1
|
Zhang W, Wu L, Zhang S. Clinical phenotype of ARDS based on K-means cluster analysis: A study from the eICU database. Heliyon 2024; 10:e39198. [PMID: 39469677 PMCID: PMC11513467 DOI: 10.1016/j.heliyon.2024.e39198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024] Open
Abstract
Purpose To explore the characteristics of the clinical phenotype of ARDS based on Machine Learning. Methods This is a study on Machine Learning. Screened cases of acute respiratory distress syndrome (ARDS) in the eICU database collected basic information in the cases and clinical data on the Day 1, Day 3, and Day 7 after the diagnosis of ARDS, respectively. Using the Calinski-Harabasz criterion, Gap Statistic, and Silhouette Coefficient, we determine the optimal clustering number k value. By the K-means cluster analysis to derive clinical phenotype, we analyzed the data collected within the first 24 h. We compared it with the survival of cases under the Berlin standard classification, and also examined the phenotypic conversion within the first 24 h, on day 3, and on day 7 after the diagnosis of ARDS. Results We collected 5054 cases and derived three clinical phenotypes using K-means cluster analysis. Phenotype-I is characterized by fewer abnormal laboratory indicators, higher oxygen partial pressure, oxygenation index, APACHE IV score, systolic and diastolic blood pressure, and lower respiratory rate and heart rate. Phenotype-II is characterized by elevated white blood cell count, blood glucose, creatinine, temperature, heart rate, and respiratory rate. Phenotype-III is characterized by elevated age, partial pressure of carbon dioxide, bicarbonate, GCS score, albumin. The differences in ICU length of stay and in-hospital mortality were significantly different between the three phenotypes (P < 0.05), with phenotype I having the lowest in-hospital mortality (10 %) and phenotype II having the highest (31.8 %). To compare the survival analysis of ARDS patients classified by phenotype and those classified according to Berlin criteria. The results showed that the differences in survival between phenotypes were statistically significant (P < 0.05) under phenotypic classification. Conclusions The clinical classification of ARDS based on K-means clustering analysis is beneficial for further identifying ARDS patients with different characteristics. Compared to the Berlin standard, the new clinical classification of ARDS provides a clearer display of the survival status of different types of patients, which helps to predict patient prognosis.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Critical Care Medicine, Kweichow Moutai Hospital, Renhuai City, Guizhou Province, 564500, China
- Department of Critical Care Medicine, People's Hospital of Leshan, Leshan City, Sichuan Province, 614008, China
| | - Linlin Wu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi City, Guizhou Province, 563000, China
| | - Shucheng Zhang
- Department of Dermatology and Venerology, Qian Foshan Hospital Affiliated to Shandong First Medical University, Jinan City, Shandong Province, 250013, China
| |
Collapse
|
2
|
Siuba MT, Bulgarelli L, Duggal A, Cavalcanti AB, Zampieri FG, Rey DA, Lucena WDR, Maia IS, Paisani DM, Laranjeira LN, Neto AS, Deliberato RO. Differential Effect of Positive End-Expiratory Pressure Strategies in Patients With ARDS: A Bayesian Analysis of Clinical Subphenotypes. Chest 2024; 166:754-764. [PMID: 38768777 PMCID: PMC11489450 DOI: 10.1016/j.chest.2024.04.011] [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: 12/28/2023] [Revised: 03/22/2024] [Accepted: 04/06/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND ARDS is a heterogeneous condition with two subphenotypes identified by different methodologies. Our group similarly identified two ARDS subphenotypes using nine routinely available clinical variables. However, whether these are associated with differential response to treatment has yet to be explored. RESEARCH QUESTION Are there differential responses to positive end-expiratory pressure (PEEP) strategies on 28-day mortality according to subphenotypes in adult patients with ARDS? STUDY DESIGN AND METHODS We evaluated data from two prior ARDS trials (Higher vs Lower Positive End-Expiratory Pressures in Patients With the ARDS [ALVEOLI] and the Alveolar Recruitment in ARDS Trial [ART]) that compared different PEEP strategies. We classified patients into one of two subphenotypes as described previously. We assessed the differential effect of PEEP with a Bayesian hierarchical logistic model for the primary outcome of 28-day mortality. RESULTS We analyzed data from 1,559 patients with ARDS. Compared with lower PEEP, a higher PEEP strategy resulted in higher 28-day mortality in patients with subphenotype A disease in the ALVEOLI study (OR, 1.61; 95% credible interval [CrI], 0.90-2.94) and ART (OR, 1.73; 95% CrI, 1.01-2.98), with a probability of harm resulting from higher PEEP in this subphenotype of 94.3% and 97.7% in the ALVEOLI and ART studies, respectively. Higher PEEP was not associated with mortality in patients with subphenotype B disease in each trial (OR, 0.95 [95% CrI, 0.51-1.73] and 1.00 [95% CrI, 0.63-1.55], respectively), with probability of benefit of 56.4% and 50.7% in the ALVEOLI and ART studies, respectively. These effects were not modified by Pao2 to Fio2 ratio, driving pressure, or the severity of illness for the cohorts. INTERPRETATION We found evidence of differential response to PEEP strategies across two ARDS subphenotypes, suggesting possible harm with a higher PEEP strategy in one subphenotype. These observations may assist with predictive enrichment in future clinical trials.
Collapse
Affiliation(s)
- Matthew T Siuba
- Department of Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, OH.
| | - Lucas Bulgarelli
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Abhijit Duggal
- Department of Critical Care Medicine, Respiratory Institute, Cleveland Clinic, Cleveland, OH
| | | | | | | | | | | | | | | | - Ary Serpa Neto
- Department of Critical Care Medicine, Hospital Israelita Albert Einstein, São Paulo, Brazil; Australian and New Zealand Intensive Care Research Centre, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia; Department of Critical Care, Melbourne Medical School, University of Melbourne, Austin Hospital, Melbourne, VIC, Australia; Department of Intensive Care, Austin Hospital, Melbourne, VIC, Australia
| | - Rodrigo Octávio Deliberato
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Department of Biostatistics, Health Informatics and Data Science (BHIDS), University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
3
|
Rezoagli E, Xin Y, Signori D, Sun W, Gerard S, Delucchi KL, Magliocca A, Vitale G, Giacomini M, Mussoni L, Montomoli J, Subert M, Ponti A, Spadaro S, Poli G, Casola F, Herrmann J, Foti G, Calfee CS, Laffey J, Bellani G, Cereda M. Phenotyping COVID-19 respiratory failure in spontaneously breathing patients with AI on lung CT-scan. Crit Care 2024; 28:263. [PMID: 39103945 PMCID: PMC11301830 DOI: 10.1186/s13054-024-05046-3] [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: 05/17/2024] [Accepted: 07/25/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Automated analysis of lung computed tomography (CT) scans may help characterize subphenotypes of acute respiratory illness. We integrated lung CT features measured via deep learning with clinical and laboratory data in spontaneously breathing subjects to enhance the identification of COVID-19 subphenotypes. METHODS This is a multicenter observational cohort study in spontaneously breathing patients with COVID-19 respiratory failure exposed to early lung CT within 7 days of admission. We explored lung CT images using deep learning approaches to quantitative and qualitative analyses; latent class analysis (LCA) by using clinical, laboratory and lung CT variables; regional differences between subphenotypes following 3D spatial trajectories. RESULTS Complete datasets were available in 559 patients. LCA identified two subphenotypes (subphenotype 1 and 2). As compared with subphenotype 2 (n = 403), subphenotype 1 patients (n = 156) were older, had higher inflammatory biomarkers, and were more hypoxemic. Lungs in subphenotype 1 had a higher density gravitational gradient with a greater proportion of consolidated lungs as compared with subphenotype 2. In contrast, subphenotype 2 had a higher density submantellar-hilar gradient with a greater proportion of ground glass opacities as compared with subphenotype 1. Subphenotype 1 showed higher prevalence of comorbidities associated with endothelial dysfunction and higher 90-day mortality than subphenotype 2, even after adjustment for clinically meaningful variables. CONCLUSIONS Integrating lung-CT data in a LCA allowed us to identify two subphenotypes of COVID-19, with different clinical trajectories. These exploratory findings suggest a role of automated imaging characterization guided by machine learning in subphenotyping patients with respiratory failure. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04395482. Registration date: 19/05/2020.
Collapse
Affiliation(s)
- Emanuele Rezoagli
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.
- Department of Emergency and Intensive Care, Fondazione IRCCS San Gerardo dei Tintori Hospital, Monza, Italy.
| | - Yi Xin
- Department of Anesthesiology, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, USA
| | - Davide Signori
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Wenli Sun
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, USA
| | - Sarah Gerard
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Kevin L Delucchi
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Aurora Magliocca
- Department of Anesthesia and Intensive Care Medicine, Policlinico San Marco, Gruppo Ospedaliero San Donato, Bergamo, Italy
- Department of Medical Physiopathology and Transplants, University of Milan, Milan, Italy
| | - Giovanni Vitale
- Department of Anesthesia and Intensive Care Medicine, Policlinico San Marco, Gruppo Ospedaliero San Donato, Bergamo, Italy
| | - Matteo Giacomini
- Department of Anesthesia and Intensive Care Medicine, Policlinico San Marco, Gruppo Ospedaliero San Donato, Bergamo, Italy
| | - Linda Mussoni
- Istituto per la Sicurezza Sociale, San Marino, San Marino
| | - Jonathan Montomoli
- Department of Anesthesia and Intensive Care, Infermi Hospital, AUSL Romagna, Rimini, Italy
| | - Matteo Subert
- Department of Anesthesia and Intensive Care Medicine, Melzo-Gorgonzola Hospital, Azienda Socio-Sanitaria Territoriale Melegnano e della Martesana, Melegnano, Milan, Italy
| | - Alessandra Ponti
- Department of Anesthesiology and Intensive Care, ASST Lecco, Lecco, Italy
| | - Savino Spadaro
- Anesthesia and Intensive Care, Azienda Ospedaliero-Universitaria of Ferrara, Ferrara, Italy
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Giancarla Poli
- Department of Anaesthesia and Critical Care Medicine, Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Francesco Casola
- Department of Physics, Harvard University, 17 Oxford St., Cambridge, MA, 02138, USA
- Harvard-Smithsonian Centre for Astrophysics, 60 Garden St., Cambridge, MA, 02138, USA
| | - Jacob Herrmann
- Roy J. Carver Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Giuseppe Foti
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Department of Emergency and Intensive Care, Fondazione IRCCS San Gerardo dei Tintori Hospital, Monza, Italy
| | - Carolyn S Calfee
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - John Laffey
- School of Medicine, National University of Ireland Galway, Galway, Ireland
- Department of Anaesthesia and Intensive Care Medicine, Galway University Hospitals, Galway, Ireland
| | - Giacomo Bellani
- University of Trento, Centre for Medical Sciences-CISMed, Trento, Italy
- Department of Anesthesia and Intensive Care, Santa Chiara Hospital, Trento, Italy
| | - Maurizio Cereda
- Department of Anesthesiology, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
4
|
Kaczka DW. Imaging the Lung in ARDS: A Primer. Respir Care 2024; 69:1011-1024. [PMID: 39048146 PMCID: PMC11298232 DOI: 10.4187/respcare.12061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Despite periodic changes in the clinical definition of ARDS, imaging of the lung remains a central component of its diagnostic identification. Several imaging modalities are available to the clinician to establish a diagnosis of the syndrome, monitor its clinical course, or assess the impact of treatment and management strategies. Each imaging modality provides unique insight into ARDS from structural and/or functional perspectives. This review will highlight several methods for lung imaging in ARDS, emphasizing basic operational and physical principles for the respiratory therapist. Advantages and disadvantages of each modality will be discussed in the context of their utility for clinical management and decision-making.
