D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
| | - Juanjuan Zhang
- Henan Comprehensive Utilization of Edible and Medicinal Plant Resources Engineering Technology Research Center, Huanghe Science and Technology College, Zhengzhou, China
| | - Jingjing Qin
- Henan Comprehensive Utilization of Edible and Medicinal Plant Resources Engineering Technology Research Center, Huanghe Science and Technology College, Zhengzhou, China
| | - Lin Guo
- National R and D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
| | - Qingfeng Guo
- Henan Comprehensive Utilization of Edible and Medicinal Plant Resources Engineering Technology Research Center, Huanghe Science and Technology College, Zhengzhou, China
| | - Wenyi Kang
- National R and D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | - Changyang Ma
- National R and D Center for Edible Fungus Processing Technology, Henan University, Kaifeng, China
- Function Food Engineering Technology Research Center, Kaifeng, China
| | - Lin Chen
- Henan Comprehensive Utilization of Edible and Medicinal Plant Resources Engineering Technology Research Center, Huanghe Science and Technology College, Zhengzhou, China
| |
Collapse
|
5
|
Yu S, Xu C, Tang X, Wang L, Hu L, Li L, Zhou X, Li Q. Exendin-4 blockade of T1R2/T1R3 activation improves Pseudomonas aeruginosa-related pneumonia in an animal model of chemically induced diabetes. Inflamm Res 2024; 73:1185-1201. [PMID: 38748233 PMCID: PMC11214611 DOI: 10.1007/s00011-024-01891-8] [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: 01/13/2024] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
OBJECTIVE Poorly controlled diabetes frequently exacerbates lung infection, thereby complicating treatment strategies. Recent studies have shown that exendin-4 exhibits not only hypoglycemic but also anti-inflammatory properties. This study aimed to explore the role of exendin-4 in lung infection with diabetes, as well as its association with NOD1/NF-κB and the T1R2/T1R3 sweet taste receptor. METHODS 16HBE human bronchial epithelial cells cultured with 20 mM glucose were stimulated with lipopolysaccharide (LPS) isolated from Pseudomonas aeruginosa (PA). Furthermore, Sprague‒Dawley rats were fed a high-fat diet, followed by intraperitoneal injection of streptozotocin and intratracheal instillation of PA. The levels of TNF-α, IL-1β and IL-6 were evaluated using ELISAs and RT‒qPCR. The expression of T1R2, T1R3, NOD1 and NF-κB p65 was assayed using western blotting and immunofluorescence staining. Pathological changes in the lungs of the rats were observed using hematoxylin and eosin (H&E) staining. RESULTS At the same dose of LPS, the 20 mM glucose group produced more proinflammatory cytokines (TNF-α, IL-1β and IL-6) and had higher levels of T1R2, T1R3, NOD1 and NF-κB p65 than the normal control group (with 5.6 mM glucose). However, preintervention with exendin-4 significantly reduced the levels of the aforementioned proinflammatory cytokines and signaling molecules. Similarly, diabetic rats infected with PA exhibited increased levels of proinflammatory cytokines in their lungs and increased expression of T1R2, T1R3, NOD1 and NF-κB p65, and these effects were reversed by exendin-4. CONCLUSIONS Diabetic hyperglycemia can exacerbate inflammation during lung infection, promote the increase in NOD1/NF-κB, and promote T1R2/T1R3. Exendin-4 can ameliorate PA-related pneumonia with diabetes and overexpression of NOD1/NF-κB. Additionally, exendin-4 suppresses T1R2/T1R3, potentially through its hypoglycemic effect or through a direct mechanism. The correlation between heightened expression of T1R2/T1R3 and an intensified inflammatory response in lung infection with diabetes requires further investigation.
Collapse
Affiliation(s)
- Shanjun Yu
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570102, China
- Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, Hainan, 570102, China
| | - Chaoqun Xu
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570102, China
- Emergency and Trauma College, Hainan Medical University, Haikou, Hainan, 579199, China
| | - Xiang Tang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570102, China
- Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, Hainan, 570102, China
| | - Lijun Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570102, China
- Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, Hainan, 570102, China
| | - Lihua Hu
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570102, China
- Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, Hainan, 570102, China
| | - Liang Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570102, China
- Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, Hainan, 570102, China
| | - Xiangdong Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570102, China.
- Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, Hainan, 570102, China.
| | - Qi Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570102, China.
- Hainan Province Clinical Medical Center of Respiratory Disease, Haikou, Hainan, 570102, China.
| |
Collapse
|
6
|
Kutumova EO, Akberdin IR, Egorova VS, Kolesova EP, Parodi A, Pokrovsky VS, Zamyatnin, Jr AA, Kolpakov FA. Physiologically based pharmacokinetic model for predicting the biodistribution of albumin nanoparticles after induction and recovery from acute lung injury. Heliyon 2024; 10:e30962. [PMID: 38803942 PMCID: PMC11128879 DOI: 10.1016/j.heliyon.2024.e30962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/02/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024] Open
Abstract
The application of nanomedicine in the treatment of acute lung injury (ALI) has great potential for the development of new therapeutic strategies. To gain insight into the kinetics of nanocarrier distribution upon time-dependent changes in tissue permeability after ALI induction in mice, we developed a physiologically based pharmacokinetic model for albumin nanoparticles (ANP). The model was calibrated using data from mice treated with intraperitoneal LPS (6 mg/kg), followed by intravenous ANP (0.5 mg/mouse or about 20.8 mg/kg) at 0.5, 6, and 24 h. The simulation results reproduced the experimental observations and indicated that the accumulation of ANP in the lungs increased, reaching a peak 6 h after LPS injury, whereas it decreased in the liver, kidney, and spleen. The model predicted that LPS caused an immediate (within the first 30 min) dramatic increase in lung and kidney tissue permeability, whereas splenic tissue permeability gradually increased over 24 h after LPS injection. This information can be used to design new therapies targeting specific organs affected by bacterial infections and potentially by other inflammatory insults.
Collapse
Affiliation(s)
- Elena O. Kutumova
- Department of Computational Biology, Sirius University of Science and Technology, 354340, Sirius, Krasnodar Region, Russia
- Laboratory of Bioinformatics, Federal Research Center for Information and Computational Technologies, 630090, Novosibirsk, Russia
- Biosoft.Ru, Ltd., 630058, Novosibirsk, Russia
| | - Ilya R. Akberdin
- Department of Computational Biology, Sirius University of Science and Technology, 354340, Sirius, Krasnodar Region, Russia
- Biosoft.Ru, Ltd., 630058, Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090, Novosibirsk, Russia
| | - Vera S. Egorova
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, 354340, Sirius, Krasnodar Region, Russia
| | - Ekaterina P. Kolesova
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, 354340, Sirius, Krasnodar Region, Russia
| | - Alessandro Parodi
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, 354340, Sirius, Krasnodar Region, Russia
| | - Vadim S. Pokrovsky
- N.N. Blokhin Medical Research Center of Oncology, 115522, Moscow, Russia
- Patrice Lumumba People's Friendship University, 117198, Moscow, Russia
| | - Andrey A. Zamyatnin, Jr
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, 354340, Sirius, Krasnodar Region, Russia
- Faculty of Bioengineering and Bioinformatics and Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119234, Moscow, Russia
- Department of Biological Chemistry, Sechenov First Moscow State Medical University, 119991, Moscow, Russia
| | - Fedor A. Kolpakov
- Department of Computational Biology, Sirius University of Science and Technology, 354340, Sirius, Krasnodar Region, Russia
- Laboratory of Bioinformatics, Federal Research Center for Information and Computational Technologies, 630090, Novosibirsk, Russia
- Biosoft.Ru, Ltd., 630058, Novosibirsk, Russia
| |
Collapse
|
7
|
Marchese G, Bungaro E, Magliocca A, Fumagalli F, Merigo G, Semeraro F, Mereto E, Babini G, Roman-Pognuz E, Stirparo G, Cucino A, Ristagno G. Acute Lung Injury after Cardiopulmonary Resuscitation: A Narrative Review. J Clin Med 2024; 13:2498. [PMID: 38731027 PMCID: PMC11084269 DOI: 10.3390/jcm13092498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/15/2024] [Accepted: 04/21/2024] [Indexed: 05/13/2024] Open
Abstract
Although cardiopulmonary resuscitation (CPR) includes lifesaving maneuvers, it might be associated with a wide spectrum of iatrogenic injuries. Among these, acute lung injury (ALI) is frequent and yields significant challenges to post-cardiac arrest recovery. Understanding the relationship between CPR and ALI is determinant for refining resuscitation techniques and improving patient outcomes. This review aims to analyze the existing literature on ALI following CPR, emphasizing prevalence, clinical implications, and contributing factors. The review seeks to elucidate the pathogenesis of ALI in the context of CPR, assess the efficacy of CPR techniques and ventilation strategies, and explore their impact on post-cardiac arrest outcomes. CPR-related injuries, ranging from skeletal fractures to severe internal organ damage, underscore the complexity of managing post-cardiac arrest patients. Chest compression, particularly when prolonged and vigorous, i.e., mechanical compression, appears to be a crucial factor contributing to ALI, with the concept of cardiopulmonary resuscitation-associated lung edema (CRALE) gaining prominence. Ventilation strategies during CPR and post-cardiac arrest syndrome also play pivotal roles in ALI development. The recognition of CPR-related lung injuries, especially CRALE and ALI, highlights the need for research on optimizing CPR techniques and tailoring ventilation strategies during and after resuscitation.
Collapse
Affiliation(s)
- Giuseppe Marchese
- UOC Anestesia e Rianimazione, Ospedale Nuovo di Legnano, ASST Ovest Milanese, 20025 Legnano, Italy
| | - Elisabetta Bungaro
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy (A.M.); (E.M.)
- Department of Anesthesiology, Intensive Care and Emergency, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (G.M.)
| | - Aurora Magliocca
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy (A.M.); (E.M.)
| | - Francesca Fumagalli
- Department of Acute Brain and Cardiovascular Injury, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20122 Milan, Italy
| | - Giulia Merigo
- Department of Anesthesiology, Intensive Care and Emergency, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (G.M.)
- Department of Biomedical Sciences for Health, University of Milan, 20122 Milan, Italy
| | - Federico Semeraro
- Department of Anesthesia, Intensive Care and Prehospital Emergency, Maggiore Hospital Carlo Alberto Pizzardi, 40133 Bologna, Italy
| | - Elisa Mereto
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy (A.M.); (E.M.)
- Department of Anesthesiology, Intensive Care and Emergency, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (G.M.)
| | - Giovanni Babini
- Department of Anesthesiology, Intensive Care and Emergency, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (G.M.)
| | - Erik Roman-Pognuz
- Department of Anesthesia and Intensive Care, University of Trieste, 34127 Trieste, Italy
| | | | - Alberto Cucino
- Department of Anaesthesia and Intensive Care Medicine, APSS, Provincia Autonoma di Trento, 38121 Trento, Italy;
| | - Giuseppe Ristagno
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy (A.M.); (E.M.)
- Department of Anesthesiology, Intensive Care and Emergency, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (G.M.)
| |
Collapse
|
8
|
Darkwah S, Kotey FCN, Ahenkorah J, Adutwum-Ofosu KK, Donkor ES. Sepsis-Related Lung Injury and the Complication of Extrapulmonary Pneumococcal Pneumonia. Diseases 2024; 12:72. [PMID: 38667530 PMCID: PMC11049144 DOI: 10.3390/diseases12040072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/18/2023] [Accepted: 11/26/2023] [Indexed: 04/28/2024] Open
Abstract
Globally, sepsis and pneumonia account for significant mortality and morbidity. A complex interplay of immune-molecular pathways underlies both sepsis and pneumonia, resulting in similar and overlapping disease characteristics. Sepsis could result from unmanaged pneumonia. Similarly, sepsis patients have pneumonia as a common complication in the intensive care unit. A significant percentage of pneumonia is misdiagnosed as septic shock. Therefore, our knowledge of the clinical relationship between pneumonia and sepsis is imperative to the proper management of these syndromes. Regarding pathogenesis and etiology, pneumococcus is one of the leading pathogens implicated in both pneumonia and sepsis syndromes. Growing evidence suggests that pneumococcal pneumonia can potentially disseminate and consequently induce systemic inflammation and severe sepsis. Streptococcus pneumoniae could potentially exploit the function of dendritic cells (DCs) to facilitate bacterial dissemination. This highlights the importance of pathogen-immune cell crosstalk in the pathophysiology of sepsis and pneumonia. The role of DCs in pneumococcal infections and sepsis is not well understood. Therefore, studying the immunologic crosstalk between pneumococcus and host immune mediators is crucial to elucidating the pathophysiology of pneumonia-induced lung injury and sepsis. This knowledge would help mitigate clinical diagnosis and management challenges.
Collapse
Affiliation(s)
- Samuel Darkwah
- Department of Medical Microbiology, University of Ghana Medical School, Accra P.O. Box KB 4236, Ghana; (F.C.N.K.); (E.S.D.)
| | - Fleischer C. N. Kotey
- Department of Medical Microbiology, University of Ghana Medical School, Accra P.O. Box KB 4236, Ghana; (F.C.N.K.); (E.S.D.)
| | - John Ahenkorah
- Department of Anatomy, University of Ghana Medical School, Accra P.O. Box KB 4236, Ghana; (J.A.); (K.K.A.-O.)
| | - Kevin Kofi Adutwum-Ofosu
- Department of Anatomy, University of Ghana Medical School, Accra P.O. Box KB 4236, Ghana; (J.A.); (K.K.A.-O.)
| | - Eric S. Donkor
- Department of Medical Microbiology, University of Ghana Medical School, Accra P.O. Box KB 4236, Ghana; (F.C.N.K.); (E.S.D.)
| |
Collapse
|
9
|
Osorio-Valencia S, Zhou B. Roles of Macrophages and Endothelial Cells and Their Crosstalk in Acute Lung Injury. Biomedicines 2024; 12:632. [PMID: 38540245 PMCID: PMC10968255 DOI: 10.3390/biomedicines12030632] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/08/2024] [Accepted: 03/10/2024] [Indexed: 11/11/2024] Open
Abstract
Acute lung injury (ALI) and its severe form, acute respiratory distress syndrome (ARDS), present life-threatening conditions characterized by inflammation and endothelial injury, leading to increased vascular permeability and lung edema. Key players in the pathogenesis and resolution of ALI are macrophages (Mφs) and endothelial cells (ECs). The crosstalk between these two cell types has emerged as a significant focus for potential therapeutic interventions in ALI. This review provides a brief overview of the roles of Mφs and ECs and their interplay in ALI/ARDS. Moreover, it highlights the significance of investigating perivascular macrophages (PVMs) and immunomodulatory endothelial cells (IMECs) as crucial participants in the Mφ-EC crosstalk. This sheds light on the pathogenesis of ALI and paves the way for innovative treatment approaches.
