1
|
Mohammadi A, De Luca D, Gauda EB. Characteristics, triggers, treatments, and experimental models of neonatal acute respiratory distress syndrome. Am J Physiol Lung Cell Mol Physiol 2025; 328:L512-L525. [PMID: 39924963 DOI: 10.1152/ajplung.00312.2024] [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: 09/24/2024] [Revised: 11/20/2024] [Accepted: 02/04/2025] [Indexed: 02/11/2025] Open
Abstract
Neonatal acute respiratory distress syndrome (NARDS) is a severe and potentially life-threatening form of lung injury recently defined by the International Neonatal ARDS Consensus. It is marked by extensive lung inflammation and damage to the alveolar epithelium and vascular endothelium. NARDS can be triggered by direct inflammatory exposures, such as pneumonia and aspiration, and indirect exposures, including sepsis, necrotizing enterocolitis, and chorioamnionitis. This review provides clinicians and researchers with the latest insights on NARDS. We adopt a cross-disciplinary approach to discuss the diagnostic criteria, pathobiology, triggers, epidemiology, and treatments of NARDS. In addition, we summarize existing clinical studies and advanced preclinical models that help address current knowledge gaps. Future research should focus on standardizing the Montreux consensus definition of NARDS in preclinical and clinical studies, identifying biomarkers, developing prediction models, and exploring novel therapies for affected infants.
Collapse
Affiliation(s)
- Atefeh Mohammadi
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Division of Neonatology and Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Daniele De Luca
- Division of Pediatrics and Neonatal Critical Care, "A. Béclère" Medical Center, Paris - Saclay University Hospitals, APHP, Paris, France
- Physiopathology and Therapeutic Innovation Unit-INSERM U999, Paris Saclay University, Paris, France
| | - Estelle B Gauda
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Division of Neonatology and Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Mitri C, Philippart F, Sacco E, Legriel S, Rousselet N, Dupuis G, Colsch B, Corvol H, Touqui L, Tabary O. Multicentric investigations of the role in the disease severity of accelerated phospholipid changes in COVID-19 patient airway. Microbes Infect 2024; 26:105354. [PMID: 38754811 DOI: 10.1016/j.micinf.2024.105354] [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: 05/12/2023] [Revised: 04/30/2024] [Accepted: 05/10/2024] [Indexed: 05/18/2024]
Abstract
CONTEXT The changes in host membrane phospholipids are crucial in airway infection pathogenesis. Phospholipase A2 hydrolyzes host cell membranes, producing lyso-phospholipids and free fatty acids, including arachidonic acid (AA), which contributes significantly to lung inflammation. AIM Follow these changes and their evolution from day 1, day 3 to day 7 in airway aspirates of 89 patients with COVID-19-associated acute respiratory distress syndrome and examine whether they correlate with the severity of the disease. The patients were recruited in three French intensive care units. The analysis was conducted from admission to the intensive care unit until the end of the first week of mechanical ventilation. RESULTS In the airway aspirates, we found significant increases in the levels of host cell phospholipids, including phosphatidyl-serine and phosphatidyl-ethanolamine, and their corresponding lyso-phospholipids. This was accompanied by increased levels of AA and its inflammatory metabolite prostaglandin E2 (PGE2). Additionally, enhanced levels of ceramides, sphingomyelin, and free cholesterol were observed in these aspirates. These lipids are known to be involved in cell death and/or apoptosis, whereas free cholesterol plays a role in virus entry and replication in host cells. However, there were no significant changes in the levels of dipalmitoyl-phosphatidylcholine, the major surfactant phospholipid. A correlation analysis revealed an association between mortality risk and levels of AA and PGE2, as well as host cell phospholipids. CONCLUSION Our findings indicate a correlation between heightened cellular phospholipid modifications and variations in AA and PGE2 with the severity of the disease in patients. Nevertheless, there is no indication of surfactant alteration in the initial phases of the illness.
Collapse
Affiliation(s)
- Christie Mitri
- Sorbonne Université, Inserm U938, Centre de Recherche Saint-Antoine (CRSA), 75012, Paris, France
| | - François Philippart
- Endotoxins, Structures and Host Response, Department of Microbiology, Institute for Integrative Biology of the Cell, UMR 9891 CNRS-CEA-Paris Saclay University, 98190 Gif-sur-Yvette, France; Medical-Surgical Intensive Care Unit, Groupe Hospitalier Paris Saint Joseph, Paris, France
| | - Emmanuelle Sacco
- Department of Clinical Research. Groupe Hospitalier Paris Saint Joseph, Paris, France
| | - Stéphane Legriel
- Medical-Surgical Intensive Care Unit, Centre Hospitalier de Versailles, Le Chesnay, France
| | - Nathalie Rousselet
- Sorbonne Université, Inserm U938, Centre de Recherche Saint-Antoine (CRSA), 75012, Paris, France
| | - Gabrielle Dupuis
- Sorbonne Université, Inserm U938, Centre de Recherche Saint-Antoine (CRSA), 75012, Paris, France
| | - Benoît Colsch
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (MTS), MetaboHUB, F-91191, Gif sur Yvette, France
| | - Harriet Corvol
- Sorbonne Université, Inserm U938, Centre de Recherche Saint-Antoine (CRSA), 75012, Paris, France; Sorbonne Université, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Trousseau, Service de Pneumologie Pédiatrique, 75012, Paris, France
| | - Lhousseine Touqui
- Sorbonne Université, Inserm U938, Centre de Recherche Saint-Antoine (CRSA), 75012, Paris, France; Inserm, Institut Pasteur, Mucoviscidose et Bronchopathies Chroniques, Département Santé Globale, Paris, France.
| | - Olivier Tabary
- Sorbonne Université, Inserm U938, Centre de Recherche Saint-Antoine (CRSA), 75012, Paris, France.
| |
Collapse
|
3
|
Mikolka P, Kosutova P, Kolomaznik M, Nemcova N, Hanusrichterova J, Curstedt T, Johansson J, Calkovska A. The Synthetic Surfactant CHF5633 Restores Lung Function and Lung Architecture in Severe Acute Respiratory Distress Syndrome in Adult Rabbits. Lung 2024; 202:299-315. [PMID: 38684519 PMCID: PMC11143048 DOI: 10.1007/s00408-024-00689-z] [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: 03/01/2024] [Accepted: 03/23/2024] [Indexed: 05/02/2024]
Abstract
PURPOSE Acute respiratory distress syndrome (ARDS) is a major cause of hypoxemic respiratory failure in adults. In ARDS extensive inflammation and leakage of fluid into the alveoli lead to dysregulation of pulmonary surfactant metabolism and function. Altered surfactant synthesis, secretion, and breakdown contribute to the clinical features of decreased lung compliance and alveolar collapse. Lung function in ARDS could potentially be restored with surfactant replacement therapy, and synthetic surfactants with modified peptide analogues may better withstand inactivation in ARDS alveoli than natural surfactants. METHODS This study aimed to investigate the activity in vitro and the bolus effect (200 mg phospholipids/kg) of synthetic surfactant CHF5633 with analogues of SP-B and SP-C, or natural surfactant Poractant alfa (Curosurf®, both preparations Chiesi Farmaceutici S.p.A.) in a severe ARDS model (the ratio of partial pressure arterial oxygen and fraction of inspired oxygen, P/F ratio ≤ 13.3 kPa) induced by hydrochloric acid instillation followed by injurious ventilation in adult New Zealand rabbits. The animals were ventilated for 4 h after surfactant treatment and the respiratory parameters, histological appearance of lung parenchyma and levels of inflammation, oxidative stress, surfactant dysfunction, and endothelial damage were evaluated. RESULTS Both surfactant preparations yielded comparable improvements in lung function parameters, reductions in lung injury score, pro-inflammatory cytokines levels, and lung edema formation compared to untreated controls. CONCLUSIONS This study indicates that surfactant replacement therapy with CHF5633 improves lung function and lung architecture, and attenuates inflammation in severe ARDS in adult rabbits similarly to Poractant alfa. Clinical trials have so far not yielded conclusive results, but exogenous surfactant may be a valid supportive treatment for patients with ARDS given its anti-inflammatory and lung-protective effects.
Collapse
Affiliation(s)
- Pavol Mikolka
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia.
| | - Petra Kosutova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Maros Kolomaznik
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Nikolett Nemcova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Juliana Hanusrichterova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Tore Curstedt
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Jan Johansson
- Department of Biosciences and Nutrition, Karolinska Institutet, Neo, Huddinge, Sweden
| | - Andrea Calkovska
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| |
Collapse
|
4
|
Battaglini D, Iavarone IG, Rocco PRM. An update on the pharmacological management of acute respiratory distress syndrome. Expert Opin Pharmacother 2024; 25:1229-1247. [PMID: 38940703 DOI: 10.1080/14656566.2024.2374461] [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: 05/10/2024] [Accepted: 06/26/2024] [Indexed: 06/29/2024]
Abstract
INTRODUCTION Acute respiratory distress syndrome (ARDS) is characterized by acute inflammatory injury to the lungs, alterations in vascular permeability, loss of aerated tissue, bilateral infiltrates, and refractory hypoxemia. ARDS is considered a heterogeneous syndrome, which complicates the search for effective therapies. The goal of this review is to provide an update on the pharmacological management of ARDS. AREAS COVERED The difficulties in finding effective pharmacological therapies are mainly due to the challenges in designing clinical trials for this unique, varied population of critically ill patients. Recently, some trials have been retrospectively analyzed by dividing patients into hyper-inflammatory and hypo-inflammatory sub-phenotypes. This approach has led to significant outcome improvements with some pharmacological treatments that previously failed to demonstrate efficacy, which suggests that a more precise selection of ARDS patients for clinical trials could be the key to identifying effective pharmacotherapies. This review is provided after searching the main studies on this topics on the PubMed and clinicaltrials.gov databases. EXPERT OPINION The future of ARDS therapy lies in precision medicine, innovative approaches to drug delivery, immunomodulation, cell-based therapies, and robust clinical trial designs. These should lead to more effective and personalized treatments for patients with ARDS.
Collapse
Affiliation(s)
- Denise Battaglini
- Anesthesia and Intensive Care, IRCCS Ospedale Policlinico, Genova, Italy
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Genova, Italy
| | - Ida Giorgia Iavarone
- Anesthesia and Intensive Care, IRCCS Ospedale Policlinico, Genova, Italy
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Genova, Italy
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
5
|
Dushianthan A, Grocott MPW, Murugan GS, Wilkinson TMA, Postle AD. Pulmonary Surfactant in Adult ARDS: Current Perspectives and Future Directions. Diagnostics (Basel) 2023; 13:2964. [PMID: 37761330 PMCID: PMC10528901 DOI: 10.3390/diagnostics13182964] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/10/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a major cause of hypoxemic respiratory failure in adults, leading to the requirement for mechanical ventilation and poorer outcomes. Dysregulated surfactant metabolism and function are characteristic of ARDS. A combination of alveolar epithelial damage leading to altered surfactant synthesis, secretion, and breakdown with increased functional inhibition from overt alveolar inflammation contributes to the clinical features of poor alveolar compliance and alveolar collapse. Quantitative and qualitative alterations in the bronchoalveolar lavage and tracheal aspirate surfactant composition contribute to ARDS pathogenesis. Compared to neonatal respiratory distress syndrome (nRDS), replacement studies of exogenous surfactants in adult ARDS suggest no survival benefit. However, these studies are limited by disease heterogeneity, variations in surfactant preparations, doses, and delivery methods. More importantly, the lack of mechanistic understanding of the exact reasons for dysregulated surfactant remains a significant issue. Moreover, studies suggest an extremely short half-life of replaced surfactant, implying increased catabolism. Refining surfactant preparations and delivery methods with additional co-interventions to counteract surfactant inhibition and degradation has the potential to enhance the biophysical characteristics of surfactant in vivo.
Collapse
Affiliation(s)
- Ahilanandan Dushianthan
- National Institute for Health Research (NIHR) Southampton Biomedical Research Centre, University Hospital Southampton National Health System Foundation Trust, Southampton SO16 6YD, UK; (M.P.W.G.); (T.M.A.W.); (A.D.P.)
- Integrative Physiology and Critical Illness Group, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Michael P. W. Grocott
- National Institute for Health Research (NIHR) Southampton Biomedical Research Centre, University Hospital Southampton National Health System Foundation Trust, Southampton SO16 6YD, UK; (M.P.W.G.); (T.M.A.W.); (A.D.P.)
- Integrative Physiology and Critical Illness Group, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | | | - Tom M. A. Wilkinson
- National Institute for Health Research (NIHR) Southampton Biomedical Research Centre, University Hospital Southampton National Health System Foundation Trust, Southampton SO16 6YD, UK; (M.P.W.G.); (T.M.A.W.); (A.D.P.)
- Integrative Physiology and Critical Illness Group, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Anthony D. Postle
- National Institute for Health Research (NIHR) Southampton Biomedical Research Centre, University Hospital Southampton National Health System Foundation Trust, Southampton SO16 6YD, UK; (M.P.W.G.); (T.M.A.W.); (A.D.P.)