Collapse
Affiliation(s)
- David W Kaczka
- Department of Anesthesia, Department of Radiology, and Roy J Carver Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa
| |
Collapse
|
5
|
Dayan RR, Blau M, Taylor J, Hasidim A, Galante O, Almog Y, Gat T, Shavialiova D, Miller JD, Khazanov G, Abu Ghalion F, Sagy I, Ben Shitrit I, Fuchs L. Lung ultrasound is associated with distinct clinical phenotypes in COVID-19 ARDS: A retrospective observational study. PLoS One 2024; 19:e0304508. [PMID: 38829891 PMCID: PMC11146726 DOI: 10.1371/journal.pone.0304508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND ARDS is a heterogeneous syndrome with distinct clinical phenotypes. Here we investigate whether the presence or absence of large pulmonary ultrasonographic consolidations can categorize COVID-19 ARDS patients requiring mechanical ventilation into distinct clinical phenotypes. METHODS This is a retrospective study performed in a tertiary-level intensive care unit in Israel between April and September 2020. Data collected included lung ultrasound (LUS) findings, respiratory parameters, and treatment interventions. The primary outcome was a composite of three ARDS interventions: prone positioning, high PEEP, or a high dose of inhaled nitric oxide. RESULTS A total of 128 LUS scans were conducted among 23 patients. The mean age was 65 and about two-thirds were males. 81 scans identified large consolidation and were classified as "C-type", and 47 scans showed multiple B-lines with no or small consolidation and were classified as "B-type". The presence of a "C-type" study had 2.5 times increased chance of receiving the composite primary outcome of advanced ARDS interventions despite similar SOFA scores, Pao2/FiO2 ratio, and markers of disease severity (OR = 2.49, %95CI 1.40-4.44). CONCLUSION The presence of a "C-type" profile with LUS consolidation potentially represents a distinct COVID-19 ARDS subphenotype that is more likely to require aggressive ARDS interventions. Further studies are required to validate this phenotype in a larger cohort and determine causality, diagnostic, and treatment responses.
Collapse
Affiliation(s)
- Roy Rafael Dayan
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Maayan Blau
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Jonathan Taylor
- Intensive Care Unit, Soroka University Medical Center, Beersheba, Israel
| | - Ariel Hasidim
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Ori Galante
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
- Division of Pulmonary, Critical Care, and Sleep Medicine, Mount Sinai Hospital, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Yaniv Almog
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
- Division of Pulmonary, Critical Care, and Sleep Medicine, Mount Sinai Hospital, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Tomer Gat
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Darya Shavialiova
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Jacob David Miller
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Georgi Khazanov
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Fahmi Abu Ghalion
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Iftach Sagy
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
- Clinical Research Center, Soroka University Medical Center, Beersheba, Israel
| | - Itamar Ben Shitrit
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
- Clinical Research Center, Soroka University Medical Center, Beersheba, Israel
| | - Lior Fuchs
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
- Division of Pulmonary, Critical Care, and Sleep Medicine, Mount Sinai Hospital, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| |
Collapse
|
6
|
Nasa P, Bos LD, Estenssoro E, van Haren FM, Serpa Neto A, Rocco PR, Slutsky AS, Schultz MJ. Consensus statements on the utility of defining ARDS and the utility of past and current definitions of ARDS-protocol for a Delphi study. BMJ Open 2024; 14:e082986. [PMID: 38670604 PMCID: PMC11057280 DOI: 10.1136/bmjopen-2023-082986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
INTRODUCTION Acute respiratory distress syndrome (ARDS), marked by acute hypoxemia and bilateral pulmonary infiltrates, has been defined in multiple ways since its first description. This Delphi study aims to collect global opinions on the conceptual framework of ARDS, assess the usefulness of components within current and past definitions and investigate the role of subphenotyping. The varied expertise of the panel will provide valuable insights for refining future ARDS definitions and improving clinical management. METHODS A diverse panel of 35-40 experts will be selected based on predefined criteria. Multiple choice questions (MCQs) or 7-point Likert-scale statements will be used in the iterative Delphi rounds to achieve consensus on key aspects related to the utility of definitions and subphenotyping. The Delphi rounds will be continued until a stable agreement or disagreement is achieved for all statements. ANALYSIS Consensus will be considered as reached when a choice in MCQs or Likert-scale statement achieved ≥80% of votes for agreement or disagreement. The stability will be checked by non-parametric χ2 tests or Kruskal Wallis test starting from the second round of Delphi process. A p-value ≥0.05 will be used to define stability. ETHICS AND DISSEMINATION The study will be conducted in full concordance with the principles of the Declaration of Helsinki and will be reported according to CREDES guidance. This study has been granted an ethical approval waiver by the NMC Healthcare Regional Research Ethics Committee, Dubai (NMCHC/CR/DXB/REC/APP/002), owing to the nature of the research. Informed consent will be obtained from all panellists before the start of the Delphi process. The study will be published in a peer-review journal with the authorship agreed as per ICMJE requirements. TRIAL REGISTRATION NUMBER NCT06159465.
Collapse
Affiliation(s)
- Prashant Nasa
- Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Critical Care Medicine, NMC Specialty Hospital, Dubai, UAE
| | - Lieuwe D Bos
- Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands
- Laboratory of Experimental Intensive Care and Anesthesiology, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Respiratory Medicine, Amsterdam UMC, Amsterdam, Netherlands
| | - Elisa Estenssoro
- Facultad de Ciencias Médicas, Universidad Nacional de la Plata, La Plata, Argentina
- Ministerio de Salud de la Provincia de Buenos Aires, La Plata, Argentina
| | - Frank Mp van Haren
- College of Health and Medicine, Australian National University, Canberra, ACT, Australia
- Intensive Care Unit, St George Hospital, Sydney, NSW, Australia
| | - Ary Serpa Neto
- Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands
- Monash University, Clayton, VIC, Australia
- Austin Hospital, Heidelberg, VIC, Australia
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Patricia Rm Rocco
- Laboratory of Pulmonary Investigations, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Arthur S Slutsky
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
- St Michael's Hospital Li Ka Shing Knowledge Institute, Toronto, Ontario, Canada
| | - Marcus J Schultz
- Department of Intensive Care, Amsterdam UMC, Amsterdam, The Netherlands
- Mahidol Oxford Tropical Medicine Research Unit, Bangkok, Thailand
- Nuffield Department of Medicine, Oxford University, Oxford, UK
- Department of Anaesthesiology, General Intensive Care and Pain Medicine, Division of Cardiac Thoracic Vascular Anesthesia and Intensive Care Medicine, Medical University Vienna, Vienna, Austria
| |
Collapse
|
7
|
Levy E, Reilly JP. Pharmacologic Treatments in Acute Respiratory Failure. Crit Care Clin 2024; 40:275-289. [PMID: 38432696 DOI: 10.1016/j.ccc.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Acute respiratory failure relies on supportive care using non-invasive and invasive oxygen and ventilatory support. Pharmacologic therapies for the most severe form of respiratory failure, acute respiratory distress syndrome (ARDS), are limited. This review focuses on the most promising therapies for ARDS, targeting different mechanisms that contribute to dysregulated inflammation and resultant hypoxemia. Significant heterogeneity exists within the ARDS population. Treatment requires prompt recognition of ARDS and an understanding of which patients may benefit most from specific pharmacologic interventions. The key to finding effective pharmacotherapies for ARDS may rely on deeper understanding of pathophysiology and bedside identification of ARDS subphenotypes.
Collapse
Affiliation(s)
- Elizabeth Levy
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, 3400 Spruce Street, Philadelphia, PA 19146, USA
| | - John P Reilly
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, 3400 Spruce Street, Philadelphia, PA 19146, USA.
| |
Collapse
|
8
|
Yang P, Sjoding MW. Acute Respiratory Distress Syndrome: Definition, Diagnosis, and Routine Management. Crit Care Clin 2024; 40:309-327. [PMID: 38432698 DOI: 10.1016/j.ccc.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Acute respiratory distress syndrome (ARDS) is an acute inflammatory lung injury characterized by severe hypoxemic respiratory failure, bilateral opacities on chest imaging, and low lung compliance. ARDS is a heterogeneous syndrome that is the common end point of a wide variety of predisposing conditions, with complex pathophysiology and underlying mechanisms. Routine management of ARDS is centered on lung-protective ventilation strategies such as low tidal volume ventilation and targeting low airway pressures to avoid exacerbation of lung injury, as well as a conservative fluid management strategy.
Collapse
Affiliation(s)
- Philip Yang
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University, 6335 Hospital Parkway, Physicians Plaza Suite 310, Johns Creek, GA 30097, USA.
| | - Michael W Sjoding
- Division of Pulmonary and Critical Care Medicine, University of Michigan, 2800 Plymouth Road, NCRC, Building 16, G027W, Ann Arbor, MI 48109, USA
| |
Collapse
|
9
|
Cysneiros A, Galvão T, Domingues N, Jorge P, Bento L, Martin-Loeches I. ARDS Mortality Prediction Model Using Evolving Clinical Data and Chest Radiograph Analysis. Biomedicines 2024; 12:439. [PMID: 38398041 PMCID: PMC10886631 DOI: 10.3390/biomedicines12020439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/12/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
INTRODUCTION Within primary ARDS, SARS-CoV-2-associated ARDS (C-ARDS) emerged in late 2019, reaching its peak during the subsequent two years. Recent efforts in ARDS research have concentrated on phenotyping this heterogeneous syndrome to enhance comprehension of its pathophysiology. METHODS AND RESULTS A retrospective study was conducted on C-ARDS patients from April 2020 to February 2021, encompassing 110 participants with a mean age of 63.2 ± 11.92 (26-83 years). Of these, 61.2% (68) were male, and 25% (17) experienced severe ARDS, resulting in a mortality rate of 47.3% (52). Ventilation settings, arterial blood gases, and chest X-ray (CXR) were evaluated on the first day of invasive mechanical ventilation and between days two and three. CXR images were scrutinized using a convolutional neural network (CNN). A binary logistic regression model for predicting C-ARDS mortality was developed based on the most influential variables: age, PaO2/FiO2 ratio (P/F) on days one and three, CNN-extracted CXR features, and age. Initial performance assessment on test data (23 patients out of the 110) revealed an area under the receiver operating characteristic (ROC) curve of 0.862 with a 95% confidence interval (0.654-0.969). CONCLUSION Integrating data available in all intensive care units enables the prediction of C-ARDS mortality by utilizing evolving P/F ratios and CXR. This approach can assist in tailoring treatment plans and initiating early discussions to escalate care and extracorporeal life support. Machine learning algorithms for imaging classification can uncover otherwise inaccessible patterns, potentially evolving into another form of ARDS phenotyping. The combined features of these algorithms and clinical variables demonstrate superior performance compared to either element alone.
Collapse
Affiliation(s)
- Ana Cysneiros
- Nova Medical School, Universidade de Lisboa, 1649-004 Lisbon, Portugal;
- Unidade de Urgência Médica, Hospital de São José, Centro Hospitalar Universitário Lisboa Central, 1169-050 Lisbon, Portugal
| | - Tiago Galvão
- Instituto Politécnico de Lisboa/Instituto Superior de Engenharia de Lisboa, 1959-007 Lisbon, Portugal; (T.G.); (N.D.); (P.J.)
| | - Nuno Domingues
- Instituto Politécnico de Lisboa/Instituto Superior de Engenharia de Lisboa, 1959-007 Lisbon, Portugal; (T.G.); (N.D.); (P.J.)
| | - Pedro Jorge
- Instituto Politécnico de Lisboa/Instituto Superior de Engenharia de Lisboa, 1959-007 Lisbon, Portugal; (T.G.); (N.D.); (P.J.)
| | - Luis Bento
- Nova Medical School, Universidade de Lisboa, 1649-004 Lisbon, Portugal;
- Unidade de Urgência Médica, Hospital de São José, Centro Hospitalar Universitário Lisboa Central, 1169-050 Lisbon, Portugal
| | | |
Collapse
|
10
|
Zhang J, Guo Y, Mak M, Tao Z. Translational medicine for acute lung injury. J Transl Med 2024; 22:25. [PMID: 38183140 PMCID: PMC10768317 DOI: 10.1186/s12967-023-04828-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/24/2023] [Indexed: 01/07/2024] Open
Abstract
Acute lung injury (ALI) is a complex disease with numerous causes. This review begins with a discussion of disease development from direct or indirect pulmonary insults, as well as varied pathogenesis. The heterogeneous nature of ALI is then elaborated upon, including its epidemiology, clinical manifestations, potential biomarkers, and genetic contributions. Although no medication is currently approved for this devastating illness, supportive care and pharmacological intervention for ALI treatment are summarized, followed by an assessment of the pathophysiological gap between human ALI and animal models. Lastly, current research progress on advanced nanomedicines for ALI therapeutics in preclinical and clinical settings is reviewed, demonstrating new opportunities towards developing an effective treatment for ALI.