Collapse
Affiliation(s)
| | - Bisheng Zhou
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA;
| |
Collapse
|
10
|
Landeck T, Schwarz H, Hammermüller S, Noreikat K, Reske S, Gottschaldt U, Nestler C, Wolf SJ, Ramm J, Lange M, Wrigge H, Girrbach F, Brehm W, Reske AW. High positive end-expiratory pressure ventilation mitigates the progression from unilateral pulmonary contusion to ARDS: An animal study. J Trauma Acute Care Surg 2024; 96:287-296. [PMID: 37335128 DOI: 10.1097/ta.0000000000004077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
BACKGROUND Pulmonary contusion (PC) is common in severely traumatized patients and can lead to respiratory failure requiring mechanical ventilation (MV). Ventilator-induced lung injury (VILI) might aggravate lung damage. Despite underrepresentation of trauma patients in trials on lung-protective MV, results are extrapolated to these patients, potentially disregarding important pathophysiological differences. METHODS Three MV protocols with different positive end-expiratory pressure (PEEP) levels: ARDSnetwork lower PEEP (ARDSnet-low), ARDSnetwork higher PEEP (ARDSnet-high), and open lung concept (OLC) were applied in swine for 24 hours following PC. Gas exchange, lung mechanics, quantitative computed tomography, and diffuse alveolar damage (DAD) score were analyzed. Results are given as median (interquartile range) at 24 hours. Statistical testing was performed using general linear models (group effect) over all measurement points and pairwise Mann-Whitney U tests for DAD. RESULTS There were significant differences between groups: PEEP ( p < 0.0001) ARDSnet-low (8 [8-10] cmH 2 O), ARDSnet-high (12 [12-12] cmH 2 O), OLC (21 [20-22] cmH 2 O). The fraction of arterial partial pressure of oxygen and inspired oxygen fraction ( p = 0.0016) was lowest in ARDSnet-low (78 (73-111) mm Hg) compared with ARDSnet-high (375 (365-423) mm Hg) and OLC (499 (430-523) mm Hg). The end-expiratory lung volume (EELV) differed significantly ( p < 0.0001), with highest values in OLC (64% [60-70%]) and lowest in ARDSnet-low (34% [24-37%]). Costa's surrogate for mechanical power differed significantly ( p < 0.0001), with lowest values for ARDSnet-high (73 [58-76]) compared with OLC (105 [108-116]). Diffuse alveolar damage was lower in ARDSnet-high compared with ARDSnet-low (0.0007). CONCLUSION Progression to ARDS, 24 hours after PC, was mitigated by OLC and ARDSnet-high. Both concepts restored EELV. ARDSnet-high had the lowest mechanical power surrogate and DAD. Our data suggest, that ARDSnet-high restored oxygenation and functional lung volume and reduced physiological and histological surrogates for VILI. ARDSnet-low generated unfavorable outcomes, such as loss of EELV, increased mechanical power and DAD after PC in swine. The high respiratory rate in the OLC may blunt favorable effects of lung recruitment.
Collapse
Affiliation(s)
- Tobias Landeck
- From the Department of Anesthesiology and Intensive Care Medicine (T.L., H.S., S.H., M.L.), University Hospital Leipzig; Department of Pediatric Cardiology and Intensive Care Medicine (H.S.), Georg-August-University Göttingen, University Medical Center, Göttingen; Department of Anesthesiology, Intensive Care Medicine, Emergency Medicine, and Pain Therapy (T.L., U.G., C.N., S.J.W., A.W.R.), Heinrich-Braun-Hospital, Zwickau; Innovation Center Computer Assisted Surgery (ICCAS) (T.L., U.G., A.W.R.), University of Leipzig, Medical Faculty, Leipzig, Germany; Bonitas Holding GmbH (S.H.), Herford; Medizinisch-Experimentelles Zentrum (K.N.), University of Leipzig, Medical Faculty, Leipzig; Department Diagnostic and Interventional Radiology (S.R.), Heinrich-Braun-Hospital Zwickau, Zwickau; Department of Urology (J.R.), Sana Kliniken Leipziger Land, Borna; Department of Anesthesiology, Intensive Care and Emergency Medicine, Pain Therapy (H.W.), Bergmannstrost Hospital Halle; Martin-Luther-University of Halle-Wittenberg (H.W.), Halle; Department of Anesthesiology and Intensive Care Medicine (F.G.), University Hospital Augsburg, Augsburg; Deutsche Fachpflege Holding, GmbH (S.H.), Herford; and Department for Horses, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany (W.B.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Wen X, Cheng M, Song Z, Hu J, Liang X, Lang W, Yang M, Zhou R, Hao Y. Molecular mechanism of honeysuckle + forsythia in treatment of acute lung injury based on network pharmacology. Biomed Rep 2024; 20:32. [PMID: 38273899 PMCID: PMC10809323 DOI: 10.3892/br.2024.1720] [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: 12/30/2022] [Accepted: 08/08/2023] [Indexed: 01/27/2024] Open
Abstract
The pathogenesis of acute lung injury (ALI) is complex and it is a common critical illness in clinical practice, seriously threatening the lives of critically ill patients, for which no specific molecular marker exists and there is a lack of effective methods for the treatment of ALI. The present study aimed to investigate the mechanism of action of honeysuckle and forsythia in treatment of acute lung injury (ALI) based on network pharmacology and in vitro modeling. The active ingredients and targets of honeysuckle and forsythia were predicted using traditional Chinese medicine systems pharmacology, PubChem and Swiss Target Prediction databases, and the Cytoscape 3.7.2 software was used to construct a drug-component-potential target network. The potential targets were imported into the Search tool for recurring instances of neighboring genes) database to obtain protein-protein interactions and subjected to Gene Ontology and Kyoto Encyclopedia of Genes and Targets analysis using Database for Annotation, Visualization, and Integrated Discovery. AutoDock Vina 1.1.2 software was used for docking between key active ingredients and the target proteins to analyze the binding ability of the active ingredients to the primary targets in honeysuckle and forsythia. A total of 64 male BALB/c mice were randomly divided into control, model, positive drug (Lianhua-Qingwen capsule), honeysuckle, forsythia, honeysuckle + forsythia high-, medium- and low-dose groups. Lipopolysaccharide (LPS) was used to induced an ALI model. The lung tissues of the mice were stained with hematoxylin-eosin and the serum levels of malondialdehyde (MDA) and the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) were measured 4 h after the LPS administration. Reverse transcription-quantitative PCR and western blotting were used to detect NF-κB mRNA and protein expression, respectively. Active ingredients of honeysuckle and forsythia acted on 265 common targets in ALI, which regulated NF-κB, tumor necrosis factor-α (TNF-α) and PI3K-AKT signaling pathway, HIF-1 signalling pathway to slow the inflammatory response in treatment of ALI. In the positive drug group, honeysuckle, forsythia group, honeysuckle + forsythia high-, medium- and low-dose groups, lung tissue damage were significantly decrease compared with the model group, and inflammatory cell infiltration was reduced. Compared with the model group, honeysuckle + forsythia groups experienced decreased damage caused by the LPS and inflammation in the lung tissues and significantly decreased TNF-α and NF-κB and MDA concentration and significantly increased the SOD and GSH-Px activities. The mechanism of the effect of honeysuckle and forsythia on ALI may be mediated by inhibition of TNF-α and NF-κB expression and the activation of antioxidant mechanisms to decrease production of pro-inflammatory cytokines in lung tissue, thus treating ALI.
Collapse
Affiliation(s)
- Xin Wen
- Shaanxi University of Chinese Medicine, Co-construction Collaborative Innovation Center for Chinese Medicine Resources Industrialization by Shaanxi and Education Ministry, State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), Xianyang, Shaanxi 712046, P.R. China
| | - Min Cheng
- Shaanxi University of Chinese Medicine, Co-construction Collaborative Innovation Center for Chinese Medicine Resources Industrialization by Shaanxi and Education Ministry, State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), Xianyang, Shaanxi 712046, P.R. China
- Shangluo University, College of Biomedical and Food Engineering, Shangluo, Shaanxi 726000, P.R. China
| | - Zhongxing Song
- Shaanxi University of Chinese Medicine, Co-construction Collaborative Innovation Center for Chinese Medicine Resources Industrialization by Shaanxi and Education Ministry, State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), Xianyang, Shaanxi 712046, P.R. China
| | - Jinhang Hu
- Shaanxi University of Chinese Medicine, Co-construction Collaborative Innovation Center for Chinese Medicine Resources Industrialization by Shaanxi and Education Ministry, State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), Xianyang, Shaanxi 712046, P.R. China
| | - Xuhu Liang
- Shangluo University, College of Biomedical and Food Engineering, Shangluo, Shaanxi 726000, P.R. China
| | - Wuying Lang
- Shangluo University, College of Biomedical and Food Engineering, Shangluo, Shaanxi 726000, P.R. China
| | - Mengqi Yang
- Shaanxi University of Chinese Medicine, Co-construction Collaborative Innovation Center for Chinese Medicine Resources Industrialization by Shaanxi and Education Ministry, State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), Xianyang, Shaanxi 712046, P.R. China
| | - Ruina Zhou
- Shaanxi University of Chinese Medicine, Co-construction Collaborative Innovation Center for Chinese Medicine Resources Industrialization by Shaanxi and Education Ministry, State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), Xianyang, Shaanxi 712046, P.R. China
| | - Yunjing Hao
- Northwest University, College of Life Sciences, Xian, Shaanxi 710075, P.R. China
| |
Collapse
|
12
|
Zafar I, Manzoor S, Mariappan N, Ahmad S, Athar M, Antony V, Ahmad A. A Murine Model of Vesicant-Induced Acute Lung Injury. J Pharmacol Exp Ther 2024; 388:568-575. [PMID: 38050084 PMCID: PMC10801773 DOI: 10.1124/jpet.123.001780] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 10/06/2023] [Accepted: 10/13/2023] [Indexed: 12/06/2023] Open
Abstract
Burn injuries including those caused by chemicals can result in systemic effects and acute lung injury (ALI). Cutaneous exposure to Lewisite, a warfare and chemical burn agent, also causes ALI. To overcome the limitations in conducting direct research on Lewisite-induced ALI in a laboratory setting, an animal model was developed using phenylarsine oxide (PAO) as a surrogate for Lewisite. Due to lack of a reliable animal model mimicking the effects of such exposures, development of effective therapies to treat such injuries is challenging. We demonstrated that a single cutaneous exposure to PAO resulted in disruption of the alveolar-capillary barrier as evidenced by elevated protein levels in the bronchoalveolar lavage fluid (BALF). BALF supernatant of PAO-exposed animals had increased levels of high mobility group box 1, a damage associated molecular pattern molecule. Arterial blood-gas measurements showed decreased pH, increased PaCO2, and decreased partial pressure of arterial O2, indicative of respiratory acidosis, hypercapnia, and hypoxemia. Increased protein levels of interleukin (IL)-6, CXCL-1, CXCL-2, CXCL-5, granulocyte-macrophage colony-stimulating factor, CXCL-10, leukemia inhibitory factor, leptin, IL-18, CCL-2, CCL-3, and CCL-7 were observed in the lung of PAO-exposed mice. Further, vascular endothelial growth factor levels were reduced in the lung. Pulmonary function evaluated using a flexiVent showed a downward shift in the pressure-volume loop, decreases in static compliance and inspiratory capacity, increases in respiratory elastance and tissue elastance. These changes are consistent with an ALI phenotype. These results demonstrate that cutaneous PAO exposure leads to ALI and that the model can be used as an effective surrogate to investigate vesicant-induced ALI. SIGNIFICANCE STATEMENT: This study presents a robust model for studying ALI resulting from cutaneous exposure to PAO, a surrogate for the toxic vesicating agent Lewisite. The findings in this study mimic the effects of cutaneous Lewisite exposure, providing a reliable model for investigating mechanisms underlying toxicity. The model can also be used to develop medical countermeasures to mitigate ALI associated with cutaneous Lewisite exposure.