- Integrative Physiology and Critical Illness Group, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
6
|
Ngo D, Pratte KA, Flexeder C, Petersen H, Dang H, Ma Y, Keyes MJ, Gao Y, Deng S, Peterson BD, Farrell LA, Bhambhani VM, Palacios C, Quadir J, Gillenwater L, Xu H, Emson C, Gieger C, Suhre K, Graumann J, Jain D, Conomos MP, Tracy RP, Guo X, Liu Y, Johnson WC, Cornell E, Durda P, Taylor KD, Papanicolaou GJ, Rich SS, Rotter JI, Rennard SI, Curtis JL, Woodruff PG, Comellas AP, Silverman EK, Crapo JD, Larson MG, Vasan RS, Wang TJ, Correa A, Sims M, Wilson JG, Gerszten RE, O’Connor GT, Barr RG, Couper D, Dupuis J, Manichaikul A, O’Neal WK, Tesfaigzi Y, Schulz H, Bowler RP. Systemic Markers of Lung Function and Forced Expiratory Volume in 1 Second Decline across Diverse Cohorts. Ann Am Thorac Soc 2023; 20:1124-1135. [PMID: 37351609 PMCID: PMC10405603 DOI: 10.1513/annalsats.202210-857oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 06/13/2023] [Indexed: 06/24/2023] Open
Abstract
Rationale: Chronic obstructive pulmonary disease (COPD) is a complex disease characterized by airway obstruction and accelerated lung function decline. Our understanding of systemic protein biomarkers associated with COPD remains incomplete. Objectives: To determine what proteins and pathways are associated with impaired pulmonary function in a diverse population. Methods: We studied 6,722 participants across six cohort studies with both aptamer-based proteomic and spirometry data (4,566 predominantly White participants in a discovery analysis and 2,156 African American cohort participants in a validation). In linear regression models, we examined protein associations with baseline forced expiratory volume in 1 second (FEV1) and FEV1/forced vital capacity (FVC). In linear mixed effects models, we investigated the associations of baseline protein levels with rate of FEV1 decline (ml/yr) in 2,777 participants with up to 7 years of follow-up spirometry. Results: We identified 254 proteins associated with FEV1 in our discovery analyses, with 80 proteins validated in the Jackson Heart Study. Novel validated protein associations include kallistatin serine protease inhibitor, growth differentiation factor 2, and tumor necrosis factor-like weak inducer of apoptosis (discovery β = 0.0561, Q = 4.05 × 10-10; β = 0.0421, Q = 1.12 × 10-3; and β = 0.0358, Q = 1.67 × 10-3, respectively). In longitudinal analyses within cohorts with follow-up spirometry, we identified 15 proteins associated with FEV1 decline (Q < 0.05), including elafin leukocyte elastase inhibitor and mucin-associated TFF2 (trefoil factor 2; β = -4.3 ml/yr, Q = 0.049; β = -6.1 ml/yr, Q = 0.032, respectively). Pathways and processes highlighted by our study include aberrant extracellular matrix remodeling, enhanced innate immune response, dysregulation of angiogenesis, and coagulation. Conclusions: In this study, we identify and validate novel biomarkers and pathways associated with lung function traits in a racially diverse population. In addition, we identify novel protein markers associated with FEV1 decline. Several protein findings are supported by previously reported genetic signals, highlighting the plausibility of certain biologic pathways. These novel proteins might represent markers for risk stratification, as well as novel molecular targets for treatment of COPD.
Collapse
Affiliation(s)
- Debby Ngo
- Cardiovascular Research Institute
- Division of Pulmonary, Critical Care, and Sleep Medicine, and
| | | | - Claudia Flexeder
- Institute of Epidemiology and
- Comprehensive Pneumology Center Munich (CPC-M) as member of the German Center for Lung Research (DZL), Munich, Germany
- Institute and Clinic for Occupational, Social, and Environmental Medicine, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Hans Petersen
- Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Hong Dang
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Yanlin Ma
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia
| | | | - Yan Gao
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi; and
- Institute and Clinic for Occupational, Social, and Environmental Medicine, University Hospital, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | | | | - Hanfei Xu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Claire Emson
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland
| | - Christian Gieger
- Institute of Epidemiology and
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München – German Research Center for Environmental Health, Neuherberg, Germany
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medicine Qatar, Education City, Doha, Qatar
| | | | - Deepti Jain
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Matthew P. Conomos
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Russell P. Tracy
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA (University of California, Los Angeles) Medical Center, Torrance, California
| | - Yongmei Liu
- Division of Cardiology, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina
| | - W. Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Elaine Cornell
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Peter Durda
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA (University of California, Los Angeles) Medical Center, Torrance, California
| | - George J. Papanicolaou
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA (University of California, Los Angeles) Medical Center, Torrance, California
| | - Steven I. Rennard
- Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California
| | | | - Prescott G. Woodruff
- Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California
| | | | | | | | - Martin G. Larson
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
- The National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts
| | - Ramachandran S. Vasan
- The National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts
- Division of Preventive Medicine and
- Division of Cardiology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Thomas J. Wang
- Department of Medicine, UT (University of Texas) Southwestern Medical Center, Dallas, Texas
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Adolfo Correa
- Jackson Heart Study, Department of Medicine, and
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi; and
| | - Mario Sims
- Jackson Heart Study, Department of Medicine, and
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi; and
| | - James G. Wilson
- Cardiovascular Research Institute
- Jackson Heart Study, Department of Medicine, and
| | - Robert E. Gerszten
- Cardiovascular Research Institute
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - George T. O’Connor
- The National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts
- Pulmonary Center, Department of Medicine, Boston University, Boston, Massachusetts
| | - R. Graham Barr
- Department of Medicine and
- Department of Epidemiology, Columbia University, New York, New York
| | - David Couper
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia
| | - Wanda K. O’Neal
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Yohannes Tesfaigzi
- Lovelace Respiratory Research Institute, Albuquerque, New Mexico
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Holger Schulz
- Institute of Epidemiology and
- Comprehensive Pneumology Center Munich (CPC-M) as member of the German Center for Lung Research (DZL), Munich, Germany
| | | |
Collapse
|
7
|
Chang Y, Yoo HJ, Kim SJ, Lee K, Lim CM, Hong SB, Koh Y, Huh JW. A targeted metabolomics approach for sepsis-induced ARDS and its subphenotypes. Crit Care 2023; 27:263. [PMID: 37408042 DOI: 10.1186/s13054-023-04552-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is etiologically and clinically a heterogeneous disease. Its diagnostic characteristics and subtype classification, and the application of these features to treatment, have been of considerable interest. Metabolomics is becoming important for identifying ARDS biology and distinguishing its subtypes. This study aimed to identify metabolites that could distinguish sepsis-induced ARDS patients from non-ARDS controls, using a targeted metabolomics approach, and to identify whether sepsis-induced direct and sepsis-induced indirect ARDS are metabolically distinct groups, and if so, confirm their metabolites and associated pathways. METHODS This study retrospectively analyzed 54 samples of ARDS patients from a sepsis registry that was prospectively collected from March 2011 to February 2018, along with 30 non-ARDS controls. The cohort was divided into direct and indirect ARDS. Metabolite concentrations of five analyte classes (energy metabolism, free fatty acids, amino acids, phospholipids, sphingolipids) were measured using liquid chromatography-tandem mass spectrometry and gas chromatography-mass spectrometry by targeted metabolomics. RESULTS In total, 186 metabolites were detected. Among them, 102 metabolites could differentiate sepsis-induced ARDS patients from the non-ARDS controls, while 14 metabolites could discriminate sepsis-induced ARDS subphenotypes. Using partial least-squares discriminant analysis, we showed that sepsis-induced ARDS patients were metabolically distinct from the non-ARDS controls. The main distinguishing metabolites were lysophosphatidylethanolamine (lysoPE) plasmalogen, PE plasmalogens, and phosphatidylcholines (PCs). Sepsis-induced direct and indirect ARDS were also metabolically distinct subgroups, with differences in lysoPCs. Glycerophospholipid and sphingolipid metabolism were the most significant metabolic pathways involved in sepsis-induced ARDS biology and in sepsis-induced direct/indirect ARDS, respectively. CONCLUSION Our study demonstrated a marked difference in metabolic patterns between sepsis-induced ARDS patients and non-ARDS controls, and between sepsis-induced direct and indirect ARDS subpheonotypes. The identified metabolites and pathways can provide clues relevant to the diagnosis and treatment of individuals with ARDS.
Collapse
Affiliation(s)
- Youjin Chang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Inje University Sanggye Paik Hospital, Seoul, Republic of Korea
| | - Hyun Ju Yoo
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Su Jung Kim
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kwangha Lee
- Department of Internal Medicine, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Chae-Man Lim
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Sang-Bum Hong
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Younsuck Koh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea
| | - Jin Won Huh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
8
|
Wu Y, Pernet E, Touqui L. Modulation of Airway Expression of the Host Bactericidal Enzyme, sPLA2-IIA, by Bacterial Toxins. Toxins (Basel) 2023; 15:440. [PMID: 37505708 PMCID: PMC10467128 DOI: 10.3390/toxins15070440] [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: 04/13/2023] [Revised: 06/23/2023] [Accepted: 06/29/2023] [Indexed: 07/29/2023] Open
Abstract
Host molecules with antimicrobial properties belong to a large family of mediators including type-IIA secreted phospholipase A2 (sPLA2-IIA). The latter is a potent bactericidal agent with high selectivity against Gram-positive bacteria, but it may also play a role in modulating the host inflammatory response. However, several pathogen-associated molecular patterns (PAMPs) or toxins produced by pathogenic bacteria can modulate the levels of sPLA2-IIA by either inducing or inhibiting its expression in host cells. Thus, the final sPLA2-IIA concentration during the infection process is determined by the orchestration between the levels of toxins that stimulate and those that downregulate the expression of this enzyme. The stimulation of sPLA2-IIA expression is a process that participates in the clearance of invading bacteria, while inhibition of this expression highlights a mechanism by which certain bacteria can subvert the immune response and invade the host. Here, we will review the major functions of sPLA2-IIA in the airways and the role of bacterial toxins in modulating the expression of this enzyme. We will also summarize the major mechanisms involved in this modulation and the potential consequences for the pulmonary host response to bacterial infection.
Collapse
Affiliation(s)
- Yongzheng Wu
- Unité de Biologie Cellulaire de l’Infection Microbionne, CNRS UMR3691, Institut Pasteur, Université de Paris Cité, 75015 Paris, France;
| | - Erwan Pernet
- Groupe de Recherche en Signalisation Cellulaire, Département de Biologie Médicale, Université du Québec à Trois-Rivières, Trois-Rivières, QC G8Z 4M3, Canada
| | - Lhousseine Touqui
- Sorbonne Université, Inserm U938, Centre de Recherche Saint-Antoine (CRSA), 75012 Paris, France
- Institut Pasteur, Université de Paris Cité, Mucoviscidose et Bronchopathies Chroniques, 75015 Paris, France
| |
Collapse
|
9
|
MIKOLKA P, KOSUTOVA P, KOLOMAZNIK M, MATEFFY S, NEMCOVA N, MOKRA D, CALKOVSKA A. Efficacy of surfactant therapy of ARDS induced by hydrochloric acid aspiration followed by ventilator-induced lung injury - an animal study. Physiol Res 2022; 71:S237-S249. [PMID: 36647912 PMCID: PMC9906666 DOI: 10.33549/physiolres.935003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The development of acute respiratory distress syndrome (ARDS) is known to be independently attributable to aspiration-induced lung injury. Mechanical ventilation as a high pressure/volume support to maintain sufficient oxygenation of a patient could initiate ventilator-induced lung injury (VILI) and thus contribute to lung damage. Although these phenomena are rare in the clinic, they could serve as the severe experimental model of alveolar-capillary membrane deterioration. Lung collapse, diffuse inflammation, alveolar epithelial and endothelial damage, leakage of fluid into the alveoli, and subsequent inactivation of pulmonary surfactant, leading to respiratory failure. Therefore, exogenous surfactant could be considered as a therapy to restore lung function in experimental ARDS. This study aimed to investigate the effect of modified porcine surfactant in animal model of severe ARDS (P/F ratio </=13.3 kPa) induced by intratracheal instillation of hydrochloric acid (HCl, 3 ml/kg, pH 1.25) followed by VILI (V(T) 20 ml/kg). Adult rabbits were divided into three groups: untreated ARDS, model treated with a bolus of poractant alfa (Curosurf®, 2.5 ml/kg, 80 mg phospholipids/ml), and healthy ventilated animals (saline), which were oxygen-ventilated for an additional 4 h. The lung function parameters, histological appearance, degree of lung edema and levels of inflammatory and oxidative markers in plasma were evaluated. Whereas surfactant therapy with poractant alfa improved lung function, attenuated inflammation and lung edema, and partially regenerated significant changes in lung architecture compared to untreated controls. This study indicates a potential of exogenous surfactant preparation in the treatment of experimental ARDS.
Collapse
Affiliation(s)
- Pavol MIKOLKA
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic
| | - Petra KOSUTOVA
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic
| | - Maros KOLOMAZNIK
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic
| | - Stanislav MATEFFY
- Diagnostic Center of Pathology in Prešov, Unilabs Slovakia, Martin, Slovak Republic
| | - Nikolett NEMCOVA
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic
| | - Daniela MOKRA
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic
| | - Andrea CALKOVSKA
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovak Republic
| |
Collapse
|
10
|
Cao C, Zhang L, Shen J. Phosgene-Induced acute lung injury: Approaches for mechanism-based treatment strategies. Front Immunol 2022; 13:917395. [PMID: 35983054 PMCID: PMC9378823 DOI: 10.3389/fimmu.2022.917395] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Phosgene (COCl2) gas is a chemical intermediate of high-volume production with numerous industrial applications worldwide. Due to its high toxicity, accidental exposure to phosgene leads to various chemical injuries, primarily resulting in chemical-induced lung injury due to inhalation. Initially, the illness is mild and presents as coughing, chest tightness, and wheezing; however, within a few hours, symptoms progress to chronic respiratory depression, refractory pulmonary edema, dyspnea, and hypoxemia, which may contribute to acute respiratory distress syndrome or even death in severe cases. Despite rapid advances in medicine, effective treatments for phosgene-inhaled poisoning are lacking. Elucidating the pathophysiology and pathogenesis of acute inhalation toxicity caused by phosgene is necessary for the development of appropriate therapeutics. In this review, we discuss extant literature on relevant mechanisms and therapeutic strategies to highlight novel ideas for the treatment of phosgene-induced acute lung injury.