Collapse
Affiliation(s)
- Jianguo Zhang
- Department of Emergency Medicine, The Affiliated Hospital, Jiangsu University, Zhenjiang, 212001, Jiangsu, China
| | - Yumeng Guo
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Michael Mak
- Department of Biomedical Engineering, School of Engineering and Applied Science, Yale University, New Haven, 06520, USA
| | - Zhimin Tao
- Department of Emergency Medicine, The Affiliated Hospital, Jiangsu University, Zhenjiang, 212001, Jiangsu, China.
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
- Department of Biomedical Engineering, School of Engineering and Applied Science, Yale University, New Haven, 06520, USA.
- Zhenjiang Key Laboratory of High Technology Research on Exosomes Foundation and Transformation Application, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
11
|
Miller E, Sampson CU, Desai AA, Karnes JH. Differential drug response in pulmonary arterial hypertension: The potential for precision medicine. Pulm Circ 2023; 13:e12304. [PMID: 37927610 PMCID: PMC10621006 DOI: 10.1002/pul2.12304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/05/2023] [Accepted: 10/11/2023] [Indexed: 11/07/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare, complex, and deadly cardiopulmonary disease. It is characterized by changes in endothelial cell function and smooth muscle cell proliferation in the pulmonary arteries, causing persistent vasoconstriction, resulting in right heart hypertrophy and failure. There are multiple drug classes specific to PAH treatment, but variation between patients may impact treatment response. A small subset of patients is responsive to pulmonary vasodilators and can be treated with calcium channel blockers, which would be deleterious if prescribed to a typical PAH patient. Little is known about the underlying cause of this important difference in vasoresponsive PAH patients. Sex, race/ethnicity, and pharmacogenomics may also factor into efficacy and safety of PAH-specific drugs. Research has indicated that endothelin receptor antagonists may be more effective in women and there have been some minor differences found in certain races and ethnicities, but these findings are muddled by the impact of socioeconomic factors and a lack of representation of non-White patients in clinical trials. Genetic variants in genes such as CYP3A5, CYP2C9, PTGIS, PTGIR, GNG2, CHST3, and CHST13 may influence the efficacy and safety of certain PAH-specific drugs. PAH research faces many challenges, but there is potential for new methodologies to glean new insights into PAH development and treatment.
Collapse
Affiliation(s)
- Elise Miller
- Department of Pharmacy Practice and ScienceUniversity of Arizona R. Ken Coit College of PharmacyTucsonArizonaUSA
| | - Chinwuwanuju Ugo‐Obi Sampson
- Department of Pharmacy Practice and ScienceUniversity of Arizona R. Ken Coit College of PharmacyTucsonArizonaUSA
| | - Ankit A. Desai
- Department of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
| | - Jason H. Karnes
- Department of Pharmacy Practice and ScienceUniversity of Arizona R. Ken Coit College of PharmacyTucsonArizonaUSA
- Department of Biomedical InformaticsVanderbilt University School of MedicineNashvilleTennesseeUSA
| |
Collapse
|
12
|
Wang Y, Liu B, Zhou C, Wang Y, Miao J, Zhao L. Pulmonary embolism induces pneumonia-like lung injury beyond pulmonary infarction. Pulm Circ 2023; 13:e12322. [PMID: 38111797 PMCID: PMC10726156 DOI: 10.1002/pul2.12322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/19/2023] [Accepted: 12/05/2023] [Indexed: 12/20/2023] Open
Abstract
Patients with pulmonary embolism (PE) commonly manifest concomitant "pneumonia," which is generally believed to be either a cause (infection) or a consequence (infarction) of PE. This study aimed to clarify the relationship between PE and "pneumonia-like" lesions beyond pulmonary infection and infarction. Chest computed tomography (CT) images of patients with PE and deep vein thrombosis (DVT) were retrospectively analyzed to compare the incidence of pneumonia lesions. The pathological damage and wet/dry ratio of lung tissues were observed in PE rats and PE plasma-injected rats. In total, 793 and 914 inpatients were enrolled in the PE and DVT groups, respectively. Pneumonia lesions were observed in 36.9% and 26.3% of patients in the PE and DVT groups, respectively (p < 0.0001). Among PE rats, 33.3% exhibited focal severe lung injury, which closely resembled the pathological damage of community-acquired pneumonia. The wet/dry ratio was significantly higher in the PE group than in the PE-control group (4.98 ± 0.08 vs. 4.39 ± 0.06, p < 0.0001). Among PE plasma-injected rats, individuals with focal proven lung injury were found at all experimental points, with an incidence of 27.6%. The lung wet/dry ratio was significantly higher in the PE plasma group than in the PE-control plasma group at 1 and 2 h postinjection (5.02 ± 0.12 vs. 4.61 ± 0.06 and 4.76 ± 0.16 vs. 4.34 ± 0.09, respectively; p < 0.05). In conclusion, the manifestation of pneumonia lesions in chest CT images was higher among PE patients than among DVT patients. Plasma of PE rats could induce focal pneumonia-like lung injury in healthy rats.
Collapse
Affiliation(s)
- Yue Wang
- Department of Pulmonary and Critical Care MedicineShengjing Hospital of China Medical UniversityShenyangChina
| | - Bo Liu
- Medical Research CenterShengjing Hospital of China Medical UniversityShenyangChina
- Liaoning Key Laboratory of Research and Application of Animal Models for Environmental and Metabolic DiseasesShengjing Hospital of China Medical UniversityShenyangChina
| | - Chuming Zhou
- Department of Pulmonary and Critical Care MedicineShengjing Hospital of China Medical UniversityShenyangChina
| | - Yuan Wang
- Department of Pulmonary and Critical Care MedicineShengjing Hospital of China Medical UniversityShenyangChina
| | - Jianing Miao
- Medical Research CenterShengjing Hospital of China Medical UniversityShenyangChina
| | - Li Zhao
- Department of Pulmonary and Critical Care MedicineShengjing Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
13
|
Mannes PZ, Barnes CE, Latoche JD, Day KE, Nedrow JR, Lee JS, Tavakoli S. 2-deoxy-2-[ 18F]fluoro-D-glucose Positron Emission Tomography to Monitor Lung Inflammation and Therapeutic Response to Dexamethasone in a Murine Model of Acute Lung Injury. Mol Imaging Biol 2023; 25:681-691. [PMID: 36941514 PMCID: PMC10027262 DOI: 10.1007/s11307-023-01813-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/30/2023] [Accepted: 03/07/2023] [Indexed: 03/23/2023]
Abstract
PURPOSE To image inflammation and monitor therapeutic response to anti-inflammatory intervention using 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) positron emission tomography (PET) in a preclinical model of acute lung injury (ALI). PROCEDURES Mice were intratracheally administered lipopolysaccharide (LPS, 2.5 mg/kg) to induce ALI or phosphate-buffered saline as the vehicle control. A subset of mice in the ALI group received two intraperitoneal doses of dexamethasone 1 and 24 h after LPS. [18F]FDG PET/CT was performed 2 days after the induction of ALI. [18F]FDG uptake in the lungs was quantified by PET (%ID/mLmean and standardized uptake value (SUVmean)) and ex vivo γ-counting (%ID/g). The severity of lung inflammation was determined by quantifying the protein level of inflammatory cytokines/chemokines and the activity of neutrophil elastase and glycolytic enzymes. In separate groups of mice, flow cytometry was performed to estimate the contribution of individual immune cell types to the total pulmonary inflammatory cell burden under different treatment conditions. RESULTS Lung uptake of [18F]FDG was significantly increased during LPS-induced ALI, and a decreased [18F]FDG uptake was observed following dexamethasone treatment to an intermediate level between that of LPS-treated and control mice. Protein expression of inflammatory biomarkers and the activity of neutrophil elastase and glycolytic enzymes were increased in the lungs of LPS-treated mice versus those of control mice, and correlated with [18F]FDG uptake. Furthermore, dexamethasone-induced decreases in cytokine/chemokine protein levels and enzyme activities correlated with [18F]FDG uptake. Neutrophils were the most abundant cells in LPS-induced ALI, and the pattern of total cell burden during ALI with or without dexamethasone therapy mirrored that of [18F]FDG uptake. CONCLUSIONS [18F]FDG PET noninvasively detects lung inflammation in ALI and its response to anti-inflammatory therapy in a preclinical model. However, high [18F]FDG uptake by bone, brown fat, and myocardium remains a technical limitation for quantification of [18F]FDG in the lungs.
Collapse
Affiliation(s)
- Philip Z Mannes
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
- Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Clayton E Barnes
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joseph D Latoche
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kathryn E Day
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jessie R Nedrow
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Janet S Lee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sina Tavakoli
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
14
|
Belizário J, Garay-Malpartida M, Faintuch J. Lung microbiome and origins of the respiratory diseases. CURRENT RESEARCH IN IMMUNOLOGY 2023; 4:100065. [PMID: 37456520 PMCID: PMC10339129 DOI: 10.1016/j.crimmu.2023.100065] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/08/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023] Open
Abstract
The studies on the composition of the human microbiomes in healthy individuals, its variability in the course of inflammation, infection, antibiotic therapy, diets and different pathological conditions have revealed their intra and inter-kingdom relationships. The lung microbiome comprises of major species members of the phylum Bacteroidetes, Firmicutes, Actinobacteria, Fusobacteria and Proteobacteria, which are distributed in ecological niches along nasal cavity, nasopharynx, oropharynx, trachea and in the lungs. Commensal and pathogenic species are maintained in equilibrium as they have strong relationships. Bacterial overgrowth after dysbiosis and/or imbalanced of CD4+ helper T cells, CD8+ cytotoxic T cells and regulatory T cells (Treg) populations can promote lung inflammatory reactions and distress, and consequently acute and chronic respiratory diseases. This review is aimed to summarize the latest advances in resident lung microbiome and its participation in most common pulmonary infections and pneumonia, community-acquired pneumonia (CAP), ventilator-associated pneumonia (VAP), immunodeficiency associated pneumonia, SARS-CoV-2-associated pneumonia, acute respiratory distress syndrome (ARDS) and chronic obstructive pulmonary disease (COPD). We briefly describe physiological and immunological mechanisms that selectively create advantages or disadvantages for relative growth of pathogenic bacterial species in the respiratory tract. At the end, we propose some directions and analytical methods that may facilitate the identification of key genera and species of resident and transient microbes involved in the respiratory diseases' initiation and progression.
Collapse
Affiliation(s)
- José Belizário
- School of Arts, Sciences and Humanities of the University of Sao Paulo, Rua Arlindo Bettio, 1000, São Paulo, CEP 03828-000, Brazil
| | - Miguel Garay-Malpartida
- School of Arts, Sciences and Humanities of the University of Sao Paulo, Rua Arlindo Bettio, 1000, São Paulo, CEP 03828-000, Brazil
| | - Joel Faintuch
- Department of Gastroenterology of the Clinics Hospital of the University of São Paulo, Av. Dr. Enéas de Carvalho Aguiar, 255, São Paulo, CEP 05403-000, Brazil
| |
Collapse
|
15
|
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: 2] [Impact Index Per Article: 1.0] [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
|
16
|
Chen Z, Huang X, Lu H, Deng W, Huang L, Wu D, Wang D, Zhan Q, Wang C. The association between early fluid strategy and prognosis of acute respiratory distress syndrome: A post hoc study of CHARDS. Pulm Circ 2023; 13:e12261. [PMID: 37404902 PMCID: PMC10315445 DOI: 10.1002/pul2.12261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/12/2023] [Accepted: 06/20/2023] [Indexed: 07/06/2023] Open
Abstract
We aimed to assess general fluid management in China and evaluate the association between fluid balance and survival outcomes in acute respiratory distress syndrome (ARDS) patients. A retrospective, multicenter study including ARDS patients was conducted. We described the fluid management of ARDS patients in China. Furthermore, clinical characteristics and outcomes of patients subdivided by cumulative fluid balance were also analyzed. Multivariable logistic regression analysis was performed with hospital mortality as the outcome. From June 2016 to February 2018, 527 ARDS patients were included in our study. The mean cumulative fluid balance was 1669 (-1101 to 4351) mL in the first 7 day after intensive care unit (ICU) admission. Patients were divided into four groups based on cumulative fluid balance of the first 7 day after ICU admission: Group I (≤0 L), Group II (>0 L, ≤3 L), Group III (>3 L, ≤5 L), and Group IV (>5 L). Significantly lower hospital mortality was observed in patients with a lower cumulative fluid balance on day 7 of ICU admission (20.5% in Group I vs. 32.8% in Group II, 38.5% in Group III, and 50% in Group IV, p < 0.001). A lower fluid balance is associated with lower hospital mortality in patients with ARDS. However, a large-scale and well-designed randomized controlled trial is needed in the future.