Collapse
Affiliation(s)
- Iram Zafar
- Departments of Anesthesiology and Perioperative Medicine (I.Z., S.M., S.A., A.A.), Department of Dermatology (M.A.), and Department of Medicine (V.A.), University of Alabama at Birmingham, Birmingham, Alabama
| | - Shajer Manzoor
- Departments of Anesthesiology and Perioperative Medicine (I.Z., S.M., S.A., A.A.), Department of Dermatology (M.A.), and Department of Medicine (V.A.), University of Alabama at Birmingham, Birmingham, Alabama
| | - Nithya Mariappan
- Departments of Anesthesiology and Perioperative Medicine (I.Z., S.M., S.A., A.A.), Department of Dermatology (M.A.), and Department of Medicine (V.A.), University of Alabama at Birmingham, Birmingham, Alabama
| | - Shama Ahmad
- Departments of Anesthesiology and Perioperative Medicine (I.Z., S.M., S.A., A.A.), Department of Dermatology (M.A.), and Department of Medicine (V.A.), University of Alabama at Birmingham, Birmingham, Alabama
| | - Mohammad Athar
- Departments of Anesthesiology and Perioperative Medicine (I.Z., S.M., S.A., A.A.), Department of Dermatology (M.A.), and Department of Medicine (V.A.), University of Alabama at Birmingham, Birmingham, Alabama
| | - Veena Antony
- Departments of Anesthesiology and Perioperative Medicine (I.Z., S.M., S.A., A.A.), Department of Dermatology (M.A.), and Department of Medicine (V.A.), University of Alabama at Birmingham, Birmingham, Alabama
| | - Aftab Ahmad
- Departments of Anesthesiology and Perioperative Medicine (I.Z., S.M., S.A., A.A.), Department of Dermatology (M.A.), and Department of Medicine (V.A.), University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
13
|
Leiphrakpam PD, Weber HR, Foster KW, Buesing KL. Intraperitoneal oxygen microbubble therapy: A novel approach to enhance systemic oxygenation in a smoke inhalation model of acute hypoxic respiratory failure. Surg Open Sci 2023; 16:127-133. [PMID: 37881512 PMCID: PMC10594636 DOI: 10.1016/j.sopen.2023.09.020] [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: 05/13/2023] [Revised: 08/02/2023] [Accepted: 09/17/2023] [Indexed: 10/27/2023] Open
Abstract
Background Patients suffering from severe acute respiratory distress syndrome (ARDS) face limited therapeutic options and alarmingly high mortality rates. Refractory hypoxemia, a hallmark of ARDS, often necessitates invasive and high-risk treatments. Oxygen microbubbles (OMB) present a promising approach for extrapulmonary oxygenation, potentially augmenting systemic oxygen levels without exposing patients to significant risks. Methods Rats with severe, acute hypoxemia secondary to wood smoke inhalation (SI) received intraperitoneal (IP) bolus injections of escalating weight-by-volume (BW/V) OMB doses or normal saline to determine optimal dosage and treatment efficacy. Subsequently, a 10 % BW/V OMB bolus or saline was administered to a group of SI rats and a control group of healthy rats (SHAM). Imaging, vital signs, and laboratory studies were compared at baseline, post-smoke inhalation, and post-treatment. Histological examination and lung tissue wet/dry weight ratios were assessed at study conclusion. Results Treatment with various OMB doses in SI-induced acute hypoxemia revealed that a 10 % BW/V OMB dose significantly augmented systemic oxygen levels while minimizing dose volume. The second set of studies demonstrated a significant increase in partial pressure of arterial oxygen (PaO2) and normalization of heart rate with OMB treatment in the SI group compared to saline treatment or control group treatment. Conclusions This study highlights the successful augmentation of systemic oxygenation following OMB treatment in a small animal model of severe hypoxemia. OMB therapy emerges as a novel and promising treatment modality with immense translational potential for oxygenation support in acute care settings.
Collapse
Affiliation(s)
- Premila D. Leiphrakpam
- University of Nebraska Medical Center College of Medicine, Department of Surgery, Omaha, NE, USA
| | - Hannah R. Weber
- University of Nebraska Medical Center College of Medicine, Department of Surgery, Omaha, NE, USA
| | - Kirk W. Foster
- University of Nebraska Medical Center College of Medicine, Department of Pathology and Microbiology, Omaha, NE, USA
| | - Keely L. Buesing
- University of Nebraska Medical Center College of Medicine, Department of Surgery, Omaha, NE, USA
| |
Collapse
|
14
|
Al-Sofyani KA. Corticosteroids treatment for pediatric acute respiratory syndrome: A critical review. Saudi Med J 2023; 44:440-449. [PMID: 37182909 PMCID: PMC10187748 DOI: 10.15537/smj.2023.44.5.20220672] [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: 05/16/2023] Open
Abstract
Approximately 25% of all pediatric consultations are due to respiratory conditions, 10% of which are for asthma. Regarding prevalence, bronchiolitis, acute bronchitis, and respiratory infections are other leading pediatric respiratory illnesses. Compared to the aforementioned diseases, pediatric acute respiratory distress syndrome (PARDS) is rare but lethal in the Intensive Care Unit patients. According to global studies, the mortality in PARDS ranges from 13.3% to 60.7%. Before the Pediatric Acute Lung Injury Consensus Conference (PALICC), adult acute respiratory distress syndrome (ARDS) management guidelines were used for PARDS. The PALICC set new criteria to identify PARDS with a different treatment and management approach. Steroids have been used to treat ARDS in some cases, although their effectiveness in treating pediatric patients is highly debated in the scientific community. This review examines steroid use in treating PARDS, emphasizes current developments in the field, and gives a broad overview of PARDS management.
Collapse
Affiliation(s)
- Khouloud A. Al-Sofyani
- From the Department of Pediatric, Pediatric Critical Care Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| |
Collapse
|
15
|
Ho JW, Quan C, Gauger MA, Alam HB, Li Y. ROLE OF PEPTIDYLARGININE DEIMINASE AND NEUTROPHIL EXTRACELLULAR TRAPS IN INJURIES: FUTURE NOVEL DIAGNOSTICS AND THERAPEUTIC TARGETS. Shock 2023; 59:247-255. [PMID: 36597759 PMCID: PMC9957939 DOI: 10.1097/shk.0000000000002052] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
ABSTRACT Injuries lead to an early systemic inflammatory state with innate immune system activation. Neutrophil extracellular traps (NETs) are a complex of chromatin and proteins released from the activated neutrophils. Although initially described as a response to bacterial infections, NETs have also been identified in the sterile postinjury inflammatory state. Peptidylarginine deiminases (PADs) are a group of isoenzymes that catalyze the conversion of arginine to citrulline, termed citrullination or deimination. PAD2 and PAD4 have been demonstrated to play a role in NET formation through citrullinated histone 3. PAD2 and PAD4 have a variety of substrates with variable organ distribution. Preclinical and clinical studies have evaluated the role of PADs and NETs in major trauma, hemorrhage, burns, and traumatic brain injury. Neutrophil extracellular trap formation and PAD activation have been shown to contribute to the postinjury inflammatory state leading to a detrimental effect on organ systems. This review describes our current understanding of the role of PAD and NET formation following injury and burn. This is a new field of study, and the emerging data appear promising for the future development of targeted biomarkers and therapies in trauma.
Collapse
Affiliation(s)
- Jessie W. Ho
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Chao Quan
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI
| | - Megan A. Gauger
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Hasan B. Alam
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Yongqing Li
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
16
|
Kaczyńska K, Jampolska M, Wojciechowski P, Sulejczak D, Andrzejewski K, Zając D. Potential of Lactoferrin in the Treatment of Lung Diseases. Pharmaceuticals (Basel) 2023; 16:192. [PMID: 37259341 PMCID: PMC9960651 DOI: 10.3390/ph16020192] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 11/07/2023] Open
Abstract
Lactoferrin (LF) is a multifunctional iron-binding glycoprotein that exhibits a variety of properties, such as immunomodulatory, anti-inflammatory, antimicrobial, and anticancer, that can be used to treat numerous diseases. Lung diseases continue to be the leading cause of death and disability worldwide. Many of the therapies currently used to treat these diseases have limited efficacy or are associated with side effects. Therefore, there is a constant pursuit for new drugs and therapies, and LF is frequently considered a therapeutic agent and/or adjunct to drug-based therapies for the treatment of lung diseases. This article focuses on a review of the existing and most up-to-date literature on the contribution of the beneficial effects of LF on the treatment of lung diseases, including asthma, viral infections, cystic fibrosis, or lung cancer, among others. Although in vitro and in vivo studies indicate significant potency of LF in the treatment of the listed diseases, only in the case of respiratory tract infections do human studies seem to confirm them by demonstrating the effectiveness of LF in reducing episodes of illness and shortening the recovery period. For lung cancer, COVID-19 and sepsis, the reports are conflicting, and for other diseases, there is a paucity of human studies conclusively confirming the beneficial effects of LF.
Collapse
Affiliation(s)
- Katarzyna Kaczyńska
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland
| | - Monika Jampolska
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland
| | - Piotr Wojciechowski
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland
| | - Dorota Sulejczak
- Department of Experimental Pharmacology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland
| | - Kryspin Andrzejewski
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland
| | - Dominika Zając
- Department of Respiration Physiology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland
| |
Collapse
|
17
|
Zhang X, Wu D, Tian Y, Chen X, Lan J, Wei F, Li Y, Luo Y, Sun X. Ganoderma lucidum polysaccharides ameliorate lipopolysaccharide-induced acute pneumonia via inhibiting NRP1-mediated inflammation. PHARMACEUTICAL BIOLOGY 2022; 60:2201-2209. [PMID: 36373992 PMCID: PMC9665083 DOI: 10.1080/13880209.2022.2142615] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/16/2022] [Accepted: 10/27/2022] [Indexed: 06/16/2023]
Abstract
CONTEXT Ganoderma lucidum polysaccharides (GLP), from Ganoderma lucidum (Leyss. ex Fr.) Karst. (Ganodermataceae), are reported to have anti-inflammatory effects, including anti-neuroinflammation and anti-colitis. Nevertheless, the role of GLP in acute pneumonia is unknown. OBJECTIVE To explore the protective role of GLP against LPS-induced acute pneumonia and investigate possible mechanisms. MATERIALS AND METHODS GLP were extracted and used for high-performance liquid chromatography (HPLC) analysis after acid hydrolysis and PMP derivatization. Sixty C57BL/6N male mice were randomly divided into six groups: Sham, Model, LPS + GLP (25, 50 and 100 mg/kg/d administered intragastrically for two weeks) and LPS + dexamethasone (6 mg/kg/d injected intraperitoneally for one week). Acute pneumonia mouse models were established by intratracheal injection of LPS. Haematoxylin and eosin (H&E) staining was examined to evaluate lung lesions. ELISA and quantitative real-time PCR were employed to assess inflammatory factors expression. Western blots were carried out to measure Neuropilin-1 expression and proteins related to apoptosis and autophagy. RESULTS GLP suppressed inflammatory cell infiltration. In BALF, cell counts were 1.1 × 106 (model) and 7.1 × 105 (100 mg/kg). Release of GM-CSF and IL-6 was reduced with GLP (25, 50 and 100 mg/kg) treatment. The expression of genes IL-1β, IL-6, TNF-α and Saa3 was reduced. GLP treatment also suppressed the activation of Neuropilin-1 (NRP1), upregulated the levels of Bcl2/Bax and LC3 and led to downregulation of the ratio C-Caspase 3/Caspase 3 and P62 expression. DISCUSSION AND CONCLUSIONS GLP could protect against LPS-induced acute pneumonia through multiple mechanisms: blocking the infiltration of inflammatory cells, inhibiting cytokine secretion, suppressing NRP1 activation and regulating pneumonocyte apoptosis and autophagy.
Collapse
Affiliation(s)
- Xuelian Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Daoshun Wu
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yu Tian
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Xiangdong Chen
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jin Lan
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Fei Wei
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Ye Li
- Ganoherb (Fujian) Technology Corporation, Nanping, China
| | - Yun Luo
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
18
|
Osawa EA, Kleiman JFF, Maciel AT. Acute respiratory distress syndrome following administration of gadolinium contrast agent: a case report. J Med Case Rep 2022; 16:421. [PMCID: PMC9668706 DOI: 10.1186/s13256-022-03643-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/11/2022] [Indexed: 11/18/2022] Open
Abstract
Background Gadolinium-based contrast agents are used extensively in magnetic resonance imaging to assist diagnosis of medical conditions. Despite their documented safety profile, severe adverse events do occur, and their documentation may serve to raise the awareness of the medical community. Case presentation We report the case of a 15-year-old white Latin American female patient admitted to the intensive care unit for acute respiratory distress syndrome following administration of gadolinium. She did not have rash or tongue swelling but developed hypotension responsive to fluid administration and severe hypoxemia. Chest computed tomography revealed bilateral pulmonary compromise with multiple confluent consolidations. She received methylprednisolone and noninvasive ventilatory support including bilevel positive airway pressure ventilation and high-flow nasal cannula, and underwent a rapid recovery. Conclusion Gadolinium-based contrast agent-induced acute respiratory distress syndrome, albeit rare, should be included in the differential diagnosis of respiratory failure shortly after magnetic resonance imaging, which is nowadays a frequent diagnostic procedure, potentially increasing the awareness of this serious complication.
Collapse
Affiliation(s)
- Eduardo Atsushi Osawa
- Imed Group Research Department, Av. Angélica, 2.530 - 9º andar, São Paulo, SP 01228-200 Brazil ,grid.477346.5Adult Intensive Care Unit, São Camilo Hospital, Pompeia Unit. Av. Pompéia, 1178, São Paulo, SP 05022-001 Brazil
| | | | - Alexandre Toledo Maciel
- Imed Group Research Department, Av. Angélica, 2.530 - 9º andar, São Paulo, SP 01228-200 Brazil ,grid.477346.5Adult Intensive Care Unit, São Camilo Hospital, Pompeia Unit. Av. Pompéia, 1178, São Paulo, SP 05022-001 Brazil
| |
Collapse
|
19
|
Dong T, Chen X, Xu H, Song Y, Wang H, Gao Y, Wang J, Du R, Lou H, Dong T. Mitochondrial metabolism mediated macrophage polarization in chronic lung diseases. Pharmacol Ther 2022; 239:108208. [DOI: 10.1016/j.pharmthera.2022.108208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/01/2022] [Accepted: 05/09/2022] [Indexed: 11/30/2022]
|
20
|
Ni YL, Shen HT, Chen SP, Kuan YH. Protective effect of genkwanin against lipopolysaccharide-induced acute lung injury in mice with p38 mitogen-activated protein kinase and nuclear factor-κB pathway inhibition. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
21
|
Huang P, Zhang J, Duan W, Jiao J, Leng A, Qu J. Plant polysaccharides with anti-lung injury effects as a potential therapeutic strategy for COVID-19. Front Pharmacol 2022; 13:982893. [DOI: 10.3389/fphar.2022.982893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
When coronavirus disease 2019 (COVID-19) develops into the severe phase, lung injury, acute respiratory distress syndrome, and/or respiratory failure could develop within a few days. As a result of pulmonary tissue injury, pathomorphological changes usually present endothelial dysfunction, inflammatory cell infiltration of the lung interstitium, defective gas exchange, and wall leakage. Consequently, COVID-19 may progress to tremendous lung injury, ongoing lung failure, and death. Exploring the treatment drugs has important implications. Recently, the application of traditional Chinese medicine had better performance in reducing fatalities, relieving symptoms, and curtailing hospitalization. Through constant research and study, plant polysaccharides may emerge as a crucial resource against lung injury with high potency and low side effects. However, the absence of a comprehensive understanding of lung-protective mechanisms impedes further investigation of polysaccharides. In the present article, a comprehensive review of research into plant polysaccharides in the past 5 years was performed. In total, 30 types of polysaccharides from 19 kinds of plants have shown lung-protective effects through the pathological processes of inflammation, oxidative stress, apoptosis, autophagy, epithelial–mesenchymal transition, and immunomodulation by mediating mucin and aquaporins, macrophage, endoplasmic reticulum stress, neutrophil, TGF-β1 pathways, Nrf2 pathway, and other mechanisms. Moreover, the deficiencies of the current studies and the future research direction are also tentatively discussed. This research provides a comprehensive perspective for better understanding the mechanism and development of polysaccharides against lung injury for the treatment of COVID-19.