Collapse
Affiliation(s)
- Chao Cao
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, China
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, China
- Training Center of Acute Poisoning Treatment Technology of Fudan University Shanghai Medical College, Shanghai, China
| | - Lin Zhang
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, China
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, China
| | - Jie Shen
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, China
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, China
- Training Center of Acute Poisoning Treatment Technology of Fudan University Shanghai Medical College, Shanghai, China
| |
Collapse
|
11
|
De Luca D, Alonso A, Autilio C. Bile acids-induced lung injury: update of reverse translational biology. Am J Physiol Lung Cell Mol Physiol 2022; 323:L93-L106. [DOI: 10.1152/ajplung.00523.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The presence of bile acids in lung tissue is associated with some clinical features observed in various medical specialties, but it took time to understand that these are due to a "bile acid-induced lung injury" since specific translational studies and cross-disciplinary awareness were lacking. We used a reverse translational approach to update and summarize the current knowledge about the mechanisms of bile acid-induced lung injury. This has been done in a cross-disciplinary fashion since these conditions may occur in patients of various age and in different medical fields. We here define these clinical conditions, then we review the physiopathology of these conditions and the animal models used to mimic them and, finally, their pathobiology. Mechanisms of bile acid-induced lung injury have been partially clarified overtime and are represented by: 1) the interaction with secretory phospholipase A2 pathway, 2) the effect on surfactant function and structure, 3) the biological effects on inflammation and local immunity, 4) the direct cellular toxicity. These mechanisms are schematically illustrated and histological comparisons between ARDS induced by bile acids and other triggers are also provided. Based on these mechanisms we propose possible direct therapeutic applications and, finally, we discuss further research steps to improve the understanding of processes that generate pathological clinical conditions.
Collapse
Affiliation(s)
- Daniele De Luca
- Division of Pediatrics and Neonatal Critical Care, Paris Saclay University Hospital, Clamart, Paris, France
- Physiopathology and Therapeutic Innovation Unit-INSERM U999, Paris Saclay University, Le Plessis Robinson, France
| | - Alejandro Alonso
- Department of Biochemistry and Molecular Biology, Faculty of Biology, and Research, Institut-Hospital, Complutense University, Madrid, Spain
| | - Chiara Autilio
- Department of Biochemistry and Molecular Biology, Faculty of Biology, and Research, Institut-Hospital, Complutense University, Madrid, Spain
| |
Collapse
|
12
|
Abstract
The secretory phospholipase A2 (sPLA2) group of secreted enzymes hydrolyze phospholipids and lead to the production of multiple biologically active lipid mediators. sPLA2s and their products (e.g., eicosanoids) play a significant role in the pathophysiology of various inflammatory diseases, including life-threatening lung disorders such as acute lung injury (ALI) and the Acute Respiratory Distress Syndrome (ARDS). The ALI/ARDS spectrum of severe inflammatory conditions is caused by direct (such as bacterial or viral pneumonia) or indirect insults (sepsis) that are associated with high morbidity and mortality. Several sPLA2 isoforms are upregulated in patients with ARDS as well as in multiple ALI preclinical models, and individual sPLA2s exert unique roles in regulating ALI pathophysiology. This brief review will summarize the contributions of specific sPLA2 isoforms as markers and mediators in ALI, supporting a potential therapeutic role for targeting them in ARDS.
Collapse
Affiliation(s)
- Eleftheria Letsiou
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, USA
| | - Yu Maw Htwe
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, USA
| | - Steven M Dudek
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
13
|
Kitsiouli E, Tenopoulou M, Papadopoulos S, Lekka ME. Phospholipases A2 as biomarkers in ARDS. Biomed J 2021; 44:663-670. [PMID: 34478892 PMCID: PMC8847824 DOI: 10.1016/j.bj.2021.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/16/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a multifactorial life-threatening lung injury, characterized by diffuse lung inflammation and increased alveolocapillary barrier permeability. The different stages of ARDS have distinctive biochemical and clinical profiles. Despite the progress of our understanding on ARDS pathobiology, the mechanisms underlying its pathogenesis are still obscure. Herein, we review the existing literature about the implications of phospholipases 2 (PLA2s), a large family of enzymes that catalyze the hydrolysis of fatty acids at the sn-2 position of glycerophospholipids, in ARDS-related pathology. We emphasize on the versatile way of participation of different PLA2s isoforms in the distinct ARDS subgroup phenotypes by either potentiating lung inflammation and damage or by preserving the normal lung. Current research supports that PLA2s are associated with the progression and the outcome of ARDS. We herein discuss the transcellular communication of PLA2s through secreted extracellular vesicles and suggest it as a new mechanism of PLA2s involvement in ARDS. Thus, the elucidation of the spatiotemporal features of PLA2s expression may give new insights and provide valuable information about the risk of an individual to develop ARDS or advance to more severe stages, and potentially identify PLA2 isoforms as biomarkers and target for pharmacological intervention.
Collapse
Affiliation(s)
- Eirini Kitsiouli
- Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Margarita Tenopoulou
- Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Stylianos Papadopoulos
- Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Marilena E Lekka
- Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece.
| |
Collapse
|
14
|
Pioselli B, Salomone F, Mazzola G, Amidani D, Sgarbi E, Amadei F, Murgia X, Catinella S, Villetti G, De Luca D, Carnielli V, Civelli M. Pulmonary surfactant: a unique biomaterial with life-saving therapeutic applications. Curr Med Chem 2021; 29:526-590. [PMID: 34525915 DOI: 10.2174/0929867328666210825110421] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/26/2021] [Accepted: 06/29/2021] [Indexed: 11/22/2022]
Abstract
Pulmonary surfactant is a complex lipoprotein mixture secreted into the alveolar lumen by type 2 pneumocytes, which is composed by tens of different lipids (approximately 90% of its entire mass) and surfactant proteins (approximately 10% of the mass). It is crucially involved in maintaining lung homeostasis by reducing the values of alveolar liquid surface tension close to zero at end-expiration, thereby avoiding the alveolar collapse, and assembling a chemical and physical barrier against inhaled pathogens. A deficient amount of surfactant or its functional inactivation is directly linked to a wide range of lung pathologies, including the neonatal respiratory distress syndrome. This paper reviews the main biophysical concepts of surfactant activity and its inactivation mechanisms, and describes the past, present and future roles of surfactant replacement therapy, focusing on the exogenous surfactant preparations marketed worldwide and new formulations under development. The closing section describes the pulmonary surfactant in the context of drug delivery. Thanks to its peculiar composition, biocompatibility, and alveolar spreading capability, the surfactant may work not only as a shuttle to the branched anatomy of the lung for other drugs but also as a modulator for their release, opening to innovative therapeutic avenues for the treatment of several respiratory diseases.
Collapse
Affiliation(s)
| | | | | | | | - Elisa Sgarbi
- Preclinical R&D, Chiesi Farmaceutici, Parma. Italy
| | | | - Xabi Murgia
- Department of Biotechnology, GAIKER Technology Centre, Zamudio. Spain
| | | | | | - Daniele De Luca
- Division of Pediatrics and Neonatal Critical Care, Antoine Béclère Medical Center, APHP, South Paris University Hospitals, Paris, France; Physiopathology and Therapeutic Innovation Unit-U999, South Paris-Saclay University, Paris. France
| | - Virgilio Carnielli
- Division of Neonatology, G Salesi Women and Children's Hospital, Polytechnical University of Marche, Ancona. Italy
| | | |
Collapse
|
15
|
De Luca D, Autilio C. Strategies to protect surfactant and enhance its activity. Biomed J 2021; 44:654-662. [PMID: 34365021 PMCID: PMC8847817 DOI: 10.1016/j.bj.2021.07.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/21/2021] [Accepted: 07/29/2021] [Indexed: 11/22/2022] Open
Abstract
The knowledge about surfactant biology is now deeper and recent research has allowed to clarify its role in several human lung disorders. The balance between surfactant production and consumption is better known and the same applies to their regulatory mechanisms. This has allowed to hypothesize and investigate several new and original strategies to protect surfactant and enhance its activity. These interventions are potentially useful for several disorders and particularly for acute respiratory distress syndrome. We here highlight the mechanisms regulating surfactant consumption, encompassing surfactant catabolism but also surfactant injury due to other mechanisms, in a physiopathology-driven fashion. We then analyze each corresponding strategy to protect surfactant and enhance its activity. Some of these strategies are more advanced in terms of research & development pathway, some others are still investigational, but all are promising and deserve a joint effort from clinical-academic researchers and the industry.
Collapse
Affiliation(s)
- Daniele De Luca
- Division of Paediatrics and Neonatal Critical Care, "A.Béclère" Medical Centre, Paris Saclay University Hospitals, APHP, Paris, France; Physiopathology and Therapeutic Innovation Unit-INSERM U999, Paris Saclay University, Paris, France.
| | - Chiara Autilio
- Dpt. of Biochemistry and Molecular Biology and Research Institute "Hospital 12 de Octubre", Complutense University, Madrid, Spain
| |
Collapse
|
16
|
De Luca D, Touqui L. The International Week of Surfactant Research: increasing knowledge about surfactant and unexploited opportunities. Biomed J 2021; 44:651-653. [PMID: 34314899 PMCID: PMC8847801 DOI: 10.1016/j.bj.2021.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/09/2021] [Accepted: 07/19/2021] [Indexed: 11/26/2022] Open
Affiliation(s)
- Daniele De Luca
- Division of Pediatrics and Neonatal Critical Care, "A.Béclère" Medical Centre, Paris Saclay University Hospitals, APHP Paris - France; Physiopathology and Therapeutic Innovation Unit-INSERM U999, Paris Saclay University Paris - France.
| | - Lhoussaine Touqui
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint Antoine, Paris - France; Mucoviscidose and Bronchopathies Chroniques", Pasteur Institute Paris - France
| |
Collapse
|
17
|
Kuypers FA, Rostad CA, Anderson EJ, Chahroudi A, Jaggi P, Wrammert J, Mantus G, Basu R, Harris F, Hanberry B, Camacho-Gonzalez A, Manoranjithan S, Vos M, Brown LA, Morris CR. Secretory phospholipase A2 in SARS-CoV-2 infection and multisystem inflammatory syndrome in children (MIS-C). Exp Biol Med (Maywood) 2021; 246:2543-2552. [PMID: 34255566 PMCID: PMC8649422 DOI: 10.1177/15353702211028560] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Secretory phospholipase 2 (sPLA2) acts as a mediator between proximal and distal events of the inflammatory cascade. Its role in SARS-CoV-2 infection is unknown, but could contribute to COVID-19 inflammasome activation and cellular damage. We present the first report of plasma sPLA2 levels in adults and children with COVID-19 compared with controls. Currently asymptomatic adults with a history of recent COVID-19 infection (≥4 weeks before) identified by SARS-CoV-2 IgG antibodies had sPLA2 levels similar to those who were seronegative (9 ± 6 vs.17 ± 28 ng/mL, P = 0.26). In contrast, children hospitalized with severe COVID-19 had significantly elevated sPLA2 compared with those with mild or asymptomatic SARS-CoV-2 infection (269 ± 137 vs. 2 ± 3 ng/mL, P = 0.01). Among children hospitalized with multisystem inflammatory syndrome in children (MIS-C), all had severe disease requiring pediatric intensive care unit (PICU) admission. sPLA2 levels were significantly higher in those with acute illness <10 days versus convalescent disease ≥10 days (540 ± 510 vs. 2 ± 1, P = 0.04). Thus, sPLA2 levels correlated with COVID-19 severity and acute MIS-C in children, implicating a role in inflammasome activation and disease pathogenesis. sPLA2 may be a useful biomarker to stratify risk and guide patient management for children with acute COVID-19 and MIS-C. Therapeutic compounds targeting sPLA2 and inflammasome activation warrant consideration.
Collapse
Affiliation(s)
- Frans A Kuypers
- Division of Hematology, Department of Pediatrics, University of California, San Francisco, CA 94609, USA
| | - Christina A Rostad
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.,Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA
| | - Evan J Anderson
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.,Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA.,Department of Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Ann Chahroudi
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.,Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA
| | - Preeti Jaggi
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Jens Wrammert
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA
| | - Grace Mantus
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Rajit Basu
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Frank Harris
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Bradley Hanberry
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Andres Camacho-Gonzalez
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.,Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA
| | | | - Miriam Vos
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.,Center for Clinical and Translational Research, Children's Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA
| | - Lou Ann Brown
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Claudia R Morris
- Department of Pediatrics1371, School of Medicine, Emory University, Atlanta, GA 30322, USA.,Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.,Center for Clinical and Translational Research, Children's Healthcare of Atlanta and Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
18
|
Da Silva E, Hickey C, Ellis G, Hougaard K, Sørli J. In vitro prediction of clinical signs of respiratory toxicity in rats following inhalation exposure. Curr Res Toxicol 2021; 2:204-209. [PMID: 34345862 PMCID: PMC8320621 DOI: 10.1016/j.crtox.2021.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/26/2021] [Accepted: 05/12/2021] [Indexed: 11/30/2022] Open
Abstract
To date there are no OECD validated alternative approaches to study toxicity following inhalation exposure to airborne chemicals. The available OECD test guidelines for acute inhalation toxicity aim to estimate a value of the lethal air concentration of the test chemical leading to the death of 50% of the exposed animals (LC50), to satisfy hazard classification and labelling requirements. This paper explores the view that alternative approaches must compare to outcomes of existing guideline methods to become accepted and implemented in a regulatory context. This case study describes the initiatives taken to validate the lung surfactant bioassay, an in vitro cell-free method, and discusses the challenges faced. While the lung surfactant bioassay could not predict the GHS classification for acute inhalation toxicity of 26 chemicals, the assay successfully predicted the clinical signs of respiratory toxicity observed during or shortly after exposure in vivo as reported in registration dossiers. The lung surfactant bioassay is a promising alternative approach to assess the potential of chemicals to cause changes to respiration remaining after exposure (indicating decreased lung function), and can be combined with other test methods in an integrated approach to testing and assessment of inhaled substances.