Collapse
Affiliation(s)
- Ziying Chen
- Peking University China‐Japan Friendship School of Clinical MedicineBeijingChina
- Department of Pulmonary and Critical Care Medicine, China‐Japan Friendship Hospital, Center for Respiratory DiseasesNational Clinical Research Center for Respiratory DiseasesBeijingChina
| | - Xu Huang
- Department of Pulmonary and Critical Care Medicine, China‐Japan Friendship Hospital, Center for Respiratory DiseasesNational Clinical Research Center for Respiratory DiseasesBeijingChina
| | - Haining Lu
- Department of Critical Care MedicineQilu Hospital of Shandong University (Qingdao)QingdaoChina
| | - Wang Deng
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Linna Huang
- Department of Pulmonary and Critical Care Medicine, China‐Japan Friendship Hospital, Center for Respiratory DiseasesNational Clinical Research Center for Respiratory DiseasesBeijingChina
| | - Dawei Wu
- Department of Critical Care MedicineQilu Hospital of Shandong University (Qingdao)QingdaoChina
| | - Daoxin Wang
- Department of Respiratory and Critical Care MedicineThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Qingyuan Zhan
- Peking University China‐Japan Friendship School of Clinical MedicineBeijingChina
- Department of Pulmonary and Critical Care Medicine, China‐Japan Friendship Hospital, Center for Respiratory DiseasesNational Clinical Research Center for Respiratory DiseasesBeijingChina
| | - Chen Wang
- Peking University China‐Japan Friendship School of Clinical MedicineBeijingChina
- Department of Pulmonary and Critical Care Medicine, China‐Japan Friendship Hospital, Center for Respiratory DiseasesNational Clinical Research Center for Respiratory DiseasesBeijingChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
17
|
Jones TW, Almuntashiri S, Chase A, Alhumaid A, Somanath PR, Sikora A, Zhang D. Plasma matrix metalloproteinase-3 predicts mortality in acute respiratory distress syndrome: a biomarker analysis of a randomized controlled trial. Respir Res 2023; 24:166. [PMID: 37349704 PMCID: PMC10286483 DOI: 10.1186/s12931-023-02476-5] [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: 03/13/2023] [Accepted: 06/09/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND Matrix metalloproteinase-3 (MMP-3) is a proteolytic enzyme involved in acute respiratory distress syndrome (ARDS) pathophysiology that may serve as a lung-specific biomarker in ARDS. METHODS This study was a secondary biomarker analysis of a subset of Albuterol for the Treatment of Acute Lung Injury (ALTA) trial patients to determine the prognostic value of MMP-3. Plasma sample MMP-3 was measured by enzyme-linked immunosorbent assay. The primary outcome was the area under the receiver operating characteristic (AUROC) of MMP-3 at day 3 for the prediction of 90-day mortality. RESULTS A total of 100 unique patient samples were evaluated and the AUROC analysis of day three MMP-3 showed an AUROC of 0.77 for the prediction of 90-day mortality (95% confidence interval: 0.67-0.87), corresponding to a sensitivity of 92% and specificity of 63% and an optimal cutoff value of 18.4 ng/mL. Patients in the high MMP-3 group (≥ 18.4 ng/mL) showed higher mortality compared to the non-elevated MMP-3 group (< 18.4 ng/mL) (47% vs. 4%, p < 0.001). A positive difference in day zero and day three MMP-3 concentration was predictive of mortality with an AUROC of 0.74 correlating to 73% sensitivity, 81% specificity, and an optimal cutoff value of + 9.5 ng/mL. CONCLUSIONS Day three MMP-3 concentration and difference in day zero and three MMP-3 concentrations demonstrated acceptable AUROCs for predicting 90-day mortality with a cut-point of 18.4 ng/mL and + 9.5 ng/mL, respectively. These results suggest a prognostic role of MMP-3 in ARDS.
Collapse
Affiliation(s)
- Timothy W. Jones
- Department of Pharmacy, Augusta University Medical Center, 1120 15th St., Augusta, GA 30912 USA
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| | - Sultan Almuntashiri
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| | - Aaron Chase
- Department of Pharmacy, Augusta University Medical Center, 1120 15th St., Augusta, GA 30912 USA
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| | - Abdullah Alhumaid
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| | - Payaningal R. Somanath
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| | - Andrea Sikora
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| | - Duo Zhang
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, 120 15th Street, HM-117, Augusta, GA 30912 USA
| |
Collapse
|
18
|
Suarez-Pajes E, Tosco-Herrera E, Ramirez-Falcon M, Gonzalez-Barbuzano S, Hernandez-Beeftink T, Guillen-Guio B, Villar J, Flores C. Genetic Determinants of the Acute Respiratory Distress Syndrome. J Clin Med 2023; 12:3713. [PMID: 37297908 PMCID: PMC10253474 DOI: 10.3390/jcm12113713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/18/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening lung condition that arises from multiple causes, including sepsis, pneumonia, trauma, and severe coronavirus disease 2019 (COVID-19). Given the heterogeneity of causes and the lack of specific therapeutic options, it is crucial to understand the genetic and molecular mechanisms that underlie this condition. The identification of genetic risks and pharmacogenetic loci, which are involved in determining drug responses, could help enhance early patient diagnosis, assist in risk stratification of patients, and reveal novel targets for pharmacological interventions, including possibilities for drug repositioning. Here, we highlight the basis and importance of the most common genetic approaches to understanding the pathogenesis of ARDS and its critical triggers. We summarize the findings of screening common genetic variation via genome-wide association studies and analyses based on other approaches, such as polygenic risk scores, multi-trait analyses, or Mendelian randomization studies. We also provide an overview of results from rare genetic variation studies using Next-Generation Sequencing techniques and their links with inborn errors of immunity. Lastly, we discuss the genetic overlap between severe COVID-19 and ARDS by other causes.
Collapse
Affiliation(s)
- Eva Suarez-Pajes
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
| | - Eva Tosco-Herrera
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
| | - Melody Ramirez-Falcon
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
| | - Silvia Gonzalez-Barbuzano
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
| | - Tamara Hernandez-Beeftink
- Department of Population Health Sciences, University of Leicester, Leicester LE1 7RH, UK
- NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester LE1 7RH, UK
| | - Beatriz Guillen-Guio
- Department of Population Health Sciences, University of Leicester, Leicester LE1 7RH, UK
- NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester LE1 7RH, UK
| | - Jesús Villar
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Research Unit, Hospital Universitario de Gran Canaria Dr. Negrín, 35019 Las Palmas de Gran Canaria, Spain
| | - Carlos Flores
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Genomics Division, Instituto Tecnológico y de Energías Renovables (ITER), 38600 Santa Cruz de Tenerife, Spain
- Faculty of Health Sciences, University of Fernando Pessoa Canarias, 35450 Las Palmas de Gran Canaria, Spain
| |
Collapse
|
19
|
Ramji HF, Hafiz M, Altaq HH, Hussain ST, Chaudry F. Acute Respiratory Distress Syndrome; A Review of Recent Updates and a Glance into the Future. Diagnostics (Basel) 2023; 13:diagnostics13091528. [PMID: 37174920 PMCID: PMC10177247 DOI: 10.3390/diagnostics13091528] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/14/2023] [Accepted: 04/15/2023] [Indexed: 05/15/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a rapidly progressive form of respiratory failure that accounts for 10% of admissions to the ICU and is associated with approximately 40% mortality in severe cases. Despite significant mortality and healthcare burden, the mainstay of management remains supportive care. The recent pandemic of SARS-CoV-2 has re-ignited a worldwide interest in exploring the pathophysiology of ARDS, looking for innovative ideas to treat this disease. Recently, many trials have been published utilizing different pharmacotherapy targets; however, the long-term benefits of these agents remain unknown. Metabolomics profiling and stem cell transplantation offer strong enthusiasm and may completely change the outlook of ARDS management in the near future.
Collapse
Affiliation(s)
- Husayn F Ramji
- University of Oklahoma College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Hudson College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Maida Hafiz
- Department of Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Hiba Hammad Altaq
- Department of Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Syed Talal Hussain
- Department of Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Fawad Chaudry
- Department of Pulmonary, Critical Care & Sleep Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
20
|
Rizzo AN, Schmidt EP. The role of the alveolar epithelial glycocalyx in acute respiratory distress syndrome. Am J Physiol Cell Physiol 2023; 324:C799-C806. [PMID: 36847444 PMCID: PMC10042597 DOI: 10.1152/ajpcell.00555.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 03/01/2023]
Abstract
The alveolar epithelial glycocalyx is a dense anionic layer of glycosaminoglycans (GAGs) and proteoglycans that lines the apical surface of the alveolar epithelium. In contrast to the pulmonary endothelial glycocalyx, which has well-established roles in vascular homeostasis and septic organ dysfunction, the alveolar epithelial glycocalyx is less understood. Recent preclinical studies demonstrated that the epithelial glycocalyx is degraded in multiple murine models of acute respiratory distress syndrome (ARDS), particularly those that result from inhaled insults (so-called "direct" lung injury), leading to shedding of GAGs into the alveolar airspaces. Epithelial glycocalyx degradation also occurs in humans with respiratory failure, as quantified by analysis of airspace fluid obtained from ventilator heat moisture exchange (HME) filters. In patients with ARDS, GAG shedding correlates with the severity of hypoxemia and is predictive of the duration of respiratory failure. These effects may be mediated by surfactant dysfunction, as targeted degradation of the epithelial glycocalyx in mice was sufficient to cause increased alveolar surface tension, diffuse microatelectasis, and impaired lung compliance. In this review, we describe the structure of the alveolar epithelial glycocalyx and the mechanisms underlying its degradation during ARDS. We additionally review the current state of knowledge regarding the attributable effect of epithelial glycocalyx degradation in lung injury pathogenesis. Finally, we address glycocalyx degradation as a potential mediator of ARDS heterogeneity, and the subsequent value of point-of-care quantification of GAG shedding to potentially identify patients who are most likely to respond to pharmacological agents aimed at attenuating glycocalyx degradation.
Collapse
Affiliation(s)
- Alicia N Rizzo
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Eric P Schmidt
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
21
|
Yang Z, Nicholson SE, Cancio TS, Cancio LC, Li Y. Complement as a vital nexus of the pathobiological connectome for acute respiratory distress syndrome: An emerging therapeutic target. Front Immunol 2023; 14:1100461. [PMID: 37006238 PMCID: PMC10064147 DOI: 10.3389/fimmu.2023.1100461] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/27/2023] [Indexed: 03/19/2023] Open
Abstract
The hallmark of acute respiratory distress syndrome (ARDS) pathobiology is unchecked inflammation-driven diffuse alveolar damage and alveolar-capillary barrier dysfunction. Currently, therapeutic interventions for ARDS remain largely limited to pulmonary-supportive strategies, and there is an unmet demand for pharmacologic therapies targeting the underlying pathology of ARDS in patients suffering from the illness. The complement cascade (ComC) plays an integral role in the regulation of both innate and adaptive immune responses. ComC activation can prime an overzealous cytokine storm and tissue/organ damage. The ARDS and acute lung injury (ALI) have an established relationship with early maladaptive ComC activation. In this review, we have collected evidence from the current studies linking ALI/ARDS with ComC dysregulation, focusing on elucidating the new emerging roles of the extracellular (canonical) and intracellular (non-canonical or complosome), ComC (complementome) in ALI/ARDS pathobiology, and highlighting complementome as a vital nexus of the pathobiological connectome for ALI/ARDS via its crosstalking with other systems of the immunome, DAMPome, PAMPome, coagulome, metabolome, and microbiome. We have also discussed the diagnostic/therapeutic potential and future direction of ALI/ARDS care with the ultimate goal of better defining mechanistic subtypes (endotypes and theratypes) through new methodologies in order to facilitate a more precise and effective complement-targeted therapy for treating these comorbidities. This information leads to support for a therapeutic anti-inflammatory strategy by targeting the ComC, where the arsenal of clinical-stage complement-specific drugs is available, especially for patients with ALI/ARDS due to COVID-19.