Collapse
|
22
|
Lampova B, Doskocil I, Kourimska L, Kopec A. N-3 polyunsaturated fatty acids may affect the course of COVID-19. Front Immunol 2022; 13:957518. [PMID: 36238306 PMCID: PMC9551352 DOI: 10.3389/fimmu.2022.957518] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/08/2022] [Indexed: 11/29/2022] Open
Abstract
The highly infectious coronavirus disease (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is a new coronavirus that has been spreading since late 2019 and has caused millions of deaths worldwide. COVID-19 continues to spread rapidly worldwide despite high vaccination coverage; therefore, it is crucial to focus on prevention. Most patients experience only mild symptoms of COVID-19. However, in some cases, serious complications can develop mainly due to an exaggerated immune response; that is, a so-called cytokine storm, which can lead to acute respiratory distress syndrome, organ failure, or, in the worst cases, death. N-3 polyunsaturated fatty acids and their metabolites can modulate inflammatory responses, thus reducing the over-release of cytokines. It has been hypothesized that supplementation of n-3 polyunsaturated fatty acids could improve clinical outcomes in critically ill COVID-19 patients. Some clinical trials have shown that administering n-3 polyunsaturated fatty acids to critically ill patients can improve their health and shorten the duration of their stay in intensive care. However, previous clinical studies have some limitations; therefore, further studies are required to confirm these findings.
Collapse
Affiliation(s)
- Barbora Lampova
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Prague, Czechia
| | - Ivo Doskocil
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Prague, Czechia
- *Correspondence: Ivo Doskocil,
| | - Lenka Kourimska
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Prague, Czechia
| | - Aneta Kopec
- Department of Human Nutrition and Dietetics, Faculty of Food Technology, The University of Agriculture in Krakow, Krakow, Poland
| |
Collapse
|
23
|
Wang R, Feng R, Xia C, Ruan F, Luo P, Guo J. Early detection of gram‑negative bacteria using metagenomic next‑generation sequencing in acute respiratory distress syndrome: A case report. Exp Ther Med 2022; 24:573. [PMID: 35949316 PMCID: PMC9353542 DOI: 10.3892/etm.2022.11510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/27/2022] [Indexed: 11/29/2022] Open
Abstract
Metagenomic next-generation sequencing (mNGS) is an effective method that can be used for the identification of early pathogens in patients with suspected severe pneumonia. However, the potential of mNGS for evaluating the prognosis of acute respiratory distress syndrome (ARDS) in patients with severe pneumonia remains unclear. In the present report, hospital-acquired gram-negative bacteria infections were detected in a case using metagenomic next-generation sequencing (mNGS) in a sample of bronchoalveolar fluid. This was obtained from a 58-year-old male patient with traumatic wet lung after a neurosurgery. According to the results, of which the profiles of the resistance genes were detected by mNGS, drugs designed to control infection were adjusted, namely to polymyxin B (500,000 U/12 h), azithromycin (0.5 g/24 h) and ganciclovir (0.25 g/12 h). Following adjusting treatment for 8 days, the symptoms of lung infection and hypoxemia were markedly improved, resulting in the patient being transferred out of the intensive care unit 15 days after treatment. To conclude, observations from the present report suggest that mNGS is a useful method for the early identification of pathogens in patients with pneumonia caused by ARDS. However, further studies are required to identify the complementary role of mNGS in supporting conventional microbiological methods in routine clinical practice.
Collapse
Affiliation(s)
- Rong Wang
- Department of Critical Medicine, Union Jiangbei Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430100, P.R. China
| | - Rong Feng
- Shanghai Topgen Biomedical Technology Co., Ltd., Shanghai 201318, P.R. China
| | - Chaoran Xia
- Shanghai Topgen Biomedical Technology Co., Ltd., Shanghai 201318, P.R. China
| | - Fangying Ruan
- Shanghai Topgen Biomedical Technology Co., Ltd., Shanghai 201318, P.R. China
| | - Peng Luo
- Shanghai Topgen Biomedical Technology Co., Ltd., Shanghai 201318, P.R. China
| | - Jun Guo
- Department of Critical Medicine, Union Jiangbei Hospital, Huazhong University of Science and Technology, Wuhan, Hubei 430100, P.R. China
| |
Collapse
|
24
|
Elma B, Mammadov R, Bilgin Y, Yazıcı GN, Süleyman H, Çoban TA. The effect of rutin on pulmonary contusion induced by blunt trauma in rats: Biochemical and histopathological evaluation. TURK GOGUS KALP DAMAR CERRAHISI DERGISI 2022; 30:431-439. [PMID: 36303692 PMCID: PMC9580287 DOI: 10.5606/tgkdc.dergisi.2022.21992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/13/2021] [Indexed: 06/16/2023]
Abstract
BACKGROUND This study aims to investigate the possible protective effects of rutin, also called vitamin P1, against pulmonary contusion induced by blunt chest trauma in a rat model. METHODS Thirty male albino Wistar rats were separated into three equal groups as healthy group, trauma group, and trauma+rutin group. After anesthesia provided by intraperitoneal administration of 60 mg/kg ketamine and xylazine by inhalation at appropriate intervals, 200 g weight was dropped from 1 m height to the anterior chest wall of the animals in the trauma group (n=10) and trauma+rutin group (n=10) and pulmonary contusion was created. Thirty min after the trauma, 50 mg/kg of rutin was administered into the stomach of trauma+rutin group animals orally with gavage. The rats received rutin once daily for two days and were sacrificed 48 h later. Their lung tissues were removed and examined biochemically and histopathologically. RESULTS Nuclear factor-kappa B, cyclooxygenase-2, and malondialdehyde levels increased in the trauma group compared to the healthy group, and rutin administration prevented this increase. Total glutathione levels decreased in the trauma group, and rutin administration also prevented this decrease. The histopathological findings were compatible with the biochemical findings. CONCLUSION Our study results suggest that rutin has a protective effect on contused lung tissue in rats.
Collapse
Affiliation(s)
- Bekir Elma
- Department of Thoracic Surgery, Medicine Faculty of Erzincan Binali Yıldırım University, Erzincan, Türkiye
| | - Renad Mammadov
- Department of Pharmacology, Medicine Faculty of Erzincan Binali Yıldırım University, Erzincan, Türkiye
| | - Yasin Bilgin
- Emergency Health Services, Erzincan Health Directorate, Erzincan, Türkiye
| | - Gülce Naz Yazıcı
- Department of Histology and Embryology, Medicine Faculty of Erzincan Binali Yıldırım University, Erzincan, Türkiye
| | - Halis Süleyman
- Department of Pharmacology, Medicine Faculty of Erzincan Binali Yıldırım University, Erzincan, Türkiye
| | - Taha Abdulkadir Çoban
- Department of Biochemistry, Medicine Faculty of Erzincan Binali Yıldırım University, Erzincan, Türkiye
| |
Collapse
|
25
|
Perilla Fruit Water Extract Attenuates Inflammatory Responses and Alleviates Neutrophil Recruitment via MAPK/JNK-AP-1/c-Fos Signaling Pathway in ARDS Animal Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4444513. [PMID: 35815275 PMCID: PMC9262517 DOI: 10.1155/2022/4444513] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 06/10/2022] [Indexed: 11/18/2022]
Abstract
Airway respiratory distress syndrome (ARDS) is usually caused by a severe pulmonary infection. However, there is currently no effective treatment for ARDS. Traditional Chinese medicine (TCM) has been shown to effectively treat inflammatory lung diseases, but a clear mechanism of action of TCM is not available. Perilla fruit water extract (PFWE) has been used to treat cough, excessive mucus production, and some pulmonary diseases. Thus, we propose that PFWE may be able to reduce lung inflammation and neutrophil infiltration in a lipopolysaccharide (LPS)-stimulated murine model. C57BL/6 mice were stimulated with LPS (10 μg/mouse) by intratracheal (IT) injection and treated with three doses of PFWE (2, 5, and 8 g/kg) by intraperitoneal (IP) injections. To investigate possible mechanisms, A549 cells were treated with PFWE and stimulated with LPS. Our results showed that PFWE decreased airway resistance, neutrophil infiltration, vessel permeability, and interleukin (IL)-6 and chemokine (C-C motif) ligand 2 (CCL2/MCP-1) expressions in vivo. In addition, the PFWE inhibited the expression of IL-6, CCL2/MCP-1, chemokine (CXC motif) ligand 1 (CXCL1/GROα), and IL-8 in vitro. Moreover, PFWE also inhibited the MAPK/JNK-AP-1/c-Fos signaling pathway in A549 cells. In conclusion, we demonstrated that PFWE attenuated pro-inflammatory cytokine and chemokine levels and downregulated neutrophil recruitment through the MAPK/JNK-AP-1/c-Fos pathway. Thus, PFWE can be a potential drug to assist the treatment of ARDS.
Collapse
|
26
|
Predictors of mortality in trauma patients with acute respiratory distress syndrome receiving extracorporeal membrane oxygenation. SURGERY IN PRACTICE AND SCIENCE 2022. [DOI: 10.1016/j.sipas.2022.100071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
27
|
Gihring A, Gärtner F, Schirmer M, Wabitsch M, Knippschild U. Recent Developments in Mouse Trauma Research Models: A Mini-Review. Front Physiol 2022; 13:866617. [PMID: 35574493 PMCID: PMC9101050 DOI: 10.3389/fphys.2022.866617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/04/2022] [Indexed: 12/02/2022] Open
Abstract
The urgency to investigate trauma in a controlled and reproducible environment rises since multiple trauma still account for the most deaths for people under the age of 45. The most common multiple trauma include head as well as blunt thorax trauma along with fractures. However, these trauma remain difficult to treat, partially because the molecular mechanisms that trigger the immediate immune response are not fully elucidated. To illuminate these mechanisms, investigators have used animal models, primarily mice as research subjects. This mini review aims to 1) emphasize the importance of the development of clinically relevant murine trauma research, 2) highlight and discuss the existing conflict between simulating clinically relevant situations and elucidating molecular mechanisms, 3) describe the advantages and disadvantages of established mouse trauma models developed to simulate clinically relevant situations, 4) summarize and list established mouse models in the field of trauma research developed to simulate clinically relevant situations.
Collapse
Affiliation(s)
- Adrian Gihring
- Department of General and Visceral Surgery, Surgery Center, Ulm University Medical Center, Ulm, Germany
| | - Fabian Gärtner
- Department of General and Visceral Surgery, Surgery Center, Ulm University Medical Center, Ulm, Germany
| | - Melanie Schirmer
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Surgery Center, Ulm University Medical Center, Ulm, Germany
- *Correspondence: Uwe Knippschild,
| |
Collapse
|
28
|
Scozzi D, Liao F, Krupnick AS, Kreisel D, Gelman AE. The role of neutrophil extracellular traps in acute lung injury. Front Immunol 2022; 13:953195. [PMID: 35967320 PMCID: PMC9374003 DOI: 10.3389/fimmu.2022.953195] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/28/2022] [Indexed: 12/14/2022] Open
Abstract
Acute lung injury (ALI) is a heterogeneous inflammatory condition associated with high morbidity and mortality. Neutrophils play a key role in the development of different forms of ALI, and the release of neutrophil extracellular traps (NETs) is emerging as a common pathogenic mechanism. NETs are essential in controlling pathogens, and their defective release or increased degradation leads to a higher risk of infection. However, NETs also contain several pro-inflammatory and cytotoxic molecules than can exacerbate thromboinflammation and lung tissue injury. To reduce NET-mediated lung damage and inflammation, DNase is frequently used in preclinical models of ALI due to its capability of digesting NET DNA scaffold. Moreover, recent advances in neutrophil biology led to the development of selective NET inhibitors, which also appear to reduce ALI in experimental models. Here we provide an overview of the role of NETs in different forms of ALI discussing existing gaps in our knowledge and novel therapeutic approaches to modulate their impact on lung injury.