Collapse
Affiliation(s)
- E. Da Silva
- Technical University of Denmark, Kgs. Lyngby, Denmark
- National Research Centre for the Working Environment, Copenhagen, Denmark
| | - C. Hickey
- Firmenich Incorporated, United States
| | | | - K.S. Hougaard
- National Research Centre for the Working Environment, Copenhagen, Denmark
- University of Copenhagen, Copenhagen, Denmark
| | - J.B. Sørli
- National Research Centre for the Working Environment, Copenhagen, Denmark
| |
Collapse
|
19
|
De Luca D, Cogo P, Kneyber MC, Biban P, Semple MG, Perez-Gil J, Conti G, Tissieres P, Rimensberger PC. Surfactant therapies for pediatric and neonatal ARDS: ESPNIC expert consensus opinion for future research steps. Crit Care 2021; 25:75. [PMID: 33618742 PMCID: PMC7898495 DOI: 10.1186/s13054-021-03489-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 02/04/2021] [Indexed: 12/14/2022] Open
Abstract
Pediatric (PARDS) and neonatal (NARDS) acute respiratory distress syndrome have different age-specific characteristics and definitions. Trials on surfactant for ARDS in children and neonates have been performed well before the PARDS and NARDS definitions and yielded conflicting results. This is mainly due to heterogeneity in study design reflecting historic lack of pathobiology knowledge. We reviewed the available clinical and preclinical data to create an expert consensus aiming to inform future research steps and advance the knowledge in this area. Eight trials investigated the use of surfactant for ARDS in children and ten in neonates, respectively. There were improvements in oxygenation (7/8 trials in children, 7/10 in neonates) and mortality (3/8 trials in children, 1/10 in neonates) improved. Trials were heterogeneous for patients' characteristics, surfactant type and administration strategy. Key pathobiological concepts were missed in study design. Consensus with strong agreement was reached on four statements: 1. There are sufficient preclinical and clinical data to support targeted research on surfactant therapies for PARDS and NARDS. Studies should be performed according to the currently available definitions and considering recent pathobiology knowledge. 2. PARDS and NARDS should be considered as syndromes and should be pre-clinically studied according to key characteristics, such as direct or indirect (primary or secondary) nature, clinical severity, infectious or non-infectious origin or patients' age. 3. Explanatory should be preferred over pragmatic design for future trials on PARDS and NARDS. 4. Different clinical outcomes need to be chosen for PARDS and NARDS, according to the trial phase and design, trigger type, severity class and/or surfactant treatment policy. We advocate for further well-designed preclinical and clinical studies to investigate the use of surfactant for PARDS and NARDS following these principles.
Collapse
Affiliation(s)
- Daniele De Luca
- Division of Pediatrics and Neonatal Critical Care, "A.Béclère" Medical Centre, Paris Saclay University Hospitals, APHP, 157 Rue de la Porte de Trivaux, 92140, Clamart (Paris-IDF), France.
- Physiopathology and Therapeutic Innovation Unit-INSERM U999, Paris Saclay University, Paris, France.
| | - Paola Cogo
- Department of Pediatrics, University of Udine, Udine, Italy
| | - Martin C Kneyber
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Beatrix Children's Hospital Groningen, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
- Critical Care, Anesthesiology, Peri-Operative and Emergency Medicine (CAPE), University of Groningen, Groningen, The Netherlands
| | - Paolo Biban
- Department of Neonatal and Pediatric Critical Care, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Malcolm Grace Semple
- Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK
| | - Jesus Perez-Gil
- Department of Biochemistry and Molecular Biology and Research Institute "Hospital 12 de Octubre", Complutense University, Madrid, Spain
| | - Giorgio Conti
- Department of Anesthesiology and Intensive Care, Catholic University of the Sacred Heart, Rome, Italy
| | - Pierre Tissieres
- Division of Pediatric Critical Care and Neonatal Medicine, "Kremlin-Bicetre" Medical Center, Paris Saclay University Hospitals, APHP, Paris, France
- Integrative Cellular Biology Institute-UMR 9198, Host-Pathogen Interactions Team, Paris Saclay University, Paris, France
| | - Peter C Rimensberger
- Division of Neonatology and Pediatric Critical Care, Department of Pediatrics, University Hospital of Geneva, University of Geneva, Geneva, Switzerland
| |
Collapse
|
20
|
De Luca D. Respiratory distress syndrome in preterm neonates in the era of precision medicine: A modern critical care-based approach. Pediatr Neonatol 2021; 62 Suppl 1:S3-S9. [PMID: 33358440 DOI: 10.1016/j.pedneo.2020.11.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023] Open
Abstract
Respiratory distress syndrome (RDS) was recognized to be caused by primary surfactant deficiency almost 70 years ago and continuous positive airway pressure was introduced approximately 50 years ago. Since then, there have been many developments in neonatology; we know many things but others are still controversial. The more we know, the more questions arise. However, this review aims to indicate what is more needed to understand and how should be the modern approach to RDS in the era of precision medicine. The review is divided between new concepts and new tools. We will explain the interaction between steroids, CPAP and surfactant, as well as the surfactant catabolism and the diagnosis of NARDS; lung ultrasound and new tools to optimize CPAP will also be covered. How these concepts are integrated in the author's personal experience is also illustrated.
Collapse
Affiliation(s)
- Daniele De Luca
- Division of Pediatrics and Neonatal Critical Care, "Antoine Béclère" Hospital, Paris Saclay University Hospitals, APHP, Paris, France; Physiopathology and Therapeutic Innovation Unit-INSERM U999, Paris Saclay University, Paris, France.
| |
Collapse
|
21
|
Mikolka P, Curstedt T, Feinstein R, Larsson A, Grendar M, Rising A, Johansson J. Impact of synthetic surfactant CHF5633 with SP-B and SP-C analogues on lung function and inflammation in rabbit model of acute respiratory distress syndrome. Physiol Rep 2021; 9:e14700. [PMID: 33403805 PMCID: PMC7786196 DOI: 10.14814/phy2.14700] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/29/2020] [Accepted: 12/03/2020] [Indexed: 02/04/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is associated with diffuse inflammation, alveolar epithelial damage, and leakage of plasma proteins into the alveolar space, which together contribute to inactivation of pulmonary surfactant and respiratory failure. Exogenous surfactant delivery is therefore considered to hold potential for ARDS treatment, but clinical trials with natural derived surfactant or synthetic surfactant containing a surfactant protein C (SP-C) analogue have been negative. Synthetic surfactant CHF5633, containing analogues of SP-B and SP-C, may be effective against ARDS. The aim here was to compare treatment effects of CHF5633 and animal-derived surfactant poractant alfa in animal model of ARDS. ARDS was induced in adult New Zealand rabbits by mild lung lavages followed by injurious ventilation until respiratory failure (P/F ratio <26.7 kPa). The animals were then treated with intratracheal bolus of 200 mg/kg CHF5633 or poractant alfa (Curosurf® ), or air as control. The animals were subsequently ventilated for an additional 4 hr and respiratory parameters were recorded regularly. Postmortem, histological analysis, degree of lung edema, and levels of the cytokines TNFα, IL-6, and IL-8 in lung homogenates were evaluated. Both surfactant preparations improved lung function, reduced the levels of pro-inflammatory cytokines, and degree of lung edema to very similar degrees versus the controls. No significant differences in any of the analyzed parameters were observed between the CHF5633- and poractant alfa-treated groups. This study indicates that single dose of CHF5633 improves lung function and attenuates inflammation as effectively as poractant alfa in experimental ARDS caused by injurious ventilation.
Collapse
Affiliation(s)
- Pavol Mikolka
- Division for NeurogeriatricsDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetHuddingeSweden
- Biomedical Center MartinJessenius Faculty of Medicine in MartinComenius University in BratislavaMartinSlovakia
- Department of PhysiologyJessenius Faculty of Medicine in MartinComenius University in BratislavaMartinSlovakia
| | - Tore Curstedt
- Department of Molecular Medicine and SurgeryKarolinska InstitutetKarolinska University HospitalStockholmSweden
| | - Riccardo Feinstein
- Department of PathologyThe Swedish National Veterinary InstituteUppsalaSweden
| | - Anders Larsson
- Hedenstierna LaboratoryDepartment of Surgical SciencesUppsala UniversityUppsalaSweden
| | - Marian Grendar
- Biomedical Center MartinJessenius Faculty of Medicine in MartinComenius University in BratislavaMartinSlovakia
| | - Anna Rising
- Division for NeurogeriatricsDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetHuddingeSweden
- Department of Anatomy, Physiology and BiochemistrySwedish University of Agricultural SciencesUppsalaSweden
| | - Jan Johansson
- Division for NeurogeriatricsDepartment of Neurobiology, Care Sciences and SocietyKarolinska InstitutetHuddingeSweden
| |
Collapse
|
22
|
Rong Z, Mo L, Pan R, Zhu X, Cheng H, Li M, Yan L, Lang Y, Zhu X, Chen L, Xia S, Han J, Chang L. Bovine surfactant in the treatment of pneumonia-induced-neonatal acute respiratory distress syndrome (NARDS) in neonates beyond 34 weeks of gestation: a multicentre, randomized, assessor-blinded, placebo-controlled trial. Eur J Pediatr 2021; 180:1107-1115. [PMID: 33084980 PMCID: PMC7575859 DOI: 10.1007/s00431-020-03821-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 11/30/2022]
Abstract
Neonatal acute respiratory distress syndrome (NARDS) reflects pulmonary surfactant dysfunction, and the usage of bovine surfactant (Calsurf) supplement may therefore be beneficial. To determine whether bovine surfactant given in NARDS can improve oxygenation and survival rate, we conducted a multicenter, randomized trial between January 2018 and June 2019, and we compared Calsurf treatment to controls in neonates with pneumonia accompanied by NARDS. Neonates who met the Montreux criteria definition of NARDS were included, and those with congenital heart and lung malformations were excluded. Primary outcomes were oxygenation index (OI) after Calsurf administration, and secondary outcomes were mortality, and duration of ventilator and oxygen between the two groups, and also other morbidities. Cumulatively, 328 neonates were recruited and analyzed, 162 in the control group, and 166 in the Calsurf group. The results shows that OI in the Calsurf group were significantly lower than that in the control group at 4 h (7.2 ± 2.7 and 11.4 ± 9.1, P = 0.001); similarly, OI in the Calsurf group were significantly lower than in the control group at 12 h ( 7.5 ± 3.1 and 11.2 ± 9.2, P = 0.001). Mortality and duration of ventilator support or oxygen use between the two groups were not significantly different.Conclusion: Calsurf acutely improved OI immediately after administration in pneumonia-induced NARDS; although, we observed no significant decrease in mortality, duration of ventilator or oxygen, or major morbidity. What is known: • The definition proposed as the Monteux criteria for neonatal acute respiratory distress syndrome (NARDS). • Surfactant acutely improved oxygenation and significantly decreased mortality in children and adolescents with acute lung injury. What is new: • This is the first large randomized controlled trail to study on surfactant treatment of neonates with acute respiratory distress syndromes. • Surfactant acutely improved oxygenation immediately after administration in pneumonia-induced NARDS at a gestational age beyond 34 weeks.
Collapse
Affiliation(s)
- Zhihui Rong
- Department of Neonatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Luxia Mo
- Department of Neonatology, Wuhan Maternal and Child Healthcare Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Rui Pan
- Department of Neonatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Xiaofang Zhu
- Department of Neonatology, Jingzhou Central Hospital, Jingzhou, China
| | - Hongbin Cheng
- Department of Neonatology, HuangShi Maternal and Child Healthcare Hospital, Huangshi, China
| | - Maojun Li
- Department of Neonatology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Sichuan, China
| | - Lubiao Yan
- Department of Neonatology, Maternal and Child Healthcare Hospital, Nanjing Medical University, Nanjing, China
| | - Yujie Lang
- Department of Neonatology, Children’s Hospital of Jinan, Jinan, China
| | - Xiaoshan Zhu
- Department of Neonatology, Anhui Provincial Children’s Hospital, Hefei, China
| | - Liping Chen
- Department of Neonatology, Jiangxi Provincial Children’s Hospital, Jiangxi, China
| | - Shiwen Xia
- Department of Neonatology, Hubei Maternity and Child Heath Hospital, Wuhan, China
| | - Jun Han
- Department of Neonatology, The First Hospital of Jilin University, Changchun, China
| | - Liwen Chang
- Department of Neonatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
23
|
Liang G, Wu R, Jiang L, Liu Y, Mao G, Huang Z, Qi L, Jiang H. The role of lipoprotein-associated phospholipase A2 in acute kidney injury of septic mice. Transl Androl Urol 2020; 9:2192-2199. [PMID: 33209683 PMCID: PMC7658152 DOI: 10.21037/tau-20-1173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background This experiment aimed to investigate the role and mechanism of lipoprotein-associated phospholipase A2 (Lp-PLA2) in kidney injury in septic mice induced by cecal ligation and perforation (CLP). Methods Male BALB/c mice were randomly divided into two groups: sham-operation group (Sham group) and septic group (CLP group). The septic model was simulated by cecal ligation and puncture method, but only cecal ligation was used for the sham operation group. The whole serum and renal tissue samples of the mice were collected 24 hours after modeling in both groups. Hematoxylin-eosin (HE) staining was used to observe the pathological changes of renal tissue, the renal injury score was recorded, and the creatinine (Cr) and blood urea nitrogen (BUN) levels were detected by automatic biochemical analyzer, while the serum Lp-PLA2 level was detected by enzyme-linked immunosorbent assay (ELISA). The 7-day survival rate and the survival curve of the two groups were statistically analyzed. Results Compared with the Sham group, the pathological score of renal injury in the CLP Group was higher, the level of Lp-PLA2 in serum was significantly increased (all P<0.01), and the expression of Lp-PLA2 in renal tissue was significantly elevated (all P<0.01). Furthermore, the 7-day survival rate of the Sham group was 90%, while that of CLP group was 25%. Conclusions The expression level of Lp-PLA2 in blood and kidney tissue of septic mice was increased and correlated with prognosis. However, the predictive value of Lp-PLA2 for prognosis in septic mice needs further study.