Collapse
Affiliation(s)
- Zhangsheng Yang
- Combat Casualty Care Research Team (CRT) 3, United States (US) Army Institute of Surgical Research, Joint Base San Antonio (JBSA)-Fort Sam Houston, TX, United States
| | - Susannah E. Nicholson
- Division of Trauma Research, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Tomas S. Cancio
- Combat Casualty Care Research Team (CRT) 3, United States (US) Army Institute of Surgical Research, Joint Base San Antonio (JBSA)-Fort Sam Houston, TX, United States
| | - Leopoldo C. Cancio
- United States (US) Army Burn Center, United States (US) Army Institute of Surgical Research, Joint Base San Antonio (JBSA)-Fort Sam Houston, TX, United States
| | - Yansong Li
- Division of Trauma Research, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
- The Geneva Foundation, Immunological Damage Control Resuscitation Program, Tacoma, WA, United States
- *Correspondence: Yansong Li,
| |
Collapse
|
22
|
Johnsson P, Fredriksson A, Tung C, Friberg H, Frigyesi A. Plasma bioactive adrenomedullin on intensive care unit admission is associated with acute respiratory distress syndrome: an observational study. Intensive Care Med Exp 2023; 11:10. [PMID: 36864354 PMCID: PMC9981258 DOI: 10.1186/s40635-023-00494-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/21/2023] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND Bioactive adrenomedullin (bio-ADM) is a vasoactive peptide with a key role in reducing vascular hyperpermeability and improving endothelial stability during infection, but it also has vasodilatory properties. Bioactive ADM has not been studied in conjunction with acute respiratory distress syndrome (ARDS), but it has recently been shown to correlate with outcomes after severe COVID-19. Therefore, this study investigated the association between circulating bio-ADM on intensive care unit (ICU) admission and ARDS. The secondary aim was the association between bio-ADM and ARDS mortality. METHODS We analysed bio-ADM levels and assessed the presence of ARDS in adult patients admitted to two general intensive care units in southern Sweden. Medical records were manually screened for the ARDS Berlin criteria. The association between bio-ADM levels and ARDS and mortality in ARDS patients was analysed using logistic regression and receiver-operating characteristics analysis. The primary outcome was an ARDS diagnosis within 72 h of ICU admission, and the secondary outcome was 30-day mortality. RESULTS Out of 1224 admissions, 11% (n = 132) developed ARDS within 72 h. We found that elevated admission bio-ADM level was associated with ARDS independently of sepsis status and of organ dysfunction as measured by the Sequential organ failure assessment (SOFA) score. Both lower levels (< 38 pg/L) and high (> 90 pg/L) levels of bio-ADM were independently (of the Simplified acute physiology score, SAPS-3) predictive of mortality. Patients with indirect mechanisms of lung injury had higher bio-ADM levels than those with a direct mechanism of injury, and bio-ADM increased with increasing ARDS severity. CONCLUSIONS High levels of bio-ADM on admission are associated with ARDS, and bio-ADM levels significantly differ depending on the injury mechanism. In contrast, both high and low levels of bio-ADM are associated with mortality, possibly due to the dual action of bio-ADM in stabilising the endothelial barrier and causing vasodilation. These findings could lead to improved diagnostic accuracy of ARDS and potentially lead to new therapeutic strategies.
Collapse
Affiliation(s)
- Patrik Johnsson
- Department of Clinical Medicine, Anaesthesiology and Intensive Care, Lund University, 22185, Lund, Sweden. .,Department of Intensive and Perioperative Care, Skåne University Hospital, 20502, Malmo, Sweden.
| | - Andrea Fredriksson
- grid.4514.40000 0001 0930 2361Department of Clinical Medicine, Anaesthesiology and Intensive Care, Lund University, 22185 Lund, Sweden ,grid.411843.b0000 0004 0623 9987Department of Intensive and Perioperative Care, Skåne University Hospital, 20502 Malmo, Sweden
| | - Christer Tung
- grid.4514.40000 0001 0930 2361Department of Clinical Medicine, Anaesthesiology and Intensive Care, Lund University, 22185 Lund, Sweden ,grid.411843.b0000 0004 0623 9987Department of Intensive and Perioperative Care, Skåne University Hospital, 20502 Malmo, Sweden
| | - Hans Friberg
- grid.4514.40000 0001 0930 2361Department of Clinical Medicine, Anaesthesiology and Intensive Care, Lund University, 22185 Lund, Sweden ,grid.411843.b0000 0004 0623 9987Department of Intensive and Perioperative Care, Skåne University Hospital, 20502 Malmo, Sweden
| | - Attila Frigyesi
- grid.4514.40000 0001 0930 2361Department of Clinical Medicine, Anaesthesiology and Intensive Care, Lund University, 22185 Lund, Sweden ,grid.411843.b0000 0004 0623 9987Department of Intensive and Perioperative Care, Skåne University Hospital, 20502 Malmo, Sweden
| |
Collapse
|
23
|
Rizzo AN, Aggarwal NR, Thompson BT, Schmidt EP. Advancing Precision Medicine for the Diagnosis and Treatment of Acute Respiratory Distress Syndrome. J Clin Med 2023; 12:1563. [PMID: 36836098 PMCID: PMC9966442 DOI: 10.3390/jcm12041563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common and life-threatening cause of respiratory failure. Despite decades of research, there are no effective pharmacologic therapies to treat this disease process and mortality remains high. The shortcomings of prior translational research efforts have been increasingly attributed to the heterogeneity of this complex syndrome, which has led to an increased focus on elucidating the mechanisms underlying the interpersonal heterogeneity of ARDS. This shift in focus aims to move the field towards personalized medicine by defining subgroups of ARDS patients with distinct biology, termed endotypes, to quickly identify patients that are most likely to benefit from mechanism targeted treatments. In this review, we first provide a historical perspective and review the key clinical trials that have advanced ARDS treatment. We then review the key challenges that exist with regards to the identification of treatable traits and the implementation of personalized medicine approaches in ARDS. Lastly, we discuss potential strategies and recommendations for future research that we believe will aid in both understanding the molecular pathogenesis of ARDS and the development of personalized treatment approaches.
Collapse
Affiliation(s)
- Alicia N. Rizzo
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02144, USA
| | - Neil R. Aggarwal
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - B. Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02144, USA
| | - Eric P. Schmidt
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02144, USA
| |
Collapse
|
24
|
Molecular imaging of chemokine-like receptor 1 (CMKLR1) in experimental acute lung injury. Proc Natl Acad Sci U S A 2023; 120:e2216458120. [PMID: 36626557 PMCID: PMC9934297 DOI: 10.1073/pnas.2216458120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The lack of techniques for noninvasive imaging of inflammation has challenged precision medicine management of acute respiratory distress syndrome (ARDS). Here, we determined the potential of positron emission tomography (PET) of chemokine-like receptor-1 (CMKLR1) to monitor lung inflammation in a murine model of lipopolysaccharide-induced injury. Lung uptake of a CMKLR1-targeting radiotracer, [64Cu]NODAGA-CG34, was significantly increased in lipopolysaccharide-induced injury, correlated with the expression of multiple inflammatory markers, and reduced by dexamethasone treatment. Monocyte-derived macrophages, followed by interstitial macrophages and monocytes were the major CMKLR1-expressing leukocytes contributing to the increased tracer uptake throughout the first week of lipopolysaccharide-induced injury. The clinical relevance of CMKLR1 as a biomarker of lung inflammation in ARDS was confirmed using single-nuclei RNA-sequencing datasets which showed significant increases in CMKLR1 expression among transcriptionally distinct subsets of lung monocytes and macrophages in COVID-19 patients vs. controls. CMKLR1-targeted PET is a promising strategy to monitor the dynamics of lung inflammation and response to anti-inflammatory treatment in ARDS.
Collapse
|
25
|
Dexmedetomidine alleviates acute lung injury by promoting Tregs differentiation via activation of AMPK/SIRT1 pathway. Inflammopharmacology 2023; 31:423-438. [PMID: 36534240 PMCID: PMC9762669 DOI: 10.1007/s10787-022-01117-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022]
Abstract
OBJECTIVES To explore the anti-inflammatory effect and the potential mechanism of dexmedetomidine in ARDS/ALI. MATERIALS AND METHODS C57BL/6 mice and EL-4 cells were used in this research. The ALI model was established by CLP. The level of inflammatory cytokines in the lung and blood, the severity of lung injury, the expression of Foxp3, and the proportion of Tregs were detected before and after dexmedetomidine treatment. The expression of the AMPK/SIRT1 after dexmedetomidine treatment was detected in vivo and in vitro. After blocking the AMPK/SIRT1 pathway or depleting Tregs in vivo, the level of the inflammatory response, tissue injury, and Tregs differentiation were detected again to clarify the effect of dexmedetomidine. RESULTS Dexmedetomidine significantly reduced systemic inflammation and lung injury in CLP mice. Dexmedetomidine enhanced the Foxp3 expression in the lungs and the frequency of Tregs in the spleen. Dexmedetomidine up-regulated the protein expression of p-AMPK and SIRT1 in lungs and EL-4 cells and facilitated the differentiation of naïve CD4+ T cells into Tregs in vitro. Meanwhile, DEX also increased the expression of Helios in Treg cells. CONCLUSIONS DEX could improve ARDS/ALI by facilitating the differentiation of Tregs from naïve CD4+ T cells via activating the AMPK/SIRT1 pathway.
Collapse
|
26
|
Wu L, Lei Q, Gao Z, Zhang W. Research Progress on Phenotypic Classification of Acute Respiratory Distress Syndrome: A Narrative Review. Int J Gen Med 2022; 15:8767-8774. [PMID: 36601648 PMCID: PMC9807128 DOI: 10.2147/ijgm.s391969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/15/2022] [Indexed: 12/30/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a clinical syndrome that is characterized by an acute onset and refractory hypoxemia. It remains an important contributor to high mortality in critically ill patients, and the majority of clinical randomized controlled trials on ARDS provide underwhelming findings, which is attributed in large part to its pathophysiological and clinical heterogeneity, among other aspects. It is now widely accepted that ARDS is highly heterogeneous, growing evidences support this. ARDS phenotypic and subphenotypic studies aim to further differentiate and identify ARDS heterogeneity in the hope that clinicians can benefit from it, then can diagnose ARDS faster and more accurately and provide targeted treatments. This review collates and evaluates the major phenotype-related research advances of recent years, with a specific focus on ARDS biomarkers and clinical factors.
Collapse
Affiliation(s)
- Linlin Wu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Qian Lei
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Zirong Gao
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Wei Zhang
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China,Correspondence: Wei Zhang, Email
| |
Collapse
|
27
|
Liu L, Yuan Y, Zhou Y, Yao L, Li J, Chen F. The transcription factor KLF14 attenuates LPS-induced acute lung injury by ameliorating apoptosis of alveolar epithelial cells in mice. Mol Immunol 2022; 152:67-77. [DOI: 10.1016/j.molimm.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 09/25/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
|
28
|
Tiboldi A, Hunyadi-Gulyas E, Wohlrab P, Schmid JA, Markstaller K, Klein KU, Tretter V. Effects of Hyperoxia and Hyperoxic Oscillations on the Proteome of Murine Lung Microvascular Endothelium. Antioxidants (Basel) 2022; 11:2349. [PMID: 36552557 PMCID: PMC9774699 DOI: 10.3390/antiox11122349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/20/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
Patients presenting with insufficient tissue oxygenation and impaired lung function as in acute respiratory distress syndrome (ARDS) frequently require mechanical ventilation with supplemental oxygen. Despite the lung being used to experiencing the highest partial pressure of oxygen during healthy breathing, the organ is susceptible to oxygen-induced injury at supraphysiological concentrations. Hyperoxia-induced lung injury (HALI) has been regarded as a second hit to pre-existing lung injury and ventilator-induced lung injury (VILI) attributed to oxidative stress. The injured lung has a tendency to form atelectasis, a cyclic collapse and reopening of alveoli. The affected lung areas experience oxygen conditions that oscillate between hyperoxia and hypoxia rather than remaining in a constant hyperoxic state. Mechanisms of HALI have been investigated in many animal models previously. These studies provided insights into the effects of hyperoxia on the whole organism. However, cell type-specific responses have not been dissected in detail, but are necessary for a complete mechanistic understanding of ongoing pathological processes. In our study, we investigated the effects of constant and intermittent hyperoxia on the lung endothelium from a mouse by an in vitro proteomic approach. We demonstrate that these oxygen conditions have characteristic effects on the pulmonary endothelial proteome that underlie the physiological (patho)mechanisms.