Collapse
Affiliation(s)
- Davide Scozzi
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, United States
| | - Fuyi Liao
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, United States
| | | | - Daniel Kreisel
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, United States
| | - Andrew E. Gelman
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, United States
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, United States
- *Correspondence: Andrew E. Gelman,
| |
Collapse
|
29
|
Zhao Y, Cui L, Yang XX, Sun X, Liu Y, Yang Z, Zhu L, Peng C, Li D, Cai J, Ma Y. Sinoacutine inhibits inflammatory responses to attenuates acute lung injury by regulating NF-κB and JNK signaling pathways. BMC Complement Med Ther 2021; 21:284. [PMID: 34801005 PMCID: PMC8605577 DOI: 10.1186/s12906-021-03458-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/03/2021] [Indexed: 12/22/2022] Open
Abstract
Background Stephania yunnanensis H. S. Lo is widely used as an antipyretic, analgesic and anti-inflammatory herbal medicine in SouthWest China. In this study, we investigated the anti-inflammatory activity and mechanism of sinoacutine (sino), one of the primary components extracted from this plant. Methods A RAW264.7 cell model was established using lipopolysaccharide (LPS) induced for estimation of cytokines in vitro, qPCR was used to estimate gene expression, western blot analysis was used to estimate protein level and investigate the regulation of NF- κB, JNK and MAPK signal pathway. In addition, an acute lung injury model was established to determine lung index and levels of influencing factors. Results Using the RAW264.7 model, we found that sino reduced levels of nitric oxide (NO), tumour necrosis factor-α (TNF-α), interleukin (IL)-1β and prostaglandin E2 (PGE2) but increased levels of IL-6. qPCR analysis revealed that sino (50, 25 μg/ml) inhibited gene expression of nitric oxide synthase (iNOS). western blot analysis showed that sino significantly inhibited protein levels of both iNOS and COX-2. Further signalling pathway analysis validated that sino also inhibited phosphorylation of p65 in the NF-κB and c-Jun NH2 terminal kinase (JNK) signalling pathways but promoted the phosphorylation of extracellular signal regulated kinase (ERK) and p38 in the MAPK signalling pathway. In addition, in a mouse model induced by LPS, we determined that sino reduced the lung index and the levels of myeloperoxidase (MPO), NO, IL-6 and TNF-α in lung tissues and bronchoalveolar lavage fluid (BALF) in acute lung injury (ALI). Conclusion Taken together, our results demonstrate that sino is a promising drug to alleviate LPS-induced inflammatory reactions. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-021-03458-0.
Collapse
Affiliation(s)
- Yuancui Zhao
- School of Chinese Material Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China.,Key Laboratory of External Drug Delivery System and Preparation Technology in University of Yunnan, Kunming, 650500, China
| | - Lili Cui
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210046, China
| | - Xing Xin Yang
- Key Laboratory of Southern Medicine Utilization, Kunming, 650500, China
| | - Xingqian Sun
- School of Chinese Material Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China.,Yunnan Key Laboratory of Dai and Yi Medicine, School of Chinese Material Medicine Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Yunkuan Liu
- School of Chinese Material Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China.,Key Laboratory of External Drug Delivery System and Preparation Technology in University of Yunnan, Kunming, 650500, China
| | - Zixian Yang
- School of Chinese Material Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Liyuan Zhu
- School of Chinese Material Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Chaorui Peng
- School of Chinese Material Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Danye Li
- School of Chinese Material Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Junfei Cai
- School of Chinese Material Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Yunshu Ma
- School of Chinese Material Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, China. .,Yunnan Key Laboratory of Dai and Yi Medicine, School of Chinese Material Medicine Yunnan University of Chinese Medicine, Kunming, 650500, China.
| |
Collapse
|
30
|
Leiphrakpam PD, Weber HR, McCain A, Matas RR, Duarte EM, Buesing KL. A novel large animal model of smoke inhalation-induced acute respiratory distress syndrome. Respir Res 2021; 22:198. [PMID: 34233680 PMCID: PMC8261975 DOI: 10.1186/s12931-021-01788-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/28/2021] [Indexed: 12/31/2022] Open
Abstract
Background Acute respiratory distress syndrome (ARDS) is multifactorial and can result from sepsis, trauma, or pneumonia, amongst other primary pathologies. It is one of the major causes of death in critically ill patients with a reported mortality rate up to 45%. The present study focuses on the development of a large animal model of smoke inhalation-induced ARDS in an effort to provide the scientific community with a reliable, reproducible large animal model of isolated toxic inhalation injury-induced ARDS. Methods Animals (n = 21) were exposed to smoke under general anesthesia for 1 to 2 h (median smoke exposure = 0.5 to 1 L of oak wood smoke) after the ultrasound-guided placement of carotid, pulmonary, and femoral artery catheters. Peripheral oxygen saturation (SpO2), vital signs, and ventilator parameters were monitored throughout the procedure. Chest x-ray, carotid, femoral and pulmonary artery blood samples were collected before, during, and after smoke exposure. Animals were euthanized and lung tissue collected for analysis 48 h after smoke inhalation. Results Animals developed ARDS 48 h after smoke inhalation as reflected by a decrease in SpO2 by approximately 31%, PaO2/FiO2 ratio by approximately 208 (50%), and development of bilateral, diffuse infiltrates on chest x-ray. Study animals also demonstrated a significant increase in IL-6 level, lung tissue injury score and wet/dry ratio, as well as changes in other arterial blood gas (ABG) parameters. Conclusions This study reports, for the first time, a novel large animal model of isolated smoke inhalation-induced ARDS without confounding variables such as cutaneous burn injury. Use of this unique model may be of benefit in studying the pathophysiology of inhalation injury or for development of novel therapeutics.
Collapse
Affiliation(s)
- Premila D Leiphrakpam
- Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-3280, USA
| | - Hannah R Weber
- Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-3280, USA
| | - Andrea McCain
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Roser Romaguera Matas
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Ernesto Martinez Duarte
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Keely L Buesing
- Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198-3280, USA.
| |
Collapse
|
31
|
Hon KL, Leung KKY, Oberender F, Leung AK. Paediatrics: how to manage acute respiratory distress syndrome. Drugs Context 2021; 10:dic-2021-1-9. [PMID: 34122589 PMCID: PMC8177958 DOI: 10.7573/dic.2021-1-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 03/17/2021] [Indexed: 12/13/2022] Open
Abstract
Background Acute respiratory distress syndrome (ARDS) is a significant cause of mortality and morbidity amongst critically ill children. The purpose of this narrative review is to provide an up-to-date review on the evaluation and management of paediatric ARDS (PARDS). Methods A PubMed search was performed with Clinical Queries using the key term "acute respiratory distress syndrome". The search strategy included clinical trials, meta-analyses, randomized controlled trials, observational studies and reviews. Google, Wikipedia and UpToDate were also searched to enrich the review. The search was restricted to the English literature and children. Discussion Non-invasive positive pressure ventilation, lung-protective ventilation strategies, conservative fluid management and adequate nutritional support all have proven efficacy in the management of PARDS. The Pediatric Acute Lung Injury Consensus Conference recommends the use of corticosteroids, high-frequency oscillation ventilation and inhaled nitric oxide in selected scenarios. Partial liquid ventilation and surfactant are not considered efficacious based on evidence from clinical trials. Conclusion PARDS is a serious but relatively rare cause of admission into the paediatric intensive care unit and is associated with high mortality. Non-invasive positive pressure ventilation, lung-protective ventilation strategies, conservative fluid management and adequate nutrition are advocated. As there has been a lack of progress in the management of PARDS in recent years, further well-designed, large-scale, randomized controlled trials in this field are urgently needed.
Collapse
Affiliation(s)
- Kam Lun Hon
- Paediatric Intensive Care Unit, Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, Hong Kong
| | - Karen Ka Yan Leung
- Paediatric Intensive Care Unit, Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, Hong Kong
| | - Felix Oberender
- Paediatric Intensive Care Unit, Monash Children's Hospital, Melbourne, Australia.,Monash University, School of Clinical Sciences, Department of Paediatrics, Melbourne, Australia
| | - Alexander Kc Leung
- Department of Pediatrics, The University of Calgary and The Alberta Children's Hospital, Calgary, Alberta, Canada
| |
Collapse
|
32
|
From sepsis to acute respiratory distress syndrome (ARDS): emerging preventive strategies based on molecular and genetic researches. Biosci Rep 2021; 40:222737. [PMID: 32319516 PMCID: PMC7199454 DOI: 10.1042/bsr20200830] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/11/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
A healthy body activates the immune response to target invading pathogens (i.e. viruses, bacteria, fungi, and parasites) and avoid further systemic infection. The activation of immunological mechanisms includes several components of the immune system, such as innate and acquired immunity. Once any component of the immune response to infections is aberrantly altered or dysregulated, resulting in a failure to clear infection, sepsis will develop through a pro-inflammatory immunological mechanism. Furthermore, the severe inflammatory responses induced by sepsis also increase vascular permeability, leading to acute pulmonary edema and resulting in acute respiratory distress syndrome (ARDS). Apparently, potential for improvement exists in the management of the transition from sepsis to ARDS; thus, this article presents an exhaustive review that highlights the previously unrecognized relationship between sepsis and ARDS and suggests a direction for future therapeutic developments, including plasma and genetic pre-diagnostic strategies and interference with proinflammatory signaling.
Collapse
|
33
|
Wang W, Chen NY, Ren D, Davies J, Philip K, Eltzschig HK, Blackburn MR, Akkanti B, Karmouty-Quintana H, Weng T. Enhancing Extracellular Adenosine Levels Restores Barrier Function in Acute Lung Injury Through Expression of Focal Adhesion Proteins. Front Mol Biosci 2021; 8:636678. [PMID: 33778007 PMCID: PMC7987656 DOI: 10.3389/fmolb.2021.636678] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/01/2021] [Indexed: 12/23/2022] Open
Abstract
Background: Acute respiratory distress syndrome (ARDS) is a clinical presentation of acute lung injury (ALI) with often fatal lung complication. Adenosine, a nucleoside generated following cellular stress provides protective effects in acute injury. The levels of extracellular adenosine can be depleted by equilibrative nucleoside transporters (ENTs). ENT inhibition by pharmaceutical agent dipyridamole promotes extracellular adenosine accumulation and is protective in ARDS. However, the therapeutic potential of dipyridamole in acute lung injury has not yet been evaluated. Methods: Adenosine acts on three adenosine receptors, the adenosine A1 (Adora1), A2a (Adora2a), the A2b (Adora2b) or the adenosine A3 (Adora 3) receptor. Accumulation of adenosine is usually required to stimulate the low-affinity Adora2b receptor. In order to investigate the effect of adenosine accumulation and the contribution of epithelial-specific ENT2 or adora2b expression in experimental ALI, dipyridamole, and epithelial specific ENT2 or Adora2b deficient mice were utilized. MLE12 cells were used to probe downstream Adora2b signaling. Adenosine receptors, transporters, and targets were determined in ARDS lungs. Results: ENT2 is mainly expressed in alveolar epithelial cells and is negatively regulated by hypoxia following tissue injury. Enhancing adenosine levels with ENT1/ENT2 inhibitor dipyridamole at a time when bleomycin-induced ALI was present, reduced further injury. Mice pretreated with the ADORA2B agonist BAY 60-6583 were protected from bleomycin-induced ALI by reducing vascular leakage (558.6 ± 50.4 vs. 379.9 ± 70.4, p < 0.05), total bronchoalveolar lavage fluid cell numbers (17.9 ± 1.8 to 13.4 ± 1.4 e4, p < 0.05), and neutrophil infiltration (6.42 ± 0.25 vs. 3.94 ± 0.29, p < 0.05). While mice lacking Adora2b in AECs were no longer protected by dipyridamole. We also identified occludin and focal adhesion kinase as downstream targets of ADORA2B, thus providing a novel mechanism for adenosine-mediated barrier protection. Similarly, we also observed similar enhanced ADORA2B (3.33 ± 0.67 to 16.12 ± 5.89, p < 0.05) and decreased occludin (81.2 ± 0.3 to 13.3 ± 0.4, p < 0.05) levels in human Acute respiratory distress syndrome lungs. Conclusion: We have highlighted a role of dipyridamole and adenosine signaling in preventing or treating ALI and identified Ent2 and Adora2b as key mediators in important for the resolution of ALI.
Collapse
Affiliation(s)
- Wei Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ning-yuan Chen
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Dewei Ren
- Houston Methodist J.C. Walter Jr. Transplant Center, Houston Methodist Hospital, Houston, TX, United States
| | - Jonathan Davies
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Kemly Philip
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Holger K. Eltzschig
- Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Michael R. Blackburn
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- UTHealth Pulmonary Center of Excellence, Houston, TX, United States
| | - Bindu Akkanti
- Divisions of Critical Care, Pulmonary and Sleep Medicine, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- UTHealth Pulmonary Center of Excellence, Houston, TX, United States
- Divisions of Critical Care, Pulmonary and Sleep Medicine, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Tingting Weng
- Department of Biochemistry and Molecular Biology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- UTHealth Pulmonary Center of Excellence, Houston, TX, United States
| |
Collapse
|
34
|
Fallon EA, Chung CS, Heffernan DS, Chen Y, De Paepe ME, Ayala A. Survival and Pulmonary Injury After Neonatal Sepsis: PD1/PDL1's Contributions to Mouse and Human Immunopathology. Front Immunol 2021; 12:634529. [PMID: 33746973 PMCID: PMC7965961 DOI: 10.3389/fimmu.2021.634529] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 02/08/2021] [Indexed: 12/14/2022] Open
Abstract
Morbidity and mortality associated with neonatal sepsis remains a healthcare crisis. PD1-/- neonatal mice endured experimental sepsis, in the form of cecal slurry (CS), and showed improved rates of survival compared to wildtype (WT) counterparts. End-organ injury, particularly of the lung, contributes to the devastation set forth by neonatal sepsis. PDL1-/- neonatal mice, in contrast to PD1-/- neonatal mice did not have a significant improvement in survival after CS. Because of this, we focused subsequent studies on the impact of PD1 gene deficiency on lung injury. Here, we observed that at 24 h post-CS (but not at 4 or 12 h) there was a marked increase in pulmonary edema (PE), neutrophil influx, myeloperoxidase (MPO) levels, and cytokine expression sham (Sh) WT mice. Regarding pulmonary endothelial cell (EC) adhesion molecule expression, we observed that Zona occludens-1 (ZO-1) within the cell shifted from a membranous location to a peri-nuclear location after CS in WT murine cultured ECs at 24hrs, but remained membranous among PD1-/- lungs. To expand the scope of this inquiry, we investigated human neonatal lung tissue. We observed that the lungs of human newborns exposed to intrauterine infection had significantly higher numbers of PD1+ cells compared to specimens who died from non-infectious causes. Together, these data suggest that PD1/PDL1, a pathway typically thought to govern adaptive immune processes in adult animals, can modulate the largely innate neonatal pulmonary immune response to experimental septic insult. The potential future significance of this area of study includes that PD1/PDL1 checkpoint proteins may be viable therapeutic targets in the septic neonate.