Collapse
Affiliation(s)
- Guiwen Liang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Ruo Wu
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Lan Jiang
- Department of Gastroenterology, First People's Hospital of Zhangjiagang City, Zhangjiagang, China
| | - Yanfang Liu
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Guomin Mao
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Zhongwei Huang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Lei Qi
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Haiyan Jiang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
24
|
De Luca D, Shankar-Aguilera S, Autilio C, Raschetti R, Vedovelli L, Fitting C, Payré C, Jeammet L, Perez-Gil J, Cogo PE, Carnielli VP, Lambeau G, Touqui L. Surfactant-secreted phospholipase A2interplay and respiratory outcome in preterm neonates. Am J Physiol Lung Cell Mol Physiol 2020; 319:L95-L104. [DOI: 10.1152/ajplung.00462.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Secreted phospholipase A2hydrolyzes surfactant phospholipids and is crucial for the inflammatory cascade; preterm neonates are treated with exogenous surfactant, but the interaction between surfactant and phospholipase is unknown. We hypothesize that this interplay is complex and the enzyme plays a relevant role in neonates needing surfactant replacement. We aimed to: 1) identify phospholipases A2isoforms expressed in preterm lung; 2) study the enzyme role on surfactant retreatment and function and the effect of exogenous surfactant on the enzyme system; and 3) verify whether phospholipase A2is linked to respiratory outcomes. In bronchoalveolar lavages of preterm neonates, we measured enzyme activity (alone or with inhibitors), enzyme subtypes, surfactant protein-A, and inflammatory mediators. Surfactant function and phospholipid profile were also tested. Urea ratio was used to obtain epithelial lining fluid concentrations. Follow-up data were prospectively collected. Subtype-IIA is the main phospholipase isoform in preterm lung, although subtype-IB may be significantly expressed. Neonates needing surfactant retreatment have higher enzyme activity ( P = 0.021) and inflammatory mediators ( P always ≤ 0.001) and lower amounts of phospholipids ( P always < 0.05). Enzyme activity was inversely correlated to surfactant adsorption (ρ = −0.6; P = 0.008; adjusted P = 0.009), total phospholipids (ρ = −0.475; P = 0.05), and phosphatidylcholine (ρ = −0.622; P = 0.017). Exogenous surfactant significantly reduced global phospholipase activity ( P < 0.001) and subtype-IIA ( P = 0.005) and increased dioleoylphosphatidylglycerol ( P < 0.001) and surfactant adsorption ( P < 0.001). Enzyme activity correlated with duration of ventilation (ρ = 0.679, P = 0.005; adjusted P = 0.04) and respiratory morbidity score at 12 mo postnatal age (τ-b = 0.349, P = 0.037; adjusted P = 0.043) but was not associated with mortality, bronchopulmonary dysplasia, or other long-term respiratory outcomes.
Collapse
Affiliation(s)
- Daniele De Luca
- Division of Pediatrics and Neonatal Critical Care, “A.Béclère” Medical Center, South Paris University Hospitals, Assistance Publique – Hôpitaux de Paris (APHP), Paris, France
- Physiopathology and Therapeutic Innovation Unit-INSERM U999, South Paris-Saclay University, Paris, France
- Cystic fibrosis and Bronchial diseases team-INSERM U938, Institut Pasteur, Paris, France
| | - Shivani Shankar-Aguilera
- Division of Pediatrics and Neonatal Critical Care, “A.Béclère” Medical Center, South Paris University Hospitals, Assistance Publique – Hôpitaux de Paris (APHP), Paris, France
- Cystic fibrosis and Bronchial diseases team-INSERM U938, Institut Pasteur, Paris, France
| | - Chiara Autilio
- Department of Biochemistry and Molecular Biology, Faculty of Biology, and Research Institut-Hospital “12 de Octubre,” Complutense University, Madrid, Spain
| | - Roberto Raschetti
- Division of Pediatrics and Neonatal Critical Care, “A.Béclère” Medical Center, South Paris University Hospitals, Assistance Publique – Hôpitaux de Paris (APHP), Paris, France
| | - Luca Vedovelli
- PCare Laboratory, Fondazione Istituto di Ricerca Pediatrica “Città della Speranza,” Padua, Italy
| | | | - Christine Payré
- Université Côte d’Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Valbonne Sophia Antipolis, France
| | - Louise Jeammet
- Université Côte d’Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Valbonne Sophia Antipolis, France
| | - Jesus Perez-Gil
- Department of Biochemistry and Molecular Biology, Faculty of Biology, and Research Institut-Hospital “12 de Octubre,” Complutense University, Madrid, Spain
| | - Paola E. Cogo
- PCare Laboratory, Fondazione Istituto di Ricerca Pediatrica “Città della Speranza,” Padua, Italy
- Division of Pediatrics, Department of Medicine and Surgery, University of Udine, Udine, Italy
| | - Virgilio P. Carnielli
- PCare Laboratory, Fondazione Istituto di Ricerca Pediatrica “Città della Speranza,” Padua, Italy
- Division of Neonatology, “G. Salesi” Women’s and Children Hospital, Polytechnical University of Marche, Ancona, Italy
| | - Gérard Lambeau
- Université Côte d’Azur, Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275, Valbonne Sophia Antipolis, France
| | - Lhousseine Touqui
- Cystic fibrosis and Bronchial diseases team-INSERM U938, Institut Pasteur, Paris, France
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine (CRSA), Paris, France
| |
Collapse
|
25
|
Zebialowicz Ahlström J, Massaro F, Mikolka P, Feinstein R, Perchiazzi G, Basabe-Burgos O, Curstedt T, Larsson A, Johansson J, Rising A. Synthetic surfactant with a recombinant surfactant protein C analogue improves lung function and attenuates inflammation in a model of acute respiratory distress syndrome in adult rabbits. Respir Res 2019; 20:245. [PMID: 31694668 PMCID: PMC6836435 DOI: 10.1186/s12931-019-1220-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 10/17/2019] [Indexed: 02/06/2023] Open
Abstract
AIM In acute respiratory distress syndrome (ARDS) damaged alveolar epithelium, leakage of plasma proteins into the alveolar space and inactivation of pulmonary surfactant lead to respiratory dysfunction. Lung function could potentially be restored with exogenous surfactant therapy, but clinical trials have so far been disappointing. These negative results may be explained by inactivation and/or too low doses of the administered surfactant. Surfactant based on a recombinant surfactant protein C analogue (rSP-C33Leu) is easy to produce and in this study we compared its effects on lung function and inflammation with a commercial surfactant preparation in an adult rabbit model of ARDS. METHODS ARDS was induced in adult New Zealand rabbits by mild lung-lavages followed by injurious ventilation (VT 20 m/kg body weight) until P/F ratio < 26.7 kPa. The animals were treated with two intratracheal boluses of 2.5 mL/kg of 2% rSP-C33Leu in DPPC/egg PC/POPG, 50:40:10 or poractant alfa (Curosurf®), both surfactants containing 80 mg phospholipids/mL, or air as control. The animals were subsequently ventilated (VT 8-9 m/kg body weight) for an additional 3 h and lung function parameters were recorded. Histological appearance of the lungs, degree of lung oedema and levels of the cytokines TNFα IL-6 and IL-8 in lung homogenates were evaluated. RESULTS Both surfactant preparations improved lung function vs. the control group and also reduced inflammation scores, production of pro-inflammatory cytokines, and formation of lung oedema to similar degrees. Poractant alfa improved compliance at 1 h, P/F ratio and PaO2 at 1.5 h compared to rSP-C33Leu surfactant. CONCLUSION This study indicates that treatment of experimental ARDS with synthetic lung surfactant based on rSP-C33Leu improves lung function and attenuates inflammation.
Collapse
Affiliation(s)
- J Zebialowicz Ahlström
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - F Massaro
- Anesthesia and Intesive Care, Villa Anthea Hospital, Bari, Italy
| | - P Mikolka
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,Biomedical Center Martin and Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - R Feinstein
- Department of Pathology, The Swedish National Veterinary Institute, Uppsala, Sweden
| | - G Perchiazzi
- Hedenstierna Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - O Basabe-Burgos
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - T Curstedt
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - A Larsson
- Hedenstierna Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - J Johansson
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - A Rising
- Division for Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden. .,Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden.
| |
Collapse
|
26
|
Hu Y, Ren J, Wang L, Zhao X, Zhang M, Shimizu K, Zhang C. Protective effects of total alkaloids from Dendrobium crepidatum against LPS-induced acute lung injury in mice and its chemical components. PHYTOCHEMISTRY 2018; 149:12-23. [PMID: 29438749 DOI: 10.1016/j.phytochem.2018.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/13/2018] [Accepted: 02/05/2018] [Indexed: 05/26/2023]
Abstract
Dendrobium crepidatum was one of the sources of Herba Dendrobii, a famous and precious traditional Chinese medicine. Indolizine-type alkaloids are the main characteristic ingredients of D. crepidatum, which possesses a variety of changeable skeletons. In the present study, we found that the total alkaloids of D. crepidatum (TAD) can inhibit the production of nitric oxide (NO) in lipopolysaccharide (LPS)-activated macrophages and showed protective effects against LPS-induced acute lung injury (ALI) in mice through downregulating the TLR4-mediated MyD88/MAPK signaling pathway. Further phytochemical study showed that six previously undescribed indolizine-type compounds, including a racemic mixture (dendrocrepidine A-E) were isolated from TAD. Meanwhile, dendrocrepidine F was separated into a pair of enantiomers by a chiral chromatography, and their absolute configurations were assigned by single-crystal X-ray diffraction analysis. The isomer (-)-dendrocrepidine F showed higher anti-inflammatory effects by inhibiting NO production in LPS-treated macrophages with an IC50 value of 13.3 μM. Taken together, indolizine-type alkaloids are the active components of D. crepidatum through downregulating the TLR4-mediated pathway, indicating some kind of therapy of TAD for ALI treatment.
Collapse
Affiliation(s)
- Yang Hu
- State Key Laboratory of Natural Medicines, Research Department of Pharmacognosy, China Pharmaceutical University, 639# Longmian Road, Nanjing, 211198, PR China; College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Jie Ren
- State Key Laboratory of Natural Medicines, Research Department of Pharmacognosy, China Pharmaceutical University, 639# Longmian Road, Nanjing, 211198, PR China
| | - Lei Wang
- State Key Laboratory of Natural Medicines, Research Department of Pharmacognosy, China Pharmaceutical University, 639# Longmian Road, Nanjing, 211198, PR China
| | - Xin Zhao
- State Key Laboratory of Natural Medicines, Research Department of Pharmacognosy, China Pharmaceutical University, 639# Longmian Road, Nanjing, 211198, PR China
| | - Mian Zhang
- State Key Laboratory of Natural Medicines, Research Department of Pharmacognosy, China Pharmaceutical University, 639# Longmian Road, Nanjing, 211198, PR China
| | - Kuniyoshi Shimizu
- Department of Forest and Forest Products Sciences, Kyushu University, 6-10-1 Hakozaki, Higashi-ku, Fukuoka, 812-8581, Japan
| | - Chaofeng Zhang
- State Key Laboratory of Natural Medicines, Research Department of Pharmacognosy, China Pharmaceutical University, 639# Longmian Road, Nanjing, 211198, PR China.
| |
Collapse
|
27
|
In silico investigation of the molecular effects caused by R123H variant in secretory phospholipase A2-IIA associated with ARDS. J Mol Graph Model 2018. [PMID: 29529495 DOI: 10.1016/j.jmgm.2018.02.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Phospholipase A2-IIA catalyzes the hydrolysis of the sn-2 ester of glycerophospholipids. A rare c.428G > A (p.Arg143His) variant in PLA2G2A gene was found in two infants affected by acute respiratory distress syndrome (ARDS) by whole coding region and exon/intron boundaries sequencing. To obtain insights into the possible molecular effects of the rare R123H mutation in secretory PLA2-IIA (sPLA2-IIA), molecular modelling, molecular dynamics (MD) using principal component analysis (PCA) and continuum electrostatic calculations were conducted on the crystal structure of the wild type protein and on a generated model structure of the R123H mutant. Analysis of MD trajectories indicate that the overall stability of the protein is not affected by this mutation but nevertheless the catalytically crucial H-bond between Tyr51 and Asp91 as well as main electrostatic interactions in the region close to the mutation site are altered. PCA results indicate that the R123H replacement alter the internal molecular motions of the enzyme and that collective motions are increased. Electrostatic surface potential studies suggest that after mutation the interfacial binding to anionic phospholipid membranes and anionic proteins may be changed. The strengthening of electrostatic interactions may be propagated into the active site region thus potentially affecting the substrate recognition and enzymatic activity. Our findings provide the basis for further investigation and advances our understanding of the effects of mutations on sPLA2 structure and function.