Collapse
Affiliation(s)
- Akos Tiboldi
- Department of Anesthesia, General Intensive Care and Pain Management, Medical University of Vienna, 1090 Vienna, Austria
| | - Eva Hunyadi-Gulyas
- Laboratory of Proteomics Research, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Temesvári krt. 62, H-6726 Szeged, Hungary
| | - Peter Wohlrab
- Department of Anesthesia, General Intensive Care and Pain Management, Medical University of Vienna, 1090 Vienna, Austria
| | - Johannes A. Schmid
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University Vienna, Schwarzspanierstraße 17, 1090 Vienna, Austria
| | - Klaus Markstaller
- Department of Anesthesia, General Intensive Care and Pain Management, Medical University of Vienna, 1090 Vienna, Austria
| | - Klaus Ulrich Klein
- Department of Anesthesia, General Intensive Care and Pain Management, Medical University of Vienna, 1090 Vienna, Austria
| | - Verena Tretter
- Department of Anesthesia, General Intensive Care and Pain Management, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
29
|
Martos-Benítez FD, Estévez-Muguercia R, Orama-Requejo V, Del Toro-Simoni T. Prognostic value of the novel P/FPE index to classify ARDS severity: A cohort study. Med Intensiva 2022:S2173-5727(22)00309-5. [PMID: 36344340 DOI: 10.1016/j.medine.2022.06.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/07/2022] [Indexed: 06/16/2023]
Abstract
OBJECTIVE To evaluate the impact of the novel P/FPE index to classify ARDS severity on mortality of patients with ARDS. DESIGN A retrospective cohort study. SETTING Twelve-bed medical and surgical intensive care unit from January 2018 to December 2020. PATIENTS A total of 217 ARDS patients managed with invasive mechanical ventilation >48h. INTERVENTIONS None. VARIABLES ARDS severity on day 1 and day 3 was measured based on PaO2/FiO2 ratio and P/FPE index [PaO2/(FiO2×PEEP)]. Primary outcome was the hospital mortality. RESULTS Hospital mortality rate was 59.9%. Relative to PaO2/FiO2 ratio, 31.8% of patients on day 1 and 77.0% on day 3 were reclassified into a different category of ARDS severity by P/FPE index. The level of PEEP was lower by P/FPE index-based ARDS severity classification than by using PaO2/FiO2 ratio. The performance for predicting mortality of P/FPE index was superior to PaO2/FiO2 ratio in term of AROC (day 1: 0.72 vs. 0.62; day 3: 0.87 vs. 0.68) and CORR (day 1: 0.370 vs. 0.213; day 3: 0.634 vs. 0.301). P/FPE index improved prediction of risk of death compared to PaO2/FiO2 ratio as showed by the qNRI (day 1: 72.0%, p<0.0001; day 3: 132.4%, p<0.0001) and IDI (day 1: 0.09, p<0.0001; day 3: 0.31, p<0.0001). CONCLUSIONS Assessment of ARDS severity based on P/FPE index seems better than PaO2/FiO2 ratio for predicting mortality. The value of P/FPE index for clinical decision-making requires confirmation by randomized controlled trials.
Collapse
Affiliation(s)
- F D Martos-Benítez
- Intensive Care Unit - 8, Hermanos Ameijeiras Hospital, Havana 10400, Cuba.
| | | | - V Orama-Requejo
- Intermediate Care Unit, Hospital of Palamos, Palamos 17230, Spain
| | - T Del Toro-Simoni
- Intensive Care Unit, Manuel Ascunce Domenech Hospital, Camagüey 70600, Cuba
| |
Collapse
|
30
|
Serial Measurements of Protein Biomarkers in Sepsis-Induced Acute Respiratory Distress Syndrome. Crit Care Explor 2022; 4:e0780. [PMID: 36284549 PMCID: PMC9586925 DOI: 10.1097/cce.0000000000000780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The role of early, serial measurements of protein biomarkers in sepsis-induced acute respiratory distress syndrome (ARDS) is not clear. OBJECTIVES To determine the differences in soluble receptor for advanced glycation end-products (sRAGEs), angiopoietin-2, and surfactant protein-D (SP-D) levels and their changes over time between sepsis patients with and without ARDS. DESIGN SETTING AND PARTICIPANTS Prospective observational cohort study of adult patients admitted to the medical ICU at Grady Memorial Hospital within 72 hours of sepsis diagnosis. MAIN OUTCOMES AND MEASURES Plasma sRAGE, angiopoietin-2, and SP-D levels were measured for 3 consecutive days after enrollment. The primary outcome was ARDS development, and the secondary outcome of 28-day mortality. The biomarker levels and their changes over time were compared between ARDS and non-ARDS patients and between nonsurvivors and survivors. RESULTS We enrolled 111 patients, and 21 patients (18.9%) developed ARDS. The three biomarker levels were not significantly different between ARDS and non-ARDS patients on all 3 days of measurement. Nonsurvivors had higher levels of all three biomarkers than did survivors on multiple days. The changes of the biomarker levels over time were not different between the outcome groups. Logistic regression analyses showed association between day 1 SP-D level and mortality (odds ratio, 1.52; 95% CI, 1.03-2.24; p = 0.03), and generalized estimating equation analyses showed association between angiopoietin-2 levels and mortality (estimate 0.0002; se 0.0001; p = 0.04). CONCLUSIONS AND RELEVANCE Among critically ill patients with sepsis, sRAGE, angiopoietin-2, and SP-D levels were not significantly different between ARDS and non-ARDS patients but were higher in nonsurvivors compared with survivors. The trend toward higher levels of sRAGE and SP-D, but not of angiopoietin-2, in ARDS patients may indicate the importance of epithelial injury in sepsis-induced ARDS. Changes of the biomarker levels over time were not different between the outcome groups.
Collapse
|
31
|
Wick KD, Aggarwal NR, Curley MAQ, Fowler AA, Jaber S, Kostrubiec M, Lassau N, Laterre PF, Lebreton G, Levitt JE, Mebazaa A, Rubin E, Sinha P, Ware LB, Matthay MA. Opportunities for improved clinical trial designs in acute respiratory distress syndrome. THE LANCET. RESPIRATORY MEDICINE 2022; 10:916-924. [PMID: 36057279 DOI: 10.1016/s2213-2600(22)00294-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 07/02/2022] [Accepted: 07/19/2022] [Indexed: 02/08/2023]
Abstract
The acute respiratory distress syndrome (ARDS) is a common critical illness syndrome with high morbidity and mortality. There are no proven pharmacological therapies for ARDS. The current definition of ARDS is based on shared clinical characteristics but does not capture the heterogeneity in clinical risk factors, imaging characteristics, physiology, timing of onset and trajectory, and biology of the syndrome. There is increasing interest within the ARDS clinical trialist community to design clinical trials that reduce heterogeneity in the trial population. This effort must be balanced with ongoing work to craft an inclusive, global definition of ARDS, with important implications for trial design. Ultimately, the two aims-to design trials that are applicable to the diverse global ARDS population while also advancing opportunities to identify targetable traits-should coexist. In this Personal View, we recommend two primary strategies to improve future ARDS trials: the development of new methods to target treatable traits in clinical trial populations, and improvements in the representativeness of ARDS trials, with the inclusion of global populations. We emphasise that these two strategies are complementary. We also discuss how a proposed expansion of the definition of ARDS could affect the future of clinical trials.
Collapse
Affiliation(s)
- Katherine D Wick
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Neil R Aggarwal
- Division of Pulmonary Sciences and Critical Care, Department of Medicine, University of Colorado, Aurora, CO, USA; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Martha A Q Curley
- Department of Family and Community Health, School of Nursing, University of Pennsylvania, Philadelphia, PA, USA
| | - Alpha A Fowler
- Division of Pulmonary Disease and Critical Care, Virginia Commonwealth University, Richmond, VA, USA
| | - Samir Jaber
- University Hospital, CHU de Montpellier Hôpital Saint Eloi, Intensive Care Unit and Transplantation, Department of Anesthesiology DAR B, Montpellier, France
| | - Maciej Kostrubiec
- Department of Internal Medicine and Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Nathalie Lassau
- Department of Imaging, Gustave Roussy, Université Paris Saclay, Villejuif, France; Biomaps, UMR1281 INSERM, CEA, CNRS, Université Paris Saclay, Villejuif, France
| | - Pierre François Laterre
- Intensive Care Medicine, Saint-Luc University Hospital, Université Catholique de Louvain, Brussels, Belgium
| | - Guillaume Lebreton
- Institute of Cardiometabolism and Nutrition, Inserm, UMRS 1166-ICAN, Sorbonne University, Paris, France; Cardiac Surgery Service, Institute of Cardiology, AP-HP, Sorbonne University, Paris, France
| | - Joseph E Levitt
- Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Alexandre Mebazaa
- Department of Anesthesiology and Critical Care Medicine, AP-HP, Saint Louis and Lariboisière University Hospitals, Paris, France
| | | | - Pratik Sinha
- Department of Anesthesiology, Washington University in St Louis, St Louis, MO, USA
| | - Lorraine B Ware
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael A Matthay
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA; Departments of Medicine and Anesthesia, University of California, San Francisco, CA, USA.
| |
Collapse
|
32
|
Battaglini D, Robba C, Pelosi P, Rocco PRM. Treatment for acute respiratory distress syndrome in adults: A narrative review of phase 2 and 3 trials. Expert Opin Emerg Drugs 2022; 27:187-209. [PMID: 35868654 DOI: 10.1080/14728214.2022.2105833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Ventilatory management and general supportive care of acute respiratory distress syndrome (ARDS) in the adult population have led to significant clinical improvements, but morbidity and mortality remain high. Pharmacologic strategies acting on the coagulation cascade, inflammation, oxidative stress, and endothelial cell injury have been targeted in the last decade for patients with ARDS, but only a few of these have shown potential benefits with a meaningful clinical response and improved patient outcomes. The lack of availability of specific pharmacologic treatments for ARDS can be attributed to its complex pathophysiology, different risk factors, huge heterogeneity, and difficult classification into specific biological phenotypes and genotypes. AREAS COVERED In this narrative review, we briefly discuss the relevance and current advances in pharmacologic treatments for ARDS in adults and the need for the development of new pharmacological strategies. EXPERT OPINION Identification of ARDS phenotypes, risk factors, heterogeneity, and pathophysiology may help to design clinical trials personalized according to ARDS-specific features, thus hopefully decreasing the rate of failed clinical pharmacologic trials. This concept is still under clinical investigation and needs further development.
Collapse
Affiliation(s)
- Denise Battaglini
- Dipartimento di Anestesia e Rianimazione, Policlinico San Martino, IRCCS per l'Oncologia e le Neuroscienze, Largo Rosanna Benzi, 10, 16132, Genoa, Italy
| | - Chiara Robba
- Dipartimento di Anestesia e Rianimazione, Policlinico San Martino, IRCCS per l'Oncologia e le Neuroscienze, Largo Rosanna Benzi, 10, 16132, Genoa, Italy.,Dipartimento di Scienze Chirurgiche e Diagnostiche Integrate, Università degli Studi di Genova, Largo Rosanna Benzi, 10, 16132, Genoa, Italy
| | - Paolo Pelosi
- Dipartimento di Anestesia e Rianimazione, Policlinico San Martino, IRCCS per l'Oncologia e le Neuroscienze, Largo Rosanna Benzi, 10, 16132, Genoa, Italy.,Dipartimento di Scienze Chirurgiche e Diagnostiche Integrate, Università degli Studi di Genova, Largo Rosanna Benzi, 10, 16132, Genoa, Italy
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G1-014, Ilha do Fundão, Rio de Janeiro, RJ 21941-902, Brazil.,COVID-19 Virus Network from Ministry of Science, Technology, and Innovation, Brazilian Council for Scientific and Technological Development, and Foundation Carlos Chagas Filho Research Support of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
33
|
Martos-Benítez F, Estévez-Muguercia R, Orama-Requejo V, del Toro-Simoni T. Prognostic value of the novel P/FPE index to classify ARDS severity: A cohort study. Med Intensiva 2022. [DOI: 10.1016/j.medin.2022.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
34
|
Aschner Y, Correll KA, Beke K, Foster DG, Roybal HM, Nelson MR, Meador CL, Strand M, Anderson KC, Moore CM, Reynolds PR, Kopf KW, Burnham EL, Downey GP. PTPα Promotes Fibroproliferative Responses After Acute Lung Injury. Am J Physiol Lung Cell Mol Physiol 2022; 323:L69-L83. [PMID: 35670474 DOI: 10.1152/ajplung.00436.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The Acute Respiratory Distress Syndrome (ARDS) is a major healthcare problem, accounting for significant mortality and long-term disability. Approximately 25% of patients with ARDS will develop an over-exuberant fibrotic response, termed fibroproliferative ARDS (FP-ARDS) that portends a poor prognosis and increased mortality. The cellular pathologic processes that drive FP-ARDS remain incompletely understood. We have previously shown that the transmembrane receptor-type tyrosine phosphatase Protein Tyrosine Phosphatase-a (PTPa) promotes pulmonary fibrosis in preclinical murine models through regulation of TGF-b signaling. In this study, we examine the role of PTPa in the pathogenesis of FP-ARDS in a preclinical murine model of acid (HCl)-induced acute lung injury. We demonstrate that while mice genetically deficient in PTPa (Ptpra-/-) are susceptible to early HCl-induced lung injury, they exhibit markedly attenuated fibroproliferative responses. Additionally, early pro-fibrotic gene expression is reduced in lung tissue after acute lung injury in Ptpra-/- mice, and stimulation of naïve lung fibroblasts with the BAL fluid from these mice results in attenuated fibrotic outcomes compared to wild type littermate controls. Transcriptomic analyses demonstrates reduced Extracellular Matrix (ECM) deposition and remodeling in mice genetically deficient in PTPa. Importantly, human lung fibroblasts modified with a CRISPR-targeted deletion of PTPRA exhibit reduced expression of profibrotic genes in response to TGF-β stimulation, demonstrating the importance of PTPa in human lung fibroblasts. Together, these findings demonstrate that PTPa is a key regulator of fibroproliferative processes following acute lung injury and could serve as a therapeutic target for patients at risk for poor long-term outcomes in ARDS.