Collapse
Affiliation(s)
- Eleanor A. Fallon
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Chun-Shiang Chung
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Daithi S. Heffernan
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
- Department of Surgery, Providence Veterans Affairs Medical Center, Providence, RI, United States
| | - Yaping Chen
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| | - Monique E. De Paepe
- Department of Pathology, Women & Infants Hospital and Alpert Medical School of Brown University, Providence, RI, United States
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| |
Collapse
|
35
|
Hemorrhagic Shock and Resuscitation Causes Glycocalyx Shedding and Endothelial Oxidative Stress Preferentially in the Lung and Intestinal Vasculature. Shock 2021; 56:803-812. [PMID: 34259440 DOI: 10.1097/shk.0000000000001764] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Hemorrhagic shock has recently been shown to cause shedding of a carbohydrate surface layer of endothelial cells known as the glycocalyx. This shedding of the glycocalyx is thought to be a mediator of the coagulopathy seen in trauma patients. Clinical studies have demonstrated increases in shed glycocalyx in the blood after trauma, and animal studies have measured glycocalyx disruption in blood vessels in the lung, skeletal muscle, and mesentery. However, no study has measured glycocalyx disruption across a wide range of vascular beds to quantify the primary locations of this shedding. METHODS In the present study, we used a rat model of hemorrhagic shock and resuscitation to more comprehensively assess glycocalyx disruption across a range of organs. Glycocalyx disruption was assessed by fluorescent-labelled wheat germ agglutinin or syndecan-1 antibody staining in flash frozen tissue. RESULTS We found that our model did elicit glycocalyx shedding, as assessed by an increase in plasma syndecan-1 levels. In tissue sections, we found that the greatest glycocalyx disruption occurred in vessels in the lung and intestine. Shedding to a lesser extent was observed in vessels of the brain, heart, and skeletal muscle. Liver vessel glycocalyx was unaffected, and kidney vessels, including the glomerular capillaries, displayed an increase in glycocalyx. We also measured reactive oxygen species (ROS) in the endothelial cells from these organs, and found that the greatest increase in ROS occurred in the two beds with the greatest glycocalyx shedding, the lungs and intestine. We also detected fibrin deposition in lung vessels following hemorrhage-resuscitation. CONCLUSIONS We conclude that the endothelium in the lungs and intestine are particularly susceptible to the oxidative stress of hemorrhage-resuscitation, as well as the resulting glycocalyx disruption. Thus these two vessel beds may be important drivers of coagulopathy in trauma patients.
Collapse
|
36
|
Wang Y, Li H, Li X, Su X, Xiao H, Yang J. Hypoxic Preconditioning of Human Umbilical Cord Mesenchymal Stem Cells Is an Effective Strategy for Treating Acute Lung Injury. Stem Cells Dev 2021; 30:128-134. [PMID: 33349130 DOI: 10.1089/scd.2020.0174] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS)/acute lung injury (ALI) is a severe clinical respiratory failure disorder associated with chronic pathology and disability and has a mortality rate of 40%-60%. However, the pathogenesis of ARDS/ALI remains unclear, and existing therapeutic options are insufficient for addressing the severity of the disease. Mesenchymal stem cells (MSCs) play an important role in the prevention and treatment of ALI, especially acute alveolar epithelial injury. However, the low survival rate of transplanted MSCs reduces their effectiveness. When human umbilical cord MSCs (hUC-MSCs) are transplanted directly, only a minority of cells migrate toward damaged tissues. Moreover, their maintenance time is short, leading to unsatisfactory therapeutic results. A moderate hypoxic environment can promote the proliferation of MSCs, inhibit apoptosis, and facilitate migration and chemotaxis. In summary, hypoxic culturing before transplantation improves the effectiveness of hUC-MSCs in treating ARDS/ALI and promises to provide novel diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Yujuan Wang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Han Li
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Xue Li
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Xin Su
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Han Xiao
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Junling Yang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
37
|
Xing D, Chen YH, Wang LT, Yu B, Ran ZB, Chen L. Evaluation of the therapeutic effect of high-flow nasal cannula oxygen therapy on patients with aspiration pneumonia accompanied by respiratory failure in the post-stroke sequelae stage. BMC Pulm Med 2021; 21:17. [PMID: 33413281 PMCID: PMC7788538 DOI: 10.1186/s12890-020-01359-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/26/2020] [Indexed: 11/22/2022] Open
Abstract
Background The aim of the present study was to evaluate the therapeutic effect of high-flow nasal cannula (HFNC) oxygen therapy on patients with aspiration pneumonia accompanied by respiratory failure in the post-stroke sequelae stage, with the goal of providing more effective oxygen therapy and improving patient prognosis. Methods Retrospective analysis was conducted on 103 elderly patients with post-stroke aspiration pneumonia and moderate respiratory failure (oxygenation index: 100–200 mmHg) that had been admitted. The patients were divided into two groups according to the mode of oxygen therapy that was used: the Venturi mask group and the HFNC treatment group. The two groups were analyzed and compared in terms of the changes in the blood gas indices measured at different points in time (4, 8, 12, 24, 48, and 72 h), the proportion of patients that required transition to invasive auxiliary ventilation, and the 28-day mortality rate. Results A total of 103 patients were retrospectively analyzed; 16 cases were excluded, and 87 patients were included in the final patient group (42 in the HFNC group and 45 in the Venturi group). There was a statistically significant difference in the oxygenation indices of the HFNC group and the Venturi group (F = 546.811, P < 0.05). There was a statistically significant interaction between the monitored oxygenation indices and the mode of oxygen therapy (F = 70.961, P < 0.05), and there was a statistically significant difference in the oxygenation indices for the two modes of oxygen therapy (F = 256.977, P < 0.05). HFNC therapy contributed to the improvement of the oxygenation indices at a rate of 75.1%. The Venturi and HFNC groups also differed significantly in terms of the proportion of patients that required transition to invasive auxiliary ventilation within 72 h (P < 0.05). The HFNC group’s risk for invasive ventilation was 0.406 times that of the Venturi group (P < 0.05). There was no statistical difference in the 28-day mortality rate of the two groups (P > 0.05). Conclusion HFNC could significantly improve the oxygenation state of patients with post-stroke aspiration pneumonia and respiratory failure, and it may reduce the incidence of invasive ventilation.
Collapse
Affiliation(s)
- Dong Xing
- Department of Emergency, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Yu-Hong Chen
- Intensive Care Unit, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Lan -Tao Wang
- Department of Emergency, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Bin Yu
- Department of Emergency, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Zhi -Bin Ran
- Department of Neurology, Shijiazhuang Great Wall Hospital, Shijiazhuang, 050000, China
| | - Li Chen
- Department of General Medicine, The Fourth Hospital of Hebei Medical University, No. 12 of Jiankang Road, Chang'an District, Shijiazhuang, 050011, China.
| |
Collapse
|
38
|
Taghavi S, Ali A, Green E, Schmitt K, Jackson-Weaver O, Tatum D, Harris C, Guidry C, McGrew P, Schroll R, Kolls J, Duchesne J. Surgical stabilization of rib fractures is associated with improved survival but increased acute respiratory distress syndrome. Surgery 2020; 169:1525-1531. [PMID: 33461776 PMCID: PMC8039755 DOI: 10.1016/j.surg.2020.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/02/2020] [Accepted: 12/08/2020] [Indexed: 11/25/2022]
Abstract
Background How the surgical stabilization of rib fractures after trauma affects the development of acute respiratory distress syndrome and impacts survival has yet to be determined in a large database. We hypothesized that surgical stabilization of rib fractures would not decrease the incidence of acute respiratory distress syndrome. Methods The National Trauma Data Bank was queried for all traumatic rib fractures in 2016. Patients were divided into groups with single rib fractures, multiple rib fractures, and flail chest. Nonoperative therapy was compared with stabilization of rib fractures of 1 to 2 ribs or 3+ ribs. Results There were 114,972 total patients with rib fractures meeting inclusion criteria, with 5,106 (4.4%) having flail chest, 24,726 (21.5%) having single rib fractures, and 85,140 (74.1%) having multiple rib fractures. Those with flail chest (15.9%) were most likely to get rib plating in comparison to multiple rib fractures (0.9%) and single rib fractures (0.2%); P < .001. On logistic regression, surgical stabilization of rib fractures 1 to 2 ribs (odds ratio: 0.17, 95% confidence interval: 0.10–0.28) or 3+ ribs (odds ratio: 0.17, 95% confidence interval: 0.11–0.28), with nonoperative therapy as the reference was associated with survival. Variables associated with mortality included increasing age, male sex, increasing injury severity score, decreased Glasgow coma scale, requirement of transfusions, and hypotension on admission. Surgical stabilization of rib fractures 3+ ribs (odds ratio: 2.30, 95% confidence interval: 1.58–3.37) was associated with acute respiratory distress syndrome but not 1 to 2 ribs (odd ratio: 1.55, 95% confidence interval: 0.97–2.48). On logistic regression of only patients with flail chest, stabilization of rib fractures was associated with decreased mortality but not increased risk of acute respiratory distress syndrome. Conclusion The increased risk of acute respiratory distress syndrome should be considered in the preoperative assessment for stabilization of rib fractures.
Collapse
Affiliation(s)
- Sharven Taghavi
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA.
| | - Ayman Ali
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA
| | - Erik Green
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA
| | - Kyle Schmitt
- Department of Surgery, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Olan Jackson-Weaver
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA
| | - Danielle Tatum
- Trauma Specialists Program, Our Lady of the Lake Regional Medical Center, Baton Rouge, LA, (d)Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA
| | - Charles Harris
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA
| | - Chrissy Guidry
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA
| | - Patrick McGrew
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA
| | - Rebecca Schroll
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA
| | - Jay Kolls
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA
| | - Juan Duchesne
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA
| |
Collapse
|
39
|
Lupu L, Palmer A, Huber-Lang M. Inflammation, Thrombosis, and Destruction: The Three-Headed Cerberus of Trauma- and SARS-CoV-2-Induced ARDS. Front Immunol 2020; 11:584514. [PMID: 33101314 PMCID: PMC7546394 DOI: 10.3389/fimmu.2020.584514] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/10/2020] [Indexed: 01/05/2023] Open
Abstract
Physical trauma can be considered an unrecognized "pandemic" because it can occur anywhere and affect anyone and represents a global burden. Following severe tissue trauma, patients frequently develop acute lung injury (ALI) and/or acute respiratory distress syndrome (ARDS) despite modern surgical and intensive care concepts. The underlying complex pathophysiology of life-threatening ALI/ARDS has been intensively studied in experimental and clinical settings. However, currently, the coronavirus family has become the focus of ALI/ARDS research because it represents an emerging global public health threat. The clinical presentation of the infection is highly heterogeneous, varying from a lack of symptoms to multiple organ dysfunction and mortality. In a particular subset of patients, the primary infection progresses rapidly to ALI and ARDS. The pathophysiological mechanisms triggering and driving severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced ALI/ARDS are still poorly understood. Although it is also generally unknown whether insights from trauma-induced ARDS may be readily translated to SARS-CoV-2-associated ARDS, it was still recommended to treat coronavirus-positive patients with ALI/ARDS with standard protocols for ALI/ARDS. However, this strategy was questioned by clinical scientists, because it was documented that some severely hypoxic SARS-CoV-2-infected patients exhibited a normal respiratory system compliance, a phenomenon rarely observed in ARDS patients with another underlying etiology. Therefore, coronavirus-induced ARDS was defined as a specific ARDS phenotype, which accordingly requires an adjusted therapeutic approach. These suggestions reflect previous attempts of classifying ARDS into different phenotypes that might overall facilitate ARDS diagnosis and treatment. Based on the clinical data from ARDS patients, two major phenotypes have been proposed: hyper- and hypo-inflammatory. Here, we provide a comparative review of the pathophysiological pathway of trauma-/hemorrhagic shock-induced ARDS and coronavirus-induced ARDS, with an emphasis on the crucial key points in the pathogenesis of both these ARDS forms. Therefore, the manifold available data on trauma-/hemorrhagic shock-induced ARDS may help to better understand coronavirus-induced ARDS.
Collapse
Affiliation(s)
| | | | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
40
|
Echinacea polysaccharide alleviates LPS-induced lung injury via inhibiting inflammation, apoptosis and activation of the TLR4/NF-κB signal pathway. Int Immunopharmacol 2020; 88:106974. [PMID: 33182056 DOI: 10.1016/j.intimp.2020.106974] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 08/24/2020] [Accepted: 09/01/2020] [Indexed: 11/23/2022]
Abstract
Lung injury is a common critical life-threatening syndrome. Inflammation is a key factor in the pathogenesis of lung injury. It is reported that Echinacea Polysaccharides (EP) has anti-inflammatory activity. However, the effect of EP on lung injury remains unclear. In our study, murine model of lung injury was induced with 2.5 mg/kg LPS before administration of 5 mg/kg or 10 mg/kg EP. EP ameliorated LPS-induced lung pathological damage, along with reduction in lung wet/dry weight ratio and myeloperoxidase activity. EP decreased the number of leukocytes, eosinophils, neutrophils, lymphocytes and macrophages in bronchoalveolar lavage fluid, and the release of tumour necrosis factor-α (TNF-α), interleukin-6 (IL-6) and interleukin-1β (IL-1β) in LPS-treated lung. EP suppressed LPS-induced apoptosis along with down-regulation of Bcl2-associated X (Bax) and cleaved caspase-3 (CC3), and elevated B-cell lymphoma-2 (Bcl-2). Besides, RAW 264.7 cells were treated with EP 100 μg/ml for 1 h and then incubated with 1 μg/ml LPS for 24 h. TNF-α, IL-6 and IL-1β levels were lowered by treatment of EP in LPS-treated RAW 264.7 cells. Moreover, EP down-regulated the expression of toll-like receptor 4 (TLR4), myeloid differentiating factor 88 (MyD88), p-IκBα, nuclear factor kappa-B (NF-κB), p-NF-κB, and up-regulated the inhibitor of NF-κB (IκBα) in vivo and in vitro following LPS induction, which is consistent with the effect of TAK-242. In conclusion, EP may alleviate LPS-induced lung injury via inhibiting inflammation, apoptosis and activation of TLR4/NF-κB signal pathway.