Collapse
|
28
|
Amigoni A, Pettenazzo A, Stritoni V, Circelli M. Surfactants in Acute Respiratory Distress Syndrome in Infants and Children: Past, Present and Future. Clin Drug Investig 2018; 37:729-736. [PMID: 28510235 PMCID: PMC5509808 DOI: 10.1007/s40261-017-0532-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There is a lack of definitive data on the effective management of acute respiratory distress syndrome (ARDS) in infants and children. The development and validation of the Berlin definition (BD) for ARDS and the Pediatric Acute Lung Injury Consensus Conference (PALICC) recommendations in children represented a major advance in optimizing research and treatment, mainly due to the introduction of a severe ARDS category. Proposed reasons for the lack of consistent results with surfactants in children and infants compared with neonates include different causes, type of lung damage (direct or indirect), timing and mode of administration as well as the type of surfactant used. Secretory phospholipase A2 plays an important role in inflammation and possible dysfunction of surfactants in ARDS. Bronchoalveolar lavage (BAL) with normal saline and surfactant allows the removal of inhaled material, the recruitment of non-ventilating areas and the maintenance of the surfactant pool size. BAL with diluted surfactant allows rapid absorption of the surfactant at the air/liquid interface, which blocks the progression of pathological lung disease and in turn disrupts the inflammatory cycle. Importantly, it is now recognized that the type of surfactant, the time of administration and the method of administration could all play an important role in the management of ARDS, and there is evidence that surfactant is effective and well tolerated in children and infants with ARDS.
Collapse
Affiliation(s)
- Angela Amigoni
- Paediatric Intensive Care Unit, Department of Women's and Child's Health, University Hospital of Padua, Via Giustiniani 3, 35128, Padua, Italy.
| | - Andrea Pettenazzo
- Paediatric Intensive Care Unit, Department of Women's and Child's Health, University Hospital of Padua, Via Giustiniani 3, 35128, Padua, Italy
| | - Valentina Stritoni
- Paediatric Intensive Care Unit, Department of Women's and Child's Health, University Hospital of Padua, Via Giustiniani 3, 35128, Padua, Italy
| | | |
Collapse
|
29
|
Gal Y, Sapoznikov A, Falach R, Ehrlich S, Aftalion M, Kronman C, Sabo T. Total Body Irradiation Mitigates Inflammation and Extends the Therapeutic Time Window for Anti-Ricin Antibody Treatment against Pulmonary Ricinosis in Mice. Toxins (Basel) 2017; 9:toxins9090278. [PMID: 28891987 PMCID: PMC5618211 DOI: 10.3390/toxins9090278] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 09/05/2017] [Accepted: 09/09/2017] [Indexed: 12/02/2022] Open
Abstract
Ricin, a highly toxic plant-derived toxin, is considered a potential weapon in biowarfare and bioterrorism due to its pronounced toxicity, high availability, and ease of preparation. Pulmonary exposure to ricin results in the generation of an acute edematous inflammation followed by respiratory insufficiency and death. Massive neutrophil recruitment to the lungs may contribute significantly to ricin-mediated morbidity. In this study, total body irradiation (TBI) served as a non-pharmacological tool to decrease the potential neutrophil-induced lung injury. TBI significantly postponed the time to death of intranasally ricin-intoxicated mice, given that leukopenia remained stable following intoxication. This increase in time to death coincided with a significant reduction in pro-inflammatory marker levels, and led to marked extension of the therapeutic time window for anti-ricin antibody treatment.
Collapse
Affiliation(s)
- Yoav Gal
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Anita Sapoznikov
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Reut Falach
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Sharon Ehrlich
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Moshe Aftalion
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Chanoch Kronman
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Tamar Sabo
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| |
Collapse
|
30
|
The Montreux definition of neonatal ARDS: biological and clinical background behind the description of a new entity. THE LANCET RESPIRATORY MEDICINE 2017; 5:657-666. [PMID: 28687343 DOI: 10.1016/s2213-2600(17)30214-x] [Citation(s) in RCA: 189] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/22/2017] [Accepted: 04/24/2017] [Indexed: 11/21/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is undefined in neonates, despite the long-standing existing formal recognition of ARDS syndrome in later life. We describe the Neonatal ARDS Project: an international, collaborative, multicentre, and multidisciplinary project which aimed to produce an ARDS consensus definition for neonates that is applicable from the perinatal period. The definition was created through discussions between five expert members of the European Society for Paediatric and Neonatal Intensive Care; four experts of the European Society for Paediatric Research; two independent experts from the USA and two from Australia. This Position Paper provides the first consensus definition for neonatal ARDS (called the Montreux definition). We also provide expert consensus that mechanisms causing ARDS in adults and older children-namely complex surfactant dysfunction, lung tissue inflammation, loss of lung volume, increased shunt, and diffuse alveolar damage-are also present in several critical neonatal respiratory disorders.
Collapse
|
31
|
Walther FJ, Gordon LM, Waring AJ. Design of Surfactant Protein B Peptide Mimics Based on the Saposin Fold for Synthetic Lung Surfactants. Biomed Hub 2016; 1. [PMID: 28503550 PMCID: PMC5424708 DOI: 10.1159/000451076] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Surfactant protein (SP)-B is a 79-residue polypeptide crucial for the biophysical and physiological function of endogenous lung surfactant. SP-B is a member of the saposin or saposin-like proteins (SAPLIP) family of proteins that share an overall three-dimensional folding pattern based on secondary structures and disulfide connectivity and exhibit a wide diversity of biological functions. Here, we review the synthesis, molecular biophysics and activity of synthetic analogs of saposin proteins designed to mimic those interactions of the parent proteins with lipids that enhance interfacial activity. Saposin proteins generally interact with target lipids as either monomers or multimers via well-defined amphipathic helices, flexible hinge domains, and insertion sequences. Based on the known 3D-structural motif for the saposin family, we show how bioengineering techniques may be used to develop minimal peptide constructs that maintain desirable structural properties and activities in biomedical applications. One important application is the molecular design, synthesis and activity of Saposin mimics based on the SP-B structure. Synthetic lung surfactants containing active SP-B analogs may be potentially useful in treating diseases of surfactant deficiency or dysfunction including the neonatal respiratory distress syndrome and acute lung injury/acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Frans J Walther
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA.,Department of Pediatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Larry M Gordon
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Alan J Waring
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA.,Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
32
|
Notter RH, Gupta R, Schwan AL, Wang Z, Shkoor MG, Walther FJ. Synthetic lung surfactants containing SP-B and SP-C peptides plus novel phospholipase-resistant lipids or glycerophospholipids. PeerJ 2016; 4:e2635. [PMID: 27812430 PMCID: PMC5088750 DOI: 10.7717/peerj.2635] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 09/30/2016] [Indexed: 01/22/2023] Open
Abstract
Background This study examines the biophysical and preclinical pulmonary activity of synthetic lung surfactants containing novel phospholipase-resistant phosphonolipids or synthetic glycerophospholipids combined with Super Mini-B (S-MB) DATK and/or SP-Css ion-lock 1 peptides that replicate the functional biophysics of surfactant proteins (SP)-B and SP-C. Phospholipase-resistant phosphonolipids used in synthetic surfactants are DEPN-8 and PG-1, molecular analogs of dipalmitoyl phosphatidylcholine (DPPC) and palmitoyl-oleoyl phosphatidylglycerol (POPG), while glycerophospholipids used are active lipid components of native surfactant (DPPC:POPC:POPG 5:3:2 by weight). The objective of the work is to test whether these novel lipid/peptide synthetic surfactants have favorable preclinical activity (biophysical, pulmonary) for therapeutic use in reversing surfactant deficiency or dysfunction in lung disease or injury. Methods Surface activity of synthetic lipid/peptide surfactants was assessed in vitro at 37 °C by measuring adsorption in a stirred subphase apparatus and dynamic surface tension lowering in pulsating and captive bubble surfactometers. Shear viscosity was measured as a function of shear rate on a Wells-Brookfield micro-viscometer. In vivo pulmonary activity was determined by measuring lung function (arterial oxygenation, dynamic lung compliance) in ventilated rats and rabbits with surfactant deficiency/dysfunction induced by saline lavage to lower arterial PO2 to <100 mmHg, consistent with clinical acute respiratory distress syndrome (ARDS). Results Synthetic surfactants containing 5:3:2 DPPC:POPC:POPG or 9:1 DEPN-8:PG-1 combined with 3% (by wt) of S-MB DATK, 3% SP-Css ion-lock 1, or 1.5% each of both peptides all adsorbed rapidly to low equilibrium surface tensions and also reduced surface tension to ≤1 mN/m under dynamic compression at 37 °C. However, dual-peptide surfactants containing 1.5% S-MB DATK + 1.5% SP-Css ion-lock 1 combined with 9:1 DEPN-8:PG-1 or 5:3:2 DPPC:POPC:POPG had the greatest in vivo activity in improving arterial oxygenation and dynamic lung compliance in ventilated animals with ARDS. Saline dispersions of these dual-peptide synthetic surfactants were also found to have shear viscosities comparable to or below those of current animal-derived surfactant drugs, supporting their potential ease of deliverability by instillation in future clinical applications. Discussion Our findings support the potential of dual-peptide synthetic lipid/peptide surfactants containing S-MB DATK + SP-Css ion-lock 1 for treating diseases of surfactant deficiency or dysfunction. Moreover, phospholipase-resistant dual-peptide surfactants containing DEPN-8/PG-1 may have particular applications in treating direct forms of ARDS where endogenous phospholipases are present in the lungs.
Collapse
Affiliation(s)
- Robert H Notter
- Department of Pediatrics, University of Rochester , Rochester , NY , United States
| | - Rohun Gupta
- Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center , Torrance , CA , United States
| | - Adrian L Schwan
- Department of Chemistry, University of Guelph , Guelph , Ontario , Canada
| | - Zhengdong Wang
- Department of Pediatrics, University of Rochester , Rochester , NY , United States
| | - Mohanad Gh Shkoor
- Department of Chemistry, University of Guelph , Guelph , Ontario , Canada
| | - Frans J Walther
- Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, United States; Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
33
|
Qiu J, Chi G, Wu Q, Ren Y, Chen C, Feng H. Pretreatment with the compound asperuloside decreases acute lung injury via inhibiting MAPK and NF-κB signaling in a murine model. Int Immunopharmacol 2016; 31:109-15. [DOI: 10.1016/j.intimp.2015.12.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/07/2015] [Accepted: 12/07/2015] [Indexed: 11/16/2022]
|
34
|
Remesal A, De Luca D, San Feliciano L, Isidoro-Garcia M, Minucci A, Pocino K, Casas J, Fabrias G, Capoluongo ED, de la Cruz DL. Effect of prenatal steroidal inhibition of sPLA2 in a rat model of preterm lung. Pulm Pharmacol Ther 2016; 36:31-6. [DOI: 10.1016/j.pupt.2015.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 11/01/2015] [Accepted: 12/07/2015] [Indexed: 10/22/2022]
|
35
|
Diosmin downregulates the expression of T cell receptors, pro-inflammatory cytokines and NF-κB activation against LPS-induced acute lung injury in mice. Pharmacol Res 2015; 102:1-11. [DOI: 10.1016/j.phrs.2015.09.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 09/04/2015] [Accepted: 09/06/2015] [Indexed: 12/22/2022]
|
36
|
Li Y, Wu Q, Deng Y, Lv H, Qiu J, Chi G, Feng H. D(-)-Salicin inhibits the LPS-induced inflammation in RAW264.7 cells and mouse models. Int Immunopharmacol 2015; 26:286-94. [PMID: 25907238 DOI: 10.1016/j.intimp.2015.04.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 04/03/2015] [Accepted: 04/07/2015] [Indexed: 01/26/2023]
Abstract
D(-)-Salicin is a traditional medicine which has been known to exhibit anti-inflammation and other therapeutic activities. The present study aimed to investigate whether D(-)-Salicin inhibited the LPS-induced inflammation in vivo and in vitro. We evaluated the effect of D(-)-Salicin on cytokines (TNF-α, IL-1β, IL-6 and IL-10) in vivo and in vitro by enzyme-linked immunosorbent assay and signaling pathways (MAPKs and NF-κB) in vivo by Western blot. The results showed that D(-)-Salicin markedly decreased TNF-α, IL-1β and IL-6 concentrations and increased IL-10 concentration. In addition, western blot analysis indicated that D(-)-Salicin suppressed the activation of MAPKs and NF-κB signaling pathways stimulated by LPS. To examine whether D(-)-Salicin ameliorated LPS-induced lung inflammation, inhibitors of MAPKs and NF-κB signaling pathways were administrated intraperitoneally to mice. Interference with specific inhibitors revealed that D(-)-Salicin-mediated cytokine suppression was through MAPKs and NF-κB pathways. In the mouse model of acute lung injury, histopathologic examination indicted that D(-)-Salicin suppressed edema induced by LPS. So it is suggest that D(-)-Salicin might be a potential therapeutic agent against inflammatory diseases.