Collapse
Affiliation(s)
- Yael Aschner
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, CO, United States.,Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Kelly A Correll
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Keriann Beke
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Daniel G Foster
- Department of Medicine, National Jewish Health, Denver, CO, United States.,Department of Pediatrics, National Jewish Health, Denver, CO, United States
| | - Helen M Roybal
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Meghan R Nelson
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Carly L Meador
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Matthew Strand
- Division of Biostatistics, National Jewish Health, Denver, CO, United States
| | - Kelsey C Anderson
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, United States
| | - Camille M Moore
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO, United States.,Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, United States
| | - Paul R Reynolds
- Department of Medicine, National Jewish Health, Denver, CO, United States.,Department of Pediatrics, National Jewish Health, Denver, CO, United States
| | - Katrina W Kopf
- Office of Academic Affairs, National Jewish Health, Denver, CO, United States
| | - Ellen L Burnham
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, CO, United States
| | - Gregory P Downey
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, CO, United States.,Department of Medicine, National Jewish Health, Denver, CO, United States.,Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO, United States.,Department of Pediatrics, National Jewish Health, Denver, CO, United States.,Office of Academic Affairs, National Jewish Health, Denver, CO, United States.,Department of Immunology and Microbiology, University of Colorado, Aurora, CO, United States
| |
Collapse
|
35
|
Nirvik P, Kertai MD. Future of Perioperative Precision Medicine: Integration of Molecular Science, Dynamic Health Care Informatics, and Implementation of Predictive Pathways in Real Time. Anesth Analg 2022; 134:900-908. [PMID: 35320133 DOI: 10.1213/ane.0000000000005966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Conceptually, precision medicine is a deep dive to discover disease origin at the molecular or genetic level, thus providing insights that allow clinicians to design corresponding individualized patient therapies. We know that a disease state is created by not only certain molecular derangements but also a biologic milieu promoting the expression of such derangements. These factors together lead to manifested symptoms. At the level of molecular definition, every average, "similar" individual stands to be "dissimilar." Hence, there is the need for customized therapy, moving away from therapy based on aggregate statistics. The perioperative state is a mix of several, simultaneously active molecular mechanisms, surgical insult, drugs, severe inflammatory response, and the body's continuous adaptation to maintain a state of homeostasis. Postoperative outcomes are a net result of several of those rapid genetic and molecular transformations that do or do not ensue. With the advent and advances of artificial intelligence, the translation from identifying these intricate mechanisms to implementing them in clinical practice has made a huge leap. Precision medicine is gaining ground with the help of personalized health recorders and personal devices that identify disease mechanics, patient-reported outcomes, adverse drug reactions, and drug-drug interaction at the individual level in a closed-loop feedback system. This phenomenon is especially true given increasing surgeries in older adults, many of whom are on multiple medications and varyingly frail. In this era of precision medicine, to provide a comprehensive remedy, the perioperative surgical home must expand, incorporating not only clinicians but also basic science experts and data scientists.
Collapse
Affiliation(s)
- Pal Nirvik
- From the Department of Anesthesiology, Virginia Commonwealth University, Richmond, Virginia
| | - Miklos D Kertai
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
36
|
Michalski JE, Kurche JS, Schwartz DA. From ARDS to pulmonary fibrosis: the next phase of the COVID-19 pandemic? Transl Res 2022; 241:13-24. [PMID: 34547499 PMCID: PMC8452088 DOI: 10.1016/j.trsl.2021.09.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 01/08/2023]
Abstract
While the coronavirus disease 19 (COVID-19) pandemic has transformed the medical and scientific communites since it was first reported in late 2019, we are only beginning to understand the chronic health burdens associated with this disease. Although COVID-19 is a multi-systemic disease, the lungs are the primary source of infection and injury, resulting in pneumonia and, in severe cases, acute respiratory distress syndrome (ARDS). Given that pulmonary fibrosis is a well-recognized sequela of ARDS, many have questioned whether COVID-19 survivors will face long-term pulmonary consequences. This review is aimed at integrating our understanding of the pathophysiologic mechanisms underlying fibroproliferative ARDS with our current knowledge of the pulmonary consequences of COVID-19 disease.
Collapse
Affiliation(s)
- Jacob E Michalski
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Jonathan S Kurche
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado; Medicine Service, Pulmonary Section, Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| | - David A Schwartz
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado; Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado.
| |
Collapse
|
37
|
Wang R, Qi B, Zhang X, Meng L, Wu X. Prophetic values of lung ultrasound score on post-extubation distress in patients with acute respiratory distress syndrome. Eur J Med Res 2022; 27:27. [PMID: 35193686 PMCID: PMC8864851 DOI: 10.1186/s40001-022-00652-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 02/07/2022] [Indexed: 11/10/2022] Open
Abstract
Background Acute respiratory distress syndrome (ARDS) has been a prevalent disease in ICU with mortality of up to 27–45%. A considerable proportion of extubated ARDS patients passing spontaneous breathing trial (SBT) still requires reintubation. Methods Lung ultrasonography (LUS) was used to predict the success rate of extubation. Ninety-two patients passing the 60-min SBT were included in this prospective research. Their clinical characters including LUS, APACHE II, SOFA, CPIS, EVLWI and PaO2/FiO2 were collected before the SBT. Another two LUS assessments were performed at the end of and 4 h after SBT. LUS results were evaluated and scored by two independent experts, and the correlations of LUS scores, APACHE-II scores, SOFA scores, CPIS and EVLWI with the success rate of extubation and rate of reintubation were analyzed. Results Failed weaning and reintubation of ARDS patients were correlated with higher LUS scores both before and after SBT. Post-extubation distress was correlated with higher APACHE-II scores, SOFA scores, CPIS and EVLWI before SBT. There were positive correlations between the LUS score and APACHE-II score, SOFA score, CPIS and EVLWI before SBT, respectively. Conclusion LUS score measured at the end of 60-min SBT could be used to predict post-extubation distress in ARDS patients.
Collapse
Affiliation(s)
- Ran Wang
- Department of Emergency Internal Medicine, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu, 233004, Anhui, China
| | - Benquan Qi
- Department of Emergency Internal Medicine, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu, 233004, Anhui, China
| | - Xiaohua Zhang
- Department of Emergency Internal Medicine, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu, 233004, Anhui, China
| | - Liang Meng
- Department of Emergency Internal Medicine, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu, 233004, Anhui, China
| | - Xiaofei Wu
- Department of Emergency Internal Medicine, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu, 233004, Anhui, China.
| |
Collapse
|
38
|
Overview of the Role of Pharmacological Management of Obstructive Sleep Apnea. Medicina (B Aires) 2022; 58:medicina58020225. [PMID: 35208549 PMCID: PMC8874508 DOI: 10.3390/medicina58020225] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/13/2022] [Accepted: 01/19/2022] [Indexed: 11/17/2022] Open
Abstract
Obstructive sleep apnea (OSA) remains a prominent disease state characterized by the recurrent collapse of the upper airway while sleeping. To date, current treatment may include continuous positive airway pressure (CPAP), lifestyle changes, behavioral modification, mandibular advancement devices, and surgical treatment. However, due to the desire for a more convenient mode of management, pharmacological treatment has been thoroughly investigated as a means for a potential alternative in OSA treatment. OSA can be distinguished into various endotypic or phenotypic classes, allowing pharmacological treatment to better target the root cause or symptoms of OSA. Some medications available for use include antidepressants, CNS stimulants, nasal decongestants, carbonic anhydrase inhibitors, and potassium channel blockers. This review will cover the findings of currently available and future study medications that could potentially play a role in OSA therapy.
Collapse
|
39
|
Duggal A, Kast R, Van Ark E, Bulgarelli L, Siuba MT, Osborn J, Rey DA, Zampieri FG, Cavalcanti AB, Maia I, Paisani DM, Laranjeira LN, Serpa Neto A, Deliberato RO. Identification of acute respiratory distress syndrome subphenotypes de novo using routine clinical data: a retrospective analysis of ARDS clinical trials. BMJ Open 2022; 12:e053297. [PMID: 34992112 PMCID: PMC8739395 DOI: 10.1136/bmjopen-2021-053297] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES The acute respiratory distress syndrome (ARDS) is a heterogeneous condition, and identification of subphenotypes may help in better risk stratification. Our study objective is to identify ARDS subphenotypes using new simpler methodology and readily available clinical variables. SETTING This is a retrospective Cohort Study of ARDS trials. Data from the US ARDSNet trials and from the international ART trial. PARTICIPANTS 3763 patients from ARDSNet data sets and 1010 patients from the ART data set. PRIMARY AND SECONDARY OUTCOME MEASURES The primary outcome was 60-day or 28-day mortality, depending on what was reported in the original trial. K-means cluster analysis was performed to identify subgroups. Sets of candidate variables were tested to assess their ability to produce different probabilities for mortality in each cluster. Clusters were compared with biomarker data, allowing identification of subphenotypes. RESULTS Data from 4773 patients were analysed. Two subphenotypes (A and B) resulted in optimal separation in the final model, which included nine routinely collected clinical variables, namely heart rate, mean arterial pressure, respiratory rate, bilirubin, bicarbonate, creatinine, PaO2, arterial pH and FiO2. Participants in subphenotype B showed increased levels of proinflammatory markers, had consistently higher mortality, lower number of ventilator-free days at day 28 and longer duration of ventilation compared with patients in the subphenotype A. CONCLUSIONS Routinely available clinical data can successfully identify two distinct subphenotypes in adult ARDS patients. This work may facilitate implementation of precision therapy in ARDS clinical trials.
Collapse
Affiliation(s)
- Abhijit Duggal
- Department of Critical Care Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Rachel Kast
- Department of Clinical Data Science, Endpoint Health, Palo Alto, California, USA
| | - Emily Van Ark
- Department of Clinical Data Science, Endpoint Health, Palo Alto, California, USA
| | - Lucas Bulgarelli
- Department of Clinical Data Science, Endpoint Health, Palo Alto, California, USA
| | - Matthew T Siuba
- Department of Critical Care Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jeff Osborn
- Department of Clinical Data Science, Endpoint Health, Palo Alto, California, USA
| | - Diego Ariel Rey
- Department of Clinical Data Science, Endpoint Health, Palo Alto, California, USA
| | | | | | - Israel Maia
- Hospital do Coracao, Sao Paulo, São Paulo, Brazil
| | | | | | - Ary Serpa Neto
- Australian and New Zealand Intensive Care Research Centre (ANZIC-RC), School of Public Health and Preventive Medicine, Monash University, Clayton, Victoria, Australia
- Critical Care Medicine, Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | | |
Collapse
|
40
|
Acute Respiratory Distress Syndrome: Focus on Viral Origin and Role of Pulmonary Lymphatics. Biomedicines 2021; 9:biomedicines9111732. [PMID: 34829961 PMCID: PMC8615541 DOI: 10.3390/biomedicines9111732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/01/2021] [Accepted: 11/17/2021] [Indexed: 11/30/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a serious affection of the lung caused by a variety of pathologies. Great interest is currently focused on ARDS induced by viruses (pandemic influenza and corona viruses). The review describes pulmonary changes in ARDS and specific effects of the pandemic viruses in ARDS, and summarizes treatment options. Because the known pathogenic mechanisms cannot explain all aspects of the syndrome, the contribution of pulmonary lymphatics to the pathology is discussed. Organization and function of lymphatics in a healthy lung and in resorption of pulmonary edema are described. A future clinical trial may provide more insight into the role of hyaluronan in ARDS but the development of promising pharmacological treatments is unlikely because drugs play no important role in lymphedema therapy.