Collapse
|
41
|
Roncati L, Ligabue G, Nasillo V, Lusenti B, Gennari W, Fabbiani L, Malagoli C, Gallo G, Giovanella S, Lupi M, Salviato T, Paolini A, Costantini M, Trenti T, Maiorana A. A proof of evidence supporting abnormal immunothrombosis in severe COVID-19: naked megakaryocyte nuclei increase in the bone marrow and lungs of critically ill patients. Platelets 2020; 31:1085-1089. [PMID: 32857624 DOI: 10.1080/09537104.2020.1810224] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a global public health emergency with many clinical facets, and new knowledge about its pathogenetic mechanisms is deemed necessary; among these, there are certainly coagulation disorders. In the history of medicine, autopsies and tissue sampling have played a fundamental role in order to understand the pathogenesis of emerging diseases, including infectious ones; compared to the past, histopathology can be now expanded by innovative techniques and modern technologies. For the first time in worldwide literature, we provide a detailed postmortem and biopsy report on the marked increase, up to 1 order of magnitude, of naked megakaryocyte nuclei in the bone marrow and lungs from serious COVID-19 patients. Most likely related to high interleukin-6 serum levels stimulating megakaryocytopoiesis, this phenomenon concurs to explain well the pulmonary abnormal immunothrombosis in these critically ill patients, all without molecular or electron microscopy signs of megakaryocyte infection.
Collapse
Affiliation(s)
- Luca Roncati
- Institute of Pathology, University of Modena and Reggio Emilia , Modena, Italy.,Hemolymphopathology Team, University Hospital of Modena , Modena, Italy.,Immunohistochemistry Lab, University Hospital of Modena , Modena, Italy
| | - Giulia Ligabue
- Nephropathology Lab, University of Modena and Reggio Emilia , Modena, Italy
| | - Vincenzo Nasillo
- Hemolymphopathology Team, University Hospital of Modena , Modena, Italy
| | - Beatrice Lusenti
- Hemolymphopathology Team, University Hospital of Modena , Modena, Italy
| | - William Gennari
- Virology and Molecular Microbiology Lab, University Hospital of Modena , Modena, Italy
| | - Luca Fabbiani
- Institute of Pathology, University of Modena and Reggio Emilia , Modena, Italy.,Hemolymphopathology Team, University Hospital of Modena , Modena, Italy
| | - Claudia Malagoli
- Institute of Pathology, University of Modena and Reggio Emilia , Modena, Italy.,Hemolymphopathology Team, University Hospital of Modena , Modena, Italy
| | - Graziana Gallo
- Institute of Pathology, University of Modena and Reggio Emilia , Modena, Italy
| | - Silvia Giovanella
- Nephropathology Lab, University of Modena and Reggio Emilia , Modena, Italy
| | - Massimo Lupi
- Institute of Pathology, University of Modena and Reggio Emilia , Modena, Italy
| | - Tiziana Salviato
- Institute of Pathology, University of Modena and Reggio Emilia , Modena, Italy
| | - Ambra Paolini
- Hemolymphopathology Team, University Hospital of Modena , Modena, Italy
| | - Matteo Costantini
- Institute of Pathology, University of Modena and Reggio Emilia , Modena, Italy
| | - Tommaso Trenti
- Department of Laboratory Medicine, University Hospital of Modena , Modena, Italy
| | - Antonio Maiorana
- Institute of Pathology, University of Modena and Reggio Emilia , Modena, Italy
| |
Collapse
|
42
|
Impact of Chest Trauma and Overweight on Mortality and Outcome in Severely Injured Patients. J Clin Med 2020; 9:jcm9092752. [PMID: 32858822 PMCID: PMC7564760 DOI: 10.3390/jcm9092752] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/11/2020] [Accepted: 08/16/2020] [Indexed: 01/08/2023] Open
Abstract
The morbidity and mortality of severely injured patients are commonly affected by multiple factors. Especially, severe chest trauma has been shown to be a significant factor in considering outcome. Contemporaneously, weight-associated endocrinological, haematological, and metabolic deviations from the norm seem to have an impact on the posttraumatic course. Therefore, the aim of this study was to determine the influence of body weight on severely injured patients by emphasizing chest trauma. A total of 338 severely injured patients were included. Multivariate regression analyses were performed on patients with severe chest trauma (AIS ≥ 3) and patients with minor chest trauma (AIS < 3). The influence of body weight on in-hospital mortality was evaluated. Of all the patients, 70.4% were male, the median age was 52 years (IQR 36–68), the overall Injury Severity Score (ISS) was 24 points (IQR 17–29), and a median BMI of 25.1 points (IQR 23–28) was determined. In general, chest trauma was associated with prolonged ventilation, prolonged ICU treatment, and increased mortality. For overweight patients with severe chest trauma, an independent survival benefit was found (OR 0.158; p = 0.037). Overweight seems to have an impact on the mortality of severely injured patients with combined chest trauma. Potentially, a nutritive advantage or still-unknown immunological aspects in these patients affecting the intensive treatment course could be argued.
Collapse
|
43
|
Value of point-of-care ultrasonography compared with computed tomography scan in detecting potential life-threatening conditions in blunt chest trauma patients. Ultrasound J 2020; 12:36. [PMID: 32747992 PMCID: PMC7399008 DOI: 10.1186/s13089-020-00183-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 07/03/2020] [Indexed: 11/10/2022] Open
Abstract
Background Ultrasonography is a suitable modality that can potentially improve patient care, saving time and lives. Purpose This article has evaluated the caveats and pitfalls of point-of-care ultrasonography in the diagnosis of pneumothorax, hemothorax and contusion. Materials and methods This prospective study was performed in 157 patients with blunt chest trauma in 3 university hospitals. Ultrasonography was performed by 2 board-certified emergency medicine specialists and an emergency medicine resident PGY-3 after passing the training process successfully. Results The false-negative cases were not significantly correlated with accompanying traumatic injuries. Lung ultrasonography accompanied by chest physical examination show accuracy 91.8. Point-of-care ultrasonography (PoCUS) showed sensitivity 75.0%, specificity 100%, positive-predictive value (PPV) of 100% and a negative-predictive value (NPV) of 94.9% for the diagnosis of pneumothorax. For hemothorax, bedside PoCUS had a sensitivity of 45.4%, specificity of 100%, PPV of 100% and NPV of 91.8%. PoCUS was assessed 58.1% sensitive and 100% specific for detecting lung contusion with positive-predictive value (PPV) of 100% and a negative-predictive value (NPV) of 86.3%. Performing US resulted in no false-positive cases. Conclusions Point-of-care ultrasonography was highly sensitive to detect pneumothorax and can be beneficial for the disposition of stable patients and to detect PTX in unstable patients before transferring to the operating room. It is also moderately appropriate for the diagnosis of hemothorax and lung contusion compared to the gold standard, CT scan. It is essential to consider the false-negative and false-positive instances of lung ultrasound in various situations to enhance management and disposition of blunt thoracic injuries.
Collapse
|
44
|
Nguyen C, Pham T. Non-Cardiogenic Pulmonary Oedema Following the use of Gadolinium-Based Contrast Medium: A Case Report. J Crit Care Med (Targu Mures) 2020; 6:181-185. [PMID: 32864464 PMCID: PMC7430356 DOI: 10.2478/jccm-2020-0019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 04/21/2020] [Indexed: 11/20/2022] Open
Abstract
Non-cardiogenic pulmonary oedema can be life threatening and requires prompt treatment. While gadolinium-based contrast is generally considered safe with a low risk of severe side effects, non-cardiogenic pulmonary oedema has become increasingly recognised as a rare, but possibly life-threatening complication. We present a case of a usually well, young 23-year-old female who developed non-cardiogenic pulmonary oedema with a moderate oxygenation impairment and no mucosal or cutaneous features of anaphylaxis following the administration of gadolinium-based contrast. She did not respond to treatment of anaphylaxis but made a rapid recovery following the commencement of positive pressure ventilation. Our case highlights the importance of recognising the rare complication of non-cardiogenic pulmonary oedema following gadolinium-based contrast administration in order to promptly implement the appropriate treatment.
Collapse
Affiliation(s)
| | - Tho Pham
- Alfred Health, VIC, MelbourneAustralia
| |
Collapse
|
45
|
Chang JC. Acute Respiratory Distress Syndrome as an Organ Phenotype of Vascular Microthrombotic Disease: Based on Hemostatic Theory and Endothelial Molecular Pathogenesis. Clin Appl Thromb Hemost 2020; 25:1076029619887437. [PMID: 31775524 PMCID: PMC7019416 DOI: 10.1177/1076029619887437] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening noncardiogenic circulatory disorder of the lungs associated with critical illnesses such as sepsis, trauma, and immune and collagen vascular disease. Its mortality rate is marginally improved with the best supportive care. The demise occurs due to progressive pulmonary hypoxia and multi-organ dysfunction syndrome (MODS) with severe inflammation. Complement activation is a part of immune response against pathogen or insult in which membrane attack complex (MAC) is formed and eliminates microbes. If complement regulatory protein such as endothelial CD59 is underexpressed, MAC may also cause pulmonary vascular injury to the innocent bystander endothelial cell of host and provokes endotheliopathy that causes inflammation and pulmonary vascular microthrombosis, leading to ARDS. Its pathogenesis is based on a novel "two-path unifying theory" of hemostasis and "two-activation theory of the endothelium" promoting molecular pathogenesis. Endotheliopathy activates two independent molecular pathways: inflammatory and microthrombotic. The former triggers the release inflammatory cytokines and the latter promotes exocytosis of unusually large von Willebrand factor multimers (ULVWF) and platelet activation. Inflammatory pathway initiates inflammation, but microthrombotic pathway more seriously produces "microthrombi strings" composed of platelet-ULVWF complexes, which become anchored on the injured endothelial cells, and causes disseminated intravascular microthrombosis (DIT). DIT is a hemostatic disease due to lone activation of ULVWF path without activated tissue factor path. It leads to endotheliopathy-associated vascular microthrombotic disease (EA-VMTD), which orchestrates consumptive thrombocytopenia, microangiopathic hemolytic anemia, and MODS. Thrombotic thrombocytopenic purpura (TTP)-like syndrome is the hematologic phenotype of EA-VMTD. ARDS is one of organ phenotypes among MODS associated with TTP-like syndrome. The most effective treatment of ARDS can be achieved by counteracting the activated microthrombotic pathway based on two novel hemostatic theories.
Collapse
Affiliation(s)
- Jae C Chang
- Department of Medicine, University of California, Irvine School of Medicine, Irvine, CA, USA
| |
Collapse
|
46
|
Pottecher J, Noll E, Borel M, Audibert G, Gette S, Meyer C, Gaertner E, Legros V, Carapito R, Uring-Lambert B, Sauleau E, Land WG, Bahram S, Meyer A, Geny B, Diemunsch P. Protocol for TRAUMADORNASE: a prospective, randomized, multicentre, double-blinded, placebo-controlled clinical trial of aerosolized dornase alfa to reduce the incidence of moderate-to-severe hypoxaemia in ventilated trauma patients. Trials 2020; 21:274. [PMID: 32183886 PMCID: PMC7079402 DOI: 10.1186/s13063-020-4141-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 02/04/2020] [Indexed: 01/01/2023] Open
Abstract
Background Acute respiratory distress syndrome continues to drive significant morbidity and mortality after severe trauma. The incidence of trauma-induced, moderate-to-severe hypoxaemia, according to the Berlin definition, could be as high as 45%. Its pathophysiology includes the release of damage-associated molecular patterns (DAMPs), which propagate tissue injuries by triggering neutrophil extracellular traps (NETs). NETs include a DNA backbone coated with cytoplasmic proteins, which drive pulmonary cytotoxic effects. The structure of NETs and many DAMPs includes double-stranded DNA, which prevents their neutralization by plasma. Dornase alfa is a US Food and Drug Administration-approved recombinant DNase, which cleaves extracellular DNA and may therefore break up the backbone of NETs and DAMPs. Aerosolized dornase alfa was shown to reduce trauma-induced lung injury in experimental models and to improve arterial oxygenation in ventilated patients. Methods TRAUMADORNASE will be an institution-led, multicentre, double-blinded, placebo-controlled randomized trial in ventilated trauma patients. The primary trial objective is to demonstrate a reduction in the incidence of moderate-to-severe hypoxaemia in severe trauma patients during the first 7 days from 45% to 30% by providing aerosolized dornase alfa as compared to placebo. The secondary objectives are to demonstrate an improvement in lung function and a reduction in morbidity and mortality. Randomization of 250 patients per treatment arm will be carried out through a secure, web-based system. Statistical analyses will include a descriptive step and an inferential step using fully Bayesian techniques. The study was approved by both the Agence Nationale de la Sécurité du Médicament et des Produits de Santé (ANSM, on 5 October 2018) and a National Institutional Review Board (CPP, on 6 November 2018). Participant recruitment began in March 2019. Results will be published in international peer-reviewed medical journals. Discussion If early administration of inhaled dornase alfa actually reduces the incidence of moderate-to-severe hypoxaemia in patients with severe trauma, this new therapeutic strategy may be easily implemented in many clinical trauma care settings. This treatment may facilitate ventilator weaning, reduce the burden of trauma-induced lung inflammation and facilitate recovery and rehabilitation in severe trauma patients. Trial registration ClinicalTrials.gov, NCT03368092. Registered on 11 December 2017.