Collapse
Affiliation(s)
- Yang Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin 130062, PR China
| | - Qianchao Wu
- Key Laboratory of Zoonosis, Ministry of Education, College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin 130062, PR China
| | - Yanhong Deng
- Key Laboratory of Zoonosis, Ministry of Education, College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin 130062, PR China
| | - Hongming Lv
- Key Laboratory of Zoonosis, Ministry of Education, College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin 130062, PR China
| | - Jiaming Qiu
- Key Laboratory of Zoonosis, Ministry of Education, College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin 130062, PR China
| | - Gefu Chi
- Department of Outpatient Clinic, the Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao 028000, People's Republic of China.
| | - Haihua Feng
- Key Laboratory of Zoonosis, Ministry of Education, College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin 130062, PR China.
| |
Collapse
|
37
|
Xu M, Cao FL, Zhang YF, Shan L, Jiang XL, An XJ, Xu W, Liu XZ, Wang XY. Tanshinone IIA therapeutically reduces LPS-induced acute lung injury by inhibiting inflammation and apoptosis in mice. Acta Pharmacol Sin 2015; 36:179-87. [PMID: 25544360 DOI: 10.1038/aps.2014.112] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 08/20/2014] [Indexed: 12/21/2022]
Abstract
AIM To study the effects of tanshinone IIA (TIIA) on lipopolysaccharide (LPS)-induced acute lung injury in mice and the underlying mechanisms. METHODS Mice were injected with LPS (10 mg/kg, i.p.), then treated with TIIA (10 mg/kg, i.p.). Seven hours after LPS injection, the lungs were collected for histological study. Protein, LDH, TNF-α and IL-1β levels in bronchoalveolar lavage fluid (BALF) and myeloperoxidase (MPO) activity in lungs were measured. Cell apoptosis and Bcl-2, caspase-3, NF-κB and HIF-1α expression in lungs were assayed. RESULTS LPS caused marked histological changes in lungs, accompanied by significantly increased lung W/D ratio, protein content and LDH level in BALF, and Evans blue leakage. LPS markedly increased neutrophil infiltration in lungs and inflammatory cytokines in BALF. Furthermore, LPS induced cell apoptosis in lungs, as evidenced by increased TUNEL-positive cells, decreased Bcl-2 content and increased cleaved caspase-3 content. Moreover, LPS significantly increased the expression of NF-κB and HIF-1α in lungs. Treatment of LPS-injected mice with TIIA significantly alleviated these pathological changes in lungs. CONCLUSION TIIA alleviates LPS-induced acute lung injury in mice by suppressing inflammatory responses and apoptosis, which is mediated via inhibition of the NF-κB and HIF-1α pathways.
Collapse
|
38
|
Adenovector-mediated gene transfer of lysophosphatidylcholine acyltransferase 1 attenuates oleic acid-induced acute lung injury in rats. Crit Care Med 2014; 42:e716-24. [PMID: 25319916 DOI: 10.1097/ccm.0000000000000633] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Lysophosphatidylcholine is generated through the hydrolysis of phosphatidylcholine by phospholipase A2 and reversely converted to phosphatidylcholine by lysophosphatidylcholine acyltransferase 1. Although lysophosphatidylcholine is a potent proinflammatory mediator and increased in several types of acute lung injuries, the role of lysophosphatidylcholine acyltransferase 1 has not yet been addressed. We aimed to investigate whether the exogenous expression of lysophosphatidylcholine acyltransferase 1 could attenuate acute lung injury. DESIGN Randomized, prospective animal study, including in vitro primary cell culture test. SETTING University medical center research laboratory. SUBJECTS Adult male Sprague-Dawley rats. INTERVENTIONS Recombinant adenoviruses carrying complementary DNA encoding lysophosphatidylcholine acyltransferase 1 or lacZ (Ad-lacZ) as a control was constructed. Alveolar type II cells were isolated from rats and cultured on tissue-culture inserts. Rats were pretreated with an endobronchial administration of the recombinant adenovirus. One week later, they were IV injected with oleic acid. The lungs were examined 4 hours post oleic acid. MEASUREMENTS AND MAIN RESULTS Adenoviruses carrying complementary DNA encoding lysophosphatidylcholine acyltransferase 1-infected alveolar type II cells showed lower lysophosphatidylcholine levels and a decreased percentage of cell death compared with Ad-lacZ-infected cells or noninfected cells after exposure to hydrogen peroxide for 1 hour. Compared with Ad-lacZ plus oleic acid-treated lungs, adenoviruses carrying complementary DNA encoding lysophosphatidylcholine acyltransferase 1 plus oleic acid-treated lungs showed a lower wet-to-dry lung weight ratio, a higher lung compliance, lower lysophosphatidylcholine contents, higher phosphatidylcholine contents, and a lower apoptosis ratio of alveolar type II cells. Histological scoring revealed that the adenoviruses carrying complementary DNA encoding lysophosphatidylcholine acyltransferase 1-treated lungs developed oleic acid-induced lung injuries that were attenuated compared with those of Ad-lacZ-treated lungs. CONCLUSIONS Exogenous expression of lysophosphatidylcholine acyltransferase 1 protects alveolar type II cells from oxidant-induced cell death in vitro, and endobronchial delivery of a lysophosphatidylcholine acyltransferase 1 transgene effectively attenuates oleic acid-induced acute lung injury in vivo. These results suggest that lysophosphatidylcholine acyltransferase 1 plays a protective role in acute lung injury.
Collapse
|
39
|
Vijayaraj P, Sivaprakasam C, Varthini LV, Sarkar M, Nachiappan V. In vitro exposure of tobacco specific nitrosamines decreases the rat lung phospholipids by enhanced phospholipase A2 activity. Toxicol In Vitro 2014; 28:1097-105. [PMID: 24835565 DOI: 10.1016/j.tiv.2014.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 04/10/2014] [Accepted: 05/05/2014] [Indexed: 01/23/2023]
Abstract
Tobacco-specific nitrosamines (TSNA) have implications in the pathogenesis of various lung diseases and conditions are prevalent even in non-smokers. N-nitrosonornicotine (NNN) and 4-(methyl nitrosamino)-1-(3-pyridyl)-1-butanone (NNK) are potent pulmonary carcinogens present in tobacco product and are mainly responsible for lung cancer. TSNA reacts with pulmonary surfactants, and alters the surfactant phospholipid. The present study was undertaken to investigate the in vitro exposure of rat lung tissue slices to NNK or NNN and to monitor the phospholipid alteration by [(32)P]orthophosphate labeling. Phospholipid content decreased significantly in the presence of either NNK or NNN with concentration and time dependent manner. Phosphatidylcholine (PC) is the main phospholipid of lung and significant reduction was observed in PC ∼61%, followed by phosphatidylglycerol (PG) with 100μM of NNK, whereas NNN treated tissues showed a reduction in phosphatidylserine (PS) ∼60% and PC at 250μM concentration. The phospholipase A2 assays and expression studies reveal that both compounds enhanced phospholipid hydrolysis, thereby reducing the phospholipid content. Collectively, our data demonstrated that both NNK and NNN significantly influenced the surfactant phospholipid level by enhanced phospholipase A2 activity.
Collapse
Affiliation(s)
- Panneerselvam Vijayaraj
- Biomembrane Lab, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India
| | - Chinnarasu Sivaprakasam
- Biomembrane Lab, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India
| | | | - Mary Sarkar
- Biochemistry Department, Indian Institute of Science, Bangalore 560012, India
| | - Vasanthi Nachiappan
- Biomembrane Lab, Department of Biochemistry, School of Life Sciences, Bharathidasan University, Tiruchirappalli, India.
| |
Collapse
|
40
|
Gal Y, Mazor O, Alcalay R, Seliger N, Aftalion M, Sapoznikov A, Falach R, Kronman C, Sabo T. Antibody/doxycycline combined therapy for pulmonary ricinosis: Attenuation of inflammation improves survival of ricin-intoxicated mice. Toxicol Rep 2014; 1:496-504. [PMID: 28962263 PMCID: PMC5598361 DOI: 10.1016/j.toxrep.2014.07.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 07/22/2014] [Accepted: 07/22/2014] [Indexed: 11/17/2022] Open
Abstract
Ricin, a highly toxic plant-derived toxin, is considered a potential weapon in biological warfare due to its high availability and ease of preparation. Pulmonary exposure to ricin results in the generation of an acute edematous inflammation followed by respiratory insufficiency and death. Passive immunization with polyclonal anti-ricin antibodies conferred protection against pulmonary ricinosis, however, at clinically-relevant time points for treatment, survival rates were limited. In this study, intranasal instillation of a lethal dose of ricin to mice, served as a lung challenge model for the evaluation and comparison of different therapeutic modalities against pulmonary ricinosis. We show that treatment with doxycycline resulted in a significant reduction of pro-inflammatory cytokines, markers of oxidative stress and capillary permeability in the lungs of the mice. Moreover, survival rates of mice intoxicated with ricin and treated 24 h later with anti-ricin antibody were significantly improved by co-administration of doxycycline. In contrast, co-administration of the steroid drug dexamethasone with anti-ricin antibodies did not increase survival rates when administered at late hours after intoxication, however dexamethasone did exert a positive effect on survival when applied in conjunction with the doxycycline treatment. These studies strongly suggest that combined therapy, comprised of neutralizing anti-ricin antibodies and an appropriate anti-inflammatory agent, can promote high-level protection against pulmonary ricinosis at clinically-relevant time points post-exposure.
Collapse
Affiliation(s)
- Yoav Gal
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Ohad Mazor
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Ron Alcalay
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Nehama Seliger
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Moshe Aftalion
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Anita Sapoznikov
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Reut Falach
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Chanoch Kronman
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Tamar Sabo
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| |
Collapse
|
41
|
Li W, Huang H, Niu X, Fan T, Hu H, Li Y, Yao H, Li H, Mu Q. Tetrahydrocoptisine Protects Rats from LPS-Induced Acute Lung Injury. Inflammation 2014; 37:2106-15. [PMID: 24928630 DOI: 10.1007/s10753-014-9945-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
42
|
De Luca D, Lopez-Rodriguez E, Minucci A, Vendittelli F, Gentile L, Stival E, Conti G, Piastra M, Antonelli M, Echaide M, Perez-Gil J, Capoluongo ED. Clinical and biological role of secretory phospholipase A2 in acute respiratory distress syndrome infants. Crit Care 2013; 17:R163. [PMID: 23883784 PMCID: PMC4057254 DOI: 10.1186/cc12842] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 06/15/2013] [Accepted: 07/24/2013] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Secretory phospholipase A2 is supposed to play a role in acute lung injury but no data are available for pediatric acute respiratory distress syndrome (ARDS). It is not clear which enzyme subtypes are secreted and what the relationships are between enzyme activity, biophysical and biochemical parameters, and clinical outcomes. We aimed to measure the enzyme and identify its subtypes and to study its biochemical and biophysical effect. The secondary aim was to correlate enzyme activity with clinical outcome. METHODS Bronchoalveolar lavage was performed in 24 infants with ARDS and 14 controls with no lung disease. Samples were assayed for secretory phospholipase A2 and molecules related to its activity and expression. Western blotting and captive bubble surfactometry were also performed. Clinical data were real time downloaded. RESULTS Tumor necrosis factor-α (814 (506-2,499) vs. 287 (111-1,315) pg/mL; P = 0.04), enzyme activity (430 (253-600) vs. 149 (61-387) IU/mL; P = 0.01), free fatty acids (4.3 (2.8-8.6) vs. 2 (0.8-4.6) mM; P = 0.026), and minimum surface tension (25.6 ± 6.1 vs. 18 ± 1.8 mN/m; P = 0.006) were higher in ARDS than in controls. Phospholipids are lower in ARDS than in controls (76.5 (54-100) vs. 1,094 (536-2,907) μg/mL; P = 0.0001). Three enzyme subtypes were identified (-IIA, -V, -X), although in lower quantities in controls; another subtype (-IB) was mainly detected in ARDS. Significant correlations exist between enzyme activity, free fatty acids (ρ = 0.823; P < 0.001), and surface tension (ρ = 0.55; P < 0.028). Correlations also exist with intensive care stay (ρ = 0.54; P = 0.001), PRISM-III24 (ρ = 0.79; P< 0.001), duration of ventilation (ρ = 0.53; P = 0.002), and oxygen therapy (ρ = 0.54; P = 0.001). CONCLUSIONS Secretory phospholipase A2 activity is raised in pediatric ARDS and constituted of four subtypes. Enzyme correlates with some inflammatory mediators, surface tension, and major clinical outcomes. Secretory phospholipase A2 may be a clinically relevant target in pediatric ARDS.