Collapse
|
41
|
Fatemi Y, Bergl PA. Diagnostic Stewardship: Appropriate Testing and Judicious Treatments. Crit Care Clin 2021; 38:69-87. [PMID: 34794632 DOI: 10.1016/j.ccc.2021.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Diagnostic stewardship encompasses the entire diagnosis-to-treatment paradigm in the intensive care unit (ICU). Initially born of the antimicrobial stewardship movement, contemporary diagnostic stewardship aims to promote timely and appropriate diagnostic testing that directly links to management decisions. In the stewardship framework, excessive diagnostic testing in low probability cases is discouraged due to its tendency to generate false-positive results, which have their own downstream consequences. Though the evidence basis for diagnostic stewardship initiatives in the ICU is nascent and largely limited to retrospective analyses, available literature generally suggests that these initiatives are safe, feasible, and associated with similar patient outcomes. As diagnostic testing of critically ill patients becomes increasingly sophisticated in the ensuing decade, a stewardship mindset will aid bedside clinicians in interpreting and incorporating new diagnostic strategies in the ICU.
Collapse
Affiliation(s)
- Yasaman Fatemi
- Division of Infectious Diseases, Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| | - Paul A Bergl
- Department of Critical Care, Gundersen Lutheran Medical Center, 1900 South Avenue, Mail Stop LM3-001, La Crosse, WI 54601, USA; Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
42
|
Clinical phenotypes from fatal cases of acute respiratory distress syndrome caused by pneumonia. Sci Rep 2021; 11:20051. [PMID: 34625618 PMCID: PMC8501115 DOI: 10.1038/s41598-021-99540-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 09/27/2021] [Indexed: 12/11/2022] Open
Abstract
There have been no report of objective clinical characteristics or prognostic factors that predict fatal outcome of acute respiratory distress syndrome (ARDS) since the Berlin definition was published. The aim of this study is to identify clinically available predictors that distinguish between two phenotypes of fatal ARDS due to pneumonia. In total, 104 cases of Japanese patients with pneumonia-induced ARDS were extracted from our prospectively collected database. Fatal cases were divided into early (< 7 days after diagnosis) and late (≥ 7 days) death groups, and clinical variables and prognostic factors were statistically evaluated. Of the 50 patients who died within 180 days, 18 (36%) and 32 (64%) were in the early (median 2 days, IQR [1, 5]) and late (median 16 days, IQR [13, 29]) death groups, respectively. According to multivariate regression analyses, the APACHE II score (HR 1.25, 95%CI 1.12–1.39, p < 0.001) and the disseminated intravascular coagulation score (HR 1.54, 95%CI 1.15–2.04, p = 0.003) were independent prognostic factors for early death. In contrast, late death was associated with high-resolution computed tomography (HRCT) score indicating early fibroproliferation (HR 1.28, 95%CI 1.13–1.42, p < 0.001) as well as the disseminated intravascular coagulation score (HR 1.24, 95%CI 1.01–1.52, p = 0.039). The extent of fibroproliferation on HRCT, and the APACHE II scores along with coagulation abnormalities, should be considered for use in predictive enrichment and personalized medicine for patients with ARDS due to pneumonia.
Collapse
|
43
|
Xu G, Wan H, Yi L, Chen W, Luo Y, Huang Y, Liu X. Berberine administrated with different routes attenuates inhaled LPS-induced acute respiratory distress syndrome through TLR4/NF-κB and JAK2/STAT3 inhibition. Eur J Pharmacol 2021; 908:174349. [PMID: 34284014 PMCID: PMC8285933 DOI: 10.1016/j.ejphar.2021.174349] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 07/07/2021] [Accepted: 07/12/2021] [Indexed: 12/17/2022]
Abstract
Accumulating evidence showed that berberine possessed the anti-inflammatory action in various diseases caused by inflammation. However, it was still unclear whether both inhalation and injection with berberine produced pulmonary protective role in acute respiratory distress syndrome (ARDS). This study was aimed to evaluate the effects of both administration routes including inhalation and injection with berberine in ARDS induced by lipopolysaccharide (LPS) inhalation. Histopathological examination and weight of lung were evaluated. Phosphorylation of NF-κB, JAK2 and STAT3 were measured to assess the activity of inflammation related signaling pathways. Proinflammatory cytokines including interleukin (IL)-1β and tumor necrosis factor (TNF)-α in the bronchoalveolar lavage fluid (BALF) and serum were also detected. The results showed that LPS caused the lung injury, while both administration routes with berberine attenuated the injury and improved the pulmonary morphology. In addition, the primary TLR4/NF-κB and secondary JAK2/STAT3 signaling pathways which were activated by LPS in lung were totally inhibited by berberine administration. Moreover, proinflammatory cytokines in both BALF and serum were decreased by berberine. Considering that molecular docking simulation indicated that berberine could bind with TLR4, the present suggested that the inhibition of the inflammation related TLR4/NF-κB and JAK2/STAT3 signaling pathways might be involved in the pulmonary protective effect of berberine in LPS-induced ARDS.
Collapse
Affiliation(s)
- Guanghui Xu
- Pharmaceutical Research Center, Xiamen Medicine Research Institute, Xiamen, 361008, Fujian province, PR China.
| | - Huiqi Wan
- Pharmaceutical Research Center, Xiamen Medicine Research Institute, Xiamen, 361008, Fujian province, PR China
| | - Litao Yi
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen, 361021, Fujian province, PR China.
| | - Wei Chen
- Pharmaceutical Research Center, Xiamen Medicine Research Institute, Xiamen, 361008, Fujian province, PR China
| | - Youhua Luo
- Pharmaceutical Research Center, Xiamen Medicine Research Institute, Xiamen, 361008, Fujian province, PR China
| | - Yiqi Huang
- Pharmaceutical Research Center, Xiamen Medicine Research Institute, Xiamen, 361008, Fujian province, PR China
| | - Xiaojuan Liu
- Pharmaceutical Research Center, Xiamen Medicine Research Institute, Xiamen, 361008, Fujian province, PR China
| |
Collapse
|
44
|
Rolandsson Enes S, Hampton TH, Barua J, McKenna DH, Dos Santos CC, Amiel E, Ashare A, Liu KD, Krasnodembskaya AD, English K, Stanton BA, Rocco PRM, Matthay MA, Weiss DJ. Healthy versus inflamed lung environments differentially affect mesenchymal stromal cells. Eur Respir J 2021; 58:2004149. [PMID: 33795318 PMCID: PMC8543758 DOI: 10.1183/13993003.04149-2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/02/2021] [Indexed: 11/05/2022]
Abstract
BACKGROUND Despite increased interest in mesenchymal stromal cell (MSC)-based cell therapies for acute respiratory distress syndrome (ARDS), clinical investigations have not yet been successful and our understanding of the potential in vivo mechanisms of MSC actions in ARDS remains limited. ARDS is driven by an acute severe innate immune dysregulation, often characterised by inflammation, coagulation and cell injury. How this inflammatory microenvironment influences MSC functions remains to be determined. AIM The aim of this study was to comparatively assess how the inflammatory environment present in ARDS lungs versus the lung environment present in healthy volunteers alters MSC behaviour. METHODS Clinical-grade human bone marrow-derived MSCs (hMSCs) were exposed to bronchoalveolar lavage fluid (BALF) samples obtained from ARDS patients or from healthy volunteers. Following exposure, hMSCs and their conditioned media were evaluated for a broad panel of relevant properties, including viability, levels of expression of inflammatory cytokines, gene expression, cell surface human leukocyte antigen expression, and activation of coagulation and complement pathways. RESULTS Pro-inflammatory, pro-coagulant and major histocompatibility complex (self-recognition) related gene expression was markedly upregulated in hMSCs exposed ex vivo to BALF obtained from healthy volunteers. These changes were less apparent and often opposite in hMSCs exposed to ARDS BALF samples. CONCLUSION These data provide new insights into how hMSCs behave in healthy versus inflamed lung environments, and strongly suggest that the inflamed environment in ARDS induces hMSC responses that are potentially beneficial for cell survival and actions. This further highlights the need to understand how different disease environments affect hMSC functions.
Collapse
Affiliation(s)
- Sara Rolandsson Enes
- Dept of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
- Dept of Experimental Medical Science, Lung Biology Unit, Lund University, Lund, Sweden
| | - Thomas H Hampton
- Dept of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Jayita Barua
- Dept of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - David H McKenna
- Dept of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Claudia C Dos Santos
- Interdepartmental Division of Critical Care, Dept of Medicine and the Keenan Center for Biomedical Research, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Eyal Amiel
- Dept of Biomedical and Health Sciences, College of Nursing and Health Sciences, University of Vermont, Burlington, VT, USA
| | - Alix Ashare
- Dept of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Section of Pulmonary and Critical Care Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Kathleen D Liu
- Depts of Medicine and Anesthesiology and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Anna D Krasnodembskaya
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queens University, Belfast, UK
| | - Karen English
- Cellular Immunology Laboratory, Biology Dept, Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Bruce A Stanton
- Dept of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Michael A Matthay
- Depts of Medicine and Anesthesiology and the Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Daniel J Weiss
- Dept of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| |
Collapse
|
45
|
Song D, Zhao M, Feng L, Wang P, Li Y, Li W. Salidroside attenuates acute lung injury via inhibition of inflammatory cytokine production. Biomed Pharmacother 2021; 142:111949. [PMID: 34325302 DOI: 10.1016/j.biopha.2021.111949] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/10/2021] [Accepted: 07/14/2021] [Indexed: 12/27/2022] Open
Abstract
Acute lung injury is a fatal condition characterized by excessive inflammation responses. Salidroside, the active constituent of Rhodiola rosea, possesses properties including anti-oxidation, anti-aging, anti-inflammatory, anti-hypoxia, and anti-cancer activities. In the present study, Salidroside attenuated acute lung injury via inhibition of inflammatory cytokine production. Rats pre-treated with Salidroside showed attenuated lipopolysaccharide (LPS)-induced pathological damage and suppressed tumor necrosis factor-alpha (TNFα) and interleukin 6 (IL-6) secretion in the lung. Furthermore, flow cytometry showed that Salidroside reduced the production of TNFα and IL-6 in NR8383 alveolar macrophages. These findings suggest that Salidroside may attenuate LPS-induced acute lung injury.
Collapse
Affiliation(s)
- Dan Song
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of Education, School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China; Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China
| | - Min Zhao
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of Education, School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China; Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China
| | - Liuxiang Feng
- People's Hospital of Yulong Naxi Autonomous County of Lijiang City, Yulong Naxi Autonomous County 674100, Yunnan, China
| | - Pingyi Wang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China
| | - Yimei Li
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China
| | - Wenhua Li
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of Education, School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China; Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang 712082, Shaanxi, China.
| |
Collapse
|
46
|
Rich TC, Leavesley SJ, Brandon AP, Evans CA, Raju SV, Wagener BM. Phosphodiesterase 4 mediates interleukin-8-induced heterologous desensitization of the β 2 -adrenergic receptor. FASEB J 2021; 35:e21946. [PMID: 34555226 DOI: 10.1096/fj.202002712rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 11/11/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening illness characterized by decreased alveolar-capillary barrier function, pulmonary edema consisting of proteinaceous fluid, and inhibition of net alveolar fluid transport responsible for resolution of pulmonary edema. There is currently no pharmacotherapy that has proven useful to prevent or treat ARDS, and two trials using beta-agonist therapy to treat ARDS demonstrated no effect. Prior studies indicated that IL-8-induced heterologous desensitization of the beta2-adrenergic receptor (β2 -AR) led to decreased beta-agonist-induced mobilization of cyclic adenosine monophosphate (cAMP). Interestingly, phosphodiesterase (PDE) 4 inhibitors have been used in human airway diseases characterized by low intracellular cAMP levels and increases in