Collapse
Affiliation(s)
- Julien Pottecher
- Hôpitaux Universitaires de Strasbourg, Hôpital de Hautepierre, Service d'Anesthésie-Réanimation Chirurgicale, 1 Avenue Molière, 67098, Strasbourg, France. .,Université de Strasbourg, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), EA3072, 4 Rue Kirschleger, 67085, Strasbourg, France. .,Fédération Hospitalo-Universitaire OMICARE, Centre de Recherche d'Immunologie et d'Hématologie, 4 rue Kirschleger, 67085, Strasbourg Cedex, France.
| | - Eric Noll
- Hôpitaux Universitaires de Strasbourg, Hôpital de Hautepierre, Service d'Anesthésie-Réanimation Chirurgicale, 1 Avenue Molière, 67098, Strasbourg, France.,Université de Strasbourg, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), EA3072, 4 Rue Kirschleger, 67085, Strasbourg, France.,Fédération Hospitalo-Universitaire OMICARE, Centre de Recherche d'Immunologie et d'Hématologie, 4 rue Kirschleger, 67085, Strasbourg Cedex, France
| | - Marie Borel
- Sorbonne Universités, UPMC Université Paris 06, INSERM UMR_S 1158 Neurophysiologie Respiratoire Expérimentale et Clinique, AP-HP, Groupe Hospitalier Pitié-Salpêtrière Charles Foix, Département d'Anesthésie Réanimation, 47-83 Boulevard de l'Hôpital, 75651, Paris Cedex 13, France
| | - Gérard Audibert
- CHRU Nancy, Hôpital Central, Service d'Anesthésie-Réanimation, 29 Avenue de Lattre de Tassigny, 54000, Nancy, France
| | - Sébastien Gette
- CHR Metz-Thionville-Site de Mercy, Service de Réanimation Polyvalente, 1 Allée du Château, 57350, Ars-Laquenexy, France
| | - Christian Meyer
- Groupe Hospitalier de la Région de Mulhouse et Sud Alsace (GHRMSA), Pôle d'Anesthésie-Réanimation, 20 rue du Dr Laennec, 68051, Mulhouse Cedex 1, France
| | - Elisabeth Gaertner
- Hôpital Louis Pasteur, Service d'Anesthésie-Réanimation Pôle 2, 39 Avenue de la Liberté, 68024, Colmar Cedex, France
| | - Vincent Legros
- CHU de Reims, Hôpital Maison Blanche, Réanimation Chirurgicale et Traumatologique, SAMU 51, 45 rue Cognacq-Jay, 51092, Reims, France
| | - Raphaël Carapito
- Fédération Hospitalo-Universitaire OMICARE, Centre de Recherche d'Immunologie et d'Hématologie, 4 rue Kirschleger, 67085, Strasbourg Cedex, France.,Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, Laboratoire Central d'Immunologie, 1 Place de l'Hôpital, 67091, Strasbourg Cedex, France.,Université de Strasbourg, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR_S 1109, 4 rue Kirschleger, 67085, Strasbourg Cedex, France
| | - Béatrice Uring-Lambert
- Fédération Hospitalo-Universitaire OMICARE, Centre de Recherche d'Immunologie et d'Hématologie, 4 rue Kirschleger, 67085, Strasbourg Cedex, France.,Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, Laboratoire Central d'Immunologie, 1 Place de l'Hôpital, 67091, Strasbourg Cedex, France.,Université de Strasbourg, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR_S 1109, 4 rue Kirschleger, 67085, Strasbourg Cedex, France
| | - Erik Sauleau
- Hôpitaux Universitaires de Strasbourg, Hôpital Civil, Pôle Santé Publique, Groupe Méthode en Recherche Clinique (GMRC), 1 Place de l'Hôpital, 67091, Strasbourg Cedex, France
| | - Walter G Land
- Fédération Hospitalo-Universitaire OMICARE, Centre de Recherche d'Immunologie et d'Hématologie, 4 rue Kirschleger, 67085, Strasbourg Cedex, France.,Université de Strasbourg, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR_S 1109, 4 rue Kirschleger, 67085, Strasbourg Cedex, France
| | - Seiamak Bahram
- Fédération Hospitalo-Universitaire OMICARE, Centre de Recherche d'Immunologie et d'Hématologie, 4 rue Kirschleger, 67085, Strasbourg Cedex, France.,Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, Laboratoire Central d'Immunologie, 1 Place de l'Hôpital, 67091, Strasbourg Cedex, France.,Université de Strasbourg, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR_S 1109, 4 rue Kirschleger, 67085, Strasbourg Cedex, France
| | - Alain Meyer
- Université de Strasbourg, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), EA3072, 4 Rue Kirschleger, 67085, Strasbourg, France.,Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, Service de Physiologie et d'Explorations Fonctionnelles, 1 Place de l'Hôpital, 67091, Strasbourg Cedex, France
| | - Bernard Geny
- Université de Strasbourg, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), EA3072, 4 Rue Kirschleger, 67085, Strasbourg, France.,Hôpitaux Universitaires de Strasbourg, Nouvel Hôpital Civil, Service de Physiologie et d'Explorations Fonctionnelles, 1 Place de l'Hôpital, 67091, Strasbourg Cedex, France
| | - Pierre Diemunsch
- Hôpitaux Universitaires de Strasbourg, Hôpital de Hautepierre, Service d'Anesthésie-Réanimation Chirurgicale, 1 Avenue Molière, 67098, Strasbourg, France.,Université de Strasbourg, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), EA3072, 4 Rue Kirschleger, 67085, Strasbourg, France.,Fédération Hospitalo-Universitaire OMICARE, Centre de Recherche d'Immunologie et d'Hématologie, 4 rue Kirschleger, 67085, Strasbourg Cedex, France
| |
Collapse
|
47
|
Risk factors affecting post-traumatic acute respiratory distress syndrome development in thoracic trauma patients. TURK GOGUS KALP DAMAR CERRAHISI DERGISI-TURKISH JOURNAL OF THORACIC AND CARDIOVASCULAR SURGERY 2020; 27:540-549. [PMID: 32082923 DOI: 10.5606/tgkdc.dergisi.2019.18124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 07/05/2019] [Indexed: 11/21/2022]
Abstract
Background This study aims to investigate the risk factors affecting post-traumatic acute respiratory distress syndrome development in thoracic trauma patients. Methods This two-centered, retrospective study included 3,080 thoracic trauma patients (2,562 males, 518 females; mean age 33.9±19.4 years; range, 2 months to 91 years) treated between January 2005 and January 2019. Demographic characteristics, mechanisms of injury, traumatic injuries, injury severity score and new injury severity score results, treatments, comorbidities, complications, morbidity and mortality rates, and durations of hospital stay were collected. Data were used to predict the risk factors for development of post-traumatic acute respiratory distress syndrome by univariate and multivariate statistical analysis. Results Acute respiratory distress syndrome was detected in 81 patients. In multivariate logistic regression analysis; age, pulmonary contusion, intracranial hemorrhage, rib fracture (unilateral and four-five pieces), femur and tibia fracture, diabetes mellitus, chronic obstructive pulmonary disease, blood transfusion (≥3 units), high white blood cell count at admission, sepsis, and hepatic injury were detected as independent risk factors (p<0.05). Optimal cutoff points (sensitivity/specificity ratios) for acute respiratory distress syndrome development risk were ≥16 (79%/68%) for injury severity score, ≥27 (90%/68.7%) for new injury severity score, and ≥16,000 (75.3%/71.6%) for admission white blood cell count. New injury severity score was superior than injury severity score to predict the development of acute respiratory distress syndrome. Conclusion Acute respiratory distress syndrome causes significant mortality and morbidity in trauma patients. In addition to the well-known risk factors, diabetes mellitus and chronic obstructive pulmonary disease were independent risk factors. We defined a cutoff value for new injury severity score to predict post-traumatic acute respiratory distress syndrome.
Collapse
|
48
|
Morgan M, Vernon T, Bradburn EH, Miller JA, Jammula S, Rogers FB. A Comprehensive Review of the Outcome for Patients Readmitted to the ICU Following Trauma and Strategies to Decrease Readmission Rates. J Intensive Care Med 2020; 35:936-942. [PMID: 31916876 DOI: 10.1177/0885066619899639] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In recent years, there has been an emphasis on evaluating the outcomes of patients who have experienced an intensive care unit (ICU) readmission. This may in part be due to the Patient Protection and Affordable Care Act's Hospital Readmission Reduction Program which imposes financial sanctions on hospitals who have excessive readmission rates, informally known as bounceback rates. The financial cost associated with avoidable bounceback combined with the potentially preventable expenses can result in unnecessary financial strain. Within the hospital readmissions, there is a subset pertaining to unplanned readmission to the ICU. Although there have been studies regarding ICU bounceback, there are limited studies regarding ICU bounceback of trauma patients and even fewer proven strategies. Although many studies have concluded that respiratory complications were the most common factor influencing ICU readmissions, there is inconclusive evidence in terms of a broadly applicable strategy that would facilitate management of these patients. The purpose of this review is to highlight the outcomes of patients readmitted to the ICU and to provide an overview of possible strategies to aid in decreasing ICU readmission rates.
Collapse
Affiliation(s)
- Madison Morgan
- Trauma Services, 4399Penn Medicine Lancaster General Health, Lancaster, PA, USA
| | - Tawnya Vernon
- Trauma Services, 4399Penn Medicine Lancaster General Health, Lancaster, PA, USA
| | - Eric H Bradburn
- Trauma Services, 4399Penn Medicine Lancaster General Health, Lancaster, PA, USA
| | - Jo Ann Miller
- Trauma Services, 4399Penn Medicine Lancaster General Health, Lancaster, PA, USA
| | - Shreya Jammula
- Trauma Services, 4399Penn Medicine Lancaster General Health, Lancaster, PA, USA
| | - Frederick B Rogers
- Trauma Services, 4399Penn Medicine Lancaster General Health, Lancaster, PA, USA
| |
Collapse
|
49
|
Fan R, Ren Q, Zhou T, Shang L, Ma M, Wang B, Xiao C. Determination of endogenous substance change in PM 2.5-induced rat plasma and lung samples by UPLC-MS/MS method to identify potential markers for lung impairment. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:22040-22050. [PMID: 31144181 DOI: 10.1007/s11356-019-05351-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 05/01/2019] [Indexed: 06/09/2023]
Abstract
Exposure to fine particulate matter (PM2.5) could induce lung impairment aggravation. Moreover, endogenous substances are known to play a significant role in lung impairment. Therefore, the research objectives was to investigate the influence of PM2.5-induced lung impairment on the levels of the eight endogenous substances, γ-aminobutyric acid (GABA), acetylcholine (ACh), glutamate (Glu), serotonin (5-HT), 5-hydroxyindole-3-acetic acid (5-HIAA), noradrenaline (NE), dopamine (DA), and 3, 4-dihydroxyphenylacetic acid (DOPAC). A sensitive UPLC-MS/MS method for the simultaneous determination of these endogenous substances in rat plasma and lung tissues was developed. The validated method was successfully applied for comparing profiles of analytes in rat plasma and lung tissues. The results indicated that five endogenous substances, namely, GABA, Ach, Glu, DA, and DOPAC, had a significant change in the rats with PM2.5-induced lung impairment.
Collapse
Affiliation(s)
- Ronghua Fan
- Department of Health Inspection, College of Public Health, Shenyang Medical College, Shenyang, 11034, Liaoning Province, China
| | - Qingquan Ren
- Department of Health Inspection, College of Public Health, Shenyang Medical College, Shenyang, 11034, Liaoning Province, China
| | - Tao Zhou
- Department of Health Inspection, College of Public Health, Shenyang Medical College, Shenyang, 11034, Liaoning Province, China
| | - Lei Shang
- Department of Key Laboratory of Environmental Pollution and Microecology, Shenyang Medical College, Shenyang, 11034, Liaoning Province, China
| | - Mingyue Ma
- Department of Toxicology, College of Public Health, Shenyang Medical College, Shenyang, 11034, Liaoning Province, China
| | - Bolun Wang
- Department of Health Inspection, College of Public Health, Shenyang Medical College, Shenyang, 11034, Liaoning Province, China
| | - Chunling Xiao
- Department of Key Laboratory of Environmental Pollution and Microecology, Shenyang Medical College, Shenyang, 11034, Liaoning Province, China.
| |
Collapse
|
50
|
Tignanelli CJ, Hemmila MR, Rogers MAM, Raghavendran K. Nationwide cohort study of independent risk factors for acute respiratory distress syndrome after trauma. Trauma Surg Acute Care Open 2019; 4:e000249. [PMID: 30899792 PMCID: PMC6407565 DOI: 10.1136/tsaco-2018-000249] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/11/2018] [Accepted: 12/24/2018] [Indexed: 12/14/2022] Open
Abstract
Background The primary objective of this study was to evaluate the effect of specific direct and indirect factors that accounted, in trauma patients, for the development of acute respiratory distress syndrome (ARDS) and mortality in patients with ARDS. Methods We performed a retrospective cohort study of patients from the National Trauma Data Bank. Multilevel mixed-effects logistic regression was used with the development of ARDS as the primary and mortality in patients with ARDS as the secondary outcome measures. We compared trauma patients with versus without thoracic (direct) and extrathoracic (indirect) risk factors, using patient demographics, physiologic, and anatomic injury severity as covariates. Subset analysis was performed for patients with trauma-induced lung contusion (TILC) and for patients with minor (Injury Severity Score [ISS] ≤15) injury. Results A total of 2 998 964 patients were studied, of whom 28 597 developed ARDS. From 2011 to 2014, the incidence of ARDS decreased; however, mortality in patients with ARDS has increased. Predictors of ARDS included direct thoracic injury (TILC, multiple rib fractures, and flail chest), as well as indirect factors (increased age, male gender, higher ISS, lower Glasgow Coma Scale motor component score, history of cardiopulmonary or hematologic disease, and history of alcoholism or obesity). Patients with ARDS secondary to direct thoracic injury had a lower risk of mortality compared with patients with ARDS due to other mechanisms. Discussion Despite the decreasing incidence of trauma-induced ARDS, mortality in patients with ARDS has increased. Direct thoracic injury was the strongest predictor of ARDS. Knowing specific contributors to trauma-induced ARDS could help identify at-risk patients early in their hospitalization and mitigate the progression to ARDS and thereby mortality. Level of evidence Prognostic study, level III.
Collapse
Affiliation(s)
- Christopher J Tignanelli
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA.,Institute for Health Informatics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Mark R Hemmila
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Mary A M Rogers
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|