Collapse
Affiliation(s)
- Daniele De Luca
- Pediatric Intensive Care Unit, Dept of Anesthesiology and Intensive Care, University Hospital 'A. Gemelli', Catholic University of the Sacred Heart, L.go A. Gemelli 8, 00168 Rome, Italy
- Laboratory of Clinical Molecular Biology, Dept of Laboratory Medicine, University Hospital 'A. Gemelli', Catholic University of the Sacred Heart, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Elena Lopez-Rodriguez
- Dept of Biochemistry, Faculty of Biology, Complutense University, av. Complutense s/n, 28040 Madrid, Spain
| | - Angelo Minucci
- Laboratory of Clinical Molecular Biology, Dept of Laboratory Medicine, University Hospital 'A. Gemelli', Catholic University of the Sacred Heart, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Francesca Vendittelli
- Laboratory of Clinical Molecular Biology, Dept of Laboratory Medicine, University Hospital 'A. Gemelli', Catholic University of the Sacred Heart, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Leonarda Gentile
- Laboratory of Clinical Molecular Biology, Dept of Laboratory Medicine, University Hospital 'A. Gemelli', Catholic University of the Sacred Heart, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Eleonora Stival
- Pediatric Intensive Care Unit, Dept of Anesthesiology and Intensive Care, University Hospital 'A. Gemelli', Catholic University of the Sacred Heart, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Giorgio Conti
- Pediatric Intensive Care Unit, Dept of Anesthesiology and Intensive Care, University Hospital 'A. Gemelli', Catholic University of the Sacred Heart, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Marco Piastra
- Pediatric Intensive Care Unit, Dept of Anesthesiology and Intensive Care, University Hospital 'A. Gemelli', Catholic University of the Sacred Heart, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Massimo Antonelli
- Pediatric Intensive Care Unit, Dept of Anesthesiology and Intensive Care, University Hospital 'A. Gemelli', Catholic University of the Sacred Heart, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Mercedes Echaide
- Dept of Biochemistry, Faculty of Biology, Complutense University, av. Complutense s/n, 28040 Madrid, Spain
| | - Jesus Perez-Gil
- Dept of Biochemistry, Faculty of Biology, Complutense University, av. Complutense s/n, 28040 Madrid, Spain
| | - Ettore D Capoluongo
- Laboratory of Clinical Molecular Biology, Dept of Laboratory Medicine, University Hospital 'A. Gemelli', Catholic University of the Sacred Heart, L.go A. Gemelli 8, 00168 Rome, Italy
| |
Collapse
|
43
|
Liu F, Sun GQ, Gao HY, Li RS, Soromou LW, Chen N, Deng YH, Feng HH. Angelicin regulates LPS-induced inflammation via inhibiting MAPK/NF-κB pathways. J Surg Res 2013; 185:300-9. [PMID: 23816246 DOI: 10.1016/j.jss.2013.05.083] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 04/26/2013] [Accepted: 05/23/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Angelicin is a furocoumarin found in Psoralea corylifolia L. fruit. The purpose of this study was to investigate the protective ability of angelicin against inflammation in lipopolysaccharide (LPS)-stimulated RAW 264.7 cells and LPS-induced in vivo acute lung injury model. MATERIALS AND METHODS The concentrations of tumor necrosis factor alpha (TNF-α) and interleukin (IL)-6 in the culture supernatants of RAW 264.7 cells were determined 24 h after LPS administration. ALI was induced by intratracheal instillation of LPS. Six hours after LPS inhalation, bronchoalveolar lavage fluid and lung tissue samples were obtained for enzyme-linked immunosorbent assay, histologic, and Western blotting analyses. RESULTS The results showed that pretreatment with angelicin markedly downregulated TNF-α and IL-6 levels in vitro and in vivo, and significantly decreased the amount of inflammatory cells, lung wet-to-dry weight ratio, and myeloperoxidase activity in LPS-induced ALI mice. Furthermore, Western blotting analysis results demonstrated that angelicin blocked the phosphorylation of IκBα, NF-κBp65, p38 MAPK, and JNK in LPS-induced ALI. CONCLUSIONS These results suggest that angelicin was potentially advantageous to prevent inflammatory diseases by inhibiting NF-κB and MAPK pathways. Our data indicated that angelicin might be a potential new agent for prevention of inflammatory reactions and diseases in the clinic.
Collapse
Affiliation(s)
- Fang Liu
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | | | | | | | | | | | | | | |
Collapse
|
44
|
De Luca D, Minucci A, Piastra M, Cogo PE, Vendittelli F, Marzano L, Gentile L, Giardina B, Conti G, Capoluongo ED. Ex vivo effect of varespladib on secretory phospholipase A2 alveolar activity in infants with ARDS. PLoS One 2012; 7:e47066. [PMID: 23071714 PMCID: PMC3469496 DOI: 10.1371/journal.pone.0047066] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2012] [Accepted: 09/07/2012] [Indexed: 11/24/2022] Open
Abstract
Background Secretory phospholipase A2 (sPLA2) plays a pivotal role in acute respiratory distress syndrome (ARDS). This enzyme seems an interesting target to reduce surfactant catabolism and lung tissue inflammation. Varespladib is a specifically designed indolic sPLA2 inhibitor, which has shown promising results in animals and adults. No specific data in pediatric ARDS patients are yet available. Methods We studied varespladib in broncho-alveolar lavage (BAL) fluids obtained ex vivo from pediatric ARDS patients. Clinical data and worst gas exchange values during the ARDS course were recorded. Samples were treated with saline or 10–40–100 µM varespladib and incubated at 37°C. Total sPLA2 activity was measured by non-radioactive method. BAL samples were subjected to western blotting to identify the main sPLA isotypes with different sensitivity to varespladib. Results was corrected for lavage dilution using the serum-to-BAL urea ratio and for varespladib absorbance. Results Varespladib reduces sPLA2 activity (p<0.0001) at 10,40 and 100 µM; both sPLA2 activity reduction and its ratio to total proteins significantly raise with increasing varespladib concentrations (p<0.001). IC50 was 80 µM. Western blotting revealed the presence of sPLA2-IIA and –IB isotypes in BAL samples. Significant correlations exist between the sPLA2 activity reduction/proteins ratio and PaO2 (rho = 0.63;p<0.001), PaO2/FiO2 (rho = 0.7; p<0.001), oxygenation (rho = −0.6; p<0.001) and ventilation (rho = −0.4;p = 0.038) indexes. Conclusions Varespladib significantly inhibits sPLA2 in BAL of infants affected by post-neonatal ARDS. Inhibition seems to be inversely related to the severity of gas exchange impairment.
Collapse
Affiliation(s)
- Daniele De Luca
- Laboratory of Clinical Molecular Biology, Department of Biochemistry, University Hospital A. Gemelli, Catholic University of the Sacred Heart, Rome, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Seeds MC, Grier BL, Suckling BN, Safta AM, Long DL, Waite BM, Morris PE, Hite RD. Secretory phospholipase A2-mediated depletion of phosphatidylglycerol in early acute respiratory distress syndrome. Am J Med Sci 2012; 343:446-51. [PMID: 22173044 DOI: 10.1097/maj.0b013e318239c96c] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Secretory phospholipases A2 (sPLA2) hydrolyze phospholipids in cell membranes and extracellular structures such as pulmonary surfactant. This study tests the hypothesis that sPLA2 are elevated in human lungs during acute respiratory distress syndrome (ARDS) and that sPLA2 levels are associated with surfactant injury by hydrolysis of surfactant phospholipids. METHODS Bronchoalveolar lavage (BAL) fluid was obtained from 18 patients with early ARDS (<72 hours) and compared with samples from 10 healthy volunteers. Secreted phospholipase A2 levels were measured (enzyme activity and enzyme immunoassay) in conjunction with ARDS subjects' surfactant abnormalities including surfactant phospholipid composition, large and small aggregates distribution and surface tension function. RESULTS BAL sPLA2 enzyme activity was markedly elevated in ARDS samples relative to healthy subjects when measured by ex vivo hydrolysis of both phosphatidylglycerol (PG) and phosphatidylcholine (PC). Enzyme immunoassay identified increased PLA2G2A protein in the ARDS BAL fluid, which was strongly correlated with the sPLA2 enzyme activity against PG. Of particular interest, the authors demonstrated an average depletion of 69% of the PG in the ARDS sample large aggregates relative to the normal controls. Furthermore, the sPLA2 enzyme activity against PG and PC ex vivo correlated with the BAL recovery of in vivo PG and PC, respectively, and also correlated with the altered distribution of the large and small surfactant aggregates. CONCLUSIONS These results support the hypothesis that sPLA2-mediated hydrolysis of surfactant phospholipid, especially PG by PLA2G2A, contributes to surfactant injury during early ARDS.
Collapse
Affiliation(s)
- Michael C Seeds
- Department of Internal Medicine, Section on Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
De Luca D, Minucci A, Trias J, Tripodi D, Conti G, Zuppi C, Capoluongo E. Varespladib Inhibits Secretory Phospholipase A2 in Bronchoalveolar Lavage of Different Types of Neonatal Lung Injury. J Clin Pharmacol 2012; 52:729-737. [DOI: 10.1177/0091270011405498] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
47
|
Huo M, Chen N, Chi G, Yuan X, Guan S, Li H, Zhong W, Guo W, Soromou LW, Gao R, Ouyang H, Deng X, Feng H. Traditional medicine alpinetin inhibits the inflammatory response in Raw 264.7 cells and mouse models. Int Immunopharmacol 2012; 12:241-8. [DOI: 10.1016/j.intimp.2011.11.017] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2011] [Revised: 11/22/2011] [Accepted: 11/28/2011] [Indexed: 11/30/2022]
|
48
|
Raghavendran K, Willson D, Notter RH. Surfactant therapy for acute lung injury and acute respiratory distress syndrome. Crit Care Clin 2011; 27:525-59. [PMID: 21742216 DOI: 10.1016/j.ccc.2011.04.005] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
This article examines exogenous lung surfactant replacement therapy and its usefulness in mitigating clinical acute lung injury (ALI) and the acute respiratory distress syndrome (ARDS). Surfactant therapy is beneficial in term infants with pneumonia and meconium aspiration lung injury, and in children up to age 21 years with direct pulmonary forms of ALI/ARDS. However, extension of exogenous surfactant therapy to adults with respiratory failure and clinical ALI/ARDS remains a challenge. This article reviews clinical studies of surfactant therapy in pediatric and adult patients with ALI/ARDS, focusing on its potential advantages in patients with direct pulmonary forms of these syndromes.
Collapse
Affiliation(s)
- Krishnan Raghavendran
- Division of Acute Care Surgery, Department of Surgery, University of Michigan Health System, 1500 East Medical Center Drive, 1C340A-UH, SPC 5033, Ann Arbor, MI 48109-5033, USA.
| | | | | |
Collapse
|
49
|
Schwan AL, Singh SP, Davy JA, Waring AJ, Gordon LM, Walther FJ, Wang Z, Notter RH. Synthesis and activity of a novel diether phosphonoglycerol in phospholipase-resistant synthetic lipid:peptide lung surfactants(). MEDCHEMCOMM 2011; 2:1167-1173. [PMID: 22530092 PMCID: PMC3331712 DOI: 10.1039/c1md00206f] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
This paper reports the chemical synthesis and purification of a novel phospholipase-resistant C16:0, C16:1 diether phosphonoglycerol with structural analogy to ester-linked anionic phosphatidylglycerol (PG) in endogenous pulmonary surfactant. This diether phosphonoglycerol (PG 1) is studied for phospholipase A(2) (PLA(2)) resistance and for surface activity in synthetic exogenous surfactants combined with Super Mini-B (S-MB) peptide and DEPN-8, a previously-reported diether phosphonolipid analog of dipalmitoyl phosphatidylcholine (DPPC, the major zwitterionic phospholipid in native lung surfactant). Activity experiments measured both adsorption and dynamic surface tension lowering due to the known importance of these surface behaviors in lung surfactant function in vivo. Synthetic surfactants containing 9 : 1 DEPN-8:PG 1 + 3% S-MB were resistant to degradation by PLA(2) in chromatographic studies, while calf lung surfactant extract (CLSE, the substance of the bovine clinical surfactant Infasurf®) was significantly degraded by PLA(2). The 9 : 1 DEPN-8:PG 1 + 3% S-MB mixture also had small but consistent increases in both adsorption and dynamic surface tension lowering ability compared to DEPN-8 + 3% S-MB. Consistent with these surface activity increases, molecular dynamics simulations using Protein Modeller, GROMACS force-field, and PyMOL showed that bilayers containing DPPC and palmitoyl-oleoyl-PC (POPC) as surrogates of DEPN-8 and PG 1 were penetrated to a greater extent by S-MB peptide than bilayers of DPPC alone. These results suggest that PG 1 or related anionic phosphono-PG analogs may have functional utility in phospholipase-resistant synthetic surfactants targeting forms of acute pulmonary injury where endogenous surfactant becomes dysfunctional due to phospholipase activity in the innate inflammatory response.
Collapse
Affiliation(s)
- Adrian L Schwan
- Department of Chemistry, University of Guelph, Guelph, ON, Canada N1G 2W1
| | | | | | | | | | | | | | | |
Collapse
|
50
|
De Luca D, Capoluongo E, Rigo V. Secretory phospholipase A2 pathway in various types of lung injury in neonates and infants: a multicentre translational study. BMC Pediatr 2011; 11:101. [PMID: 22067747 PMCID: PMC3247178 DOI: 10.1186/1471-2431-11-101] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 11/08/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Secretory phospholipase A2 (sPLA2) is a group of enzymes involved in lung tissue inflammation and surfactant catabolism. sPLA2 plays a role in adults affected by acute lung injury and seems a promising therapeutic target. Preliminary data allow foreseeing the importance of such enzyme in some critical respiratory diseases in neonates and infants, as well. Our study aim is to clarify the role of sPLA2 and its modulators in the pathogenesis and clinical severity of hyaline membrane disease, infection related respiratory failure, meconium aspiration syndrome and acute respiratory distress syndrome. sPLA2 genes will also be sequenced and possible genetic involvement will be analysed. METHODS/DESIGN Multicentre, international, translational study, including several paediatric and neonatal intensive care units and one coordinating laboratory. Babies affected by the above mentioned conditions will be enrolled: broncho-alveolar lavage fluid, serum and whole blood will be obtained at definite time-points during the disease course. Several clinical, respiratory and outcome data will be recorded. Laboratory researchers who perform the bench part of the study will be blinded to the clinical data. DISCUSSION This study, thanks to its multicenter design, will clarify the role(s) of sPLA2 and its pathway in these diseases: sPLA2 might be the crossroad between inflammation and surfactant dysfunction. This may represent a crucial target for new anti-inflammatory therapies but also a novel approach to protect surfactant or spare it, improving alveolar stability, lung mechanics and gas exchange.
Collapse
Affiliation(s)
- Daniele De Luca
- Pediatric Intensive Care Unit, Dept of Emergency and Intensive Care, University Hospital "A.Gemelli", Catholic University of the Sacred Heart - Rome, Italy
- Laboratory of Clinical Molecular Biology, Dept of Molecular Medicine, University Hospital "A.Gemelli", Catholic University of the Sacred Heart - Rome, Italy
| | - Ettore Capoluongo
- Pediatric Intensive Care Unit, Dept of Emergency and Intensive Care, University Hospital "A.Gemelli", Catholic University of the Sacred Heart - Rome, Italy
| | - Vincent Rigo
- Neonatal Intensive Care Unit, University of Liège, CHU de Liège (CHR Citadelle), Belgium
| |
Collapse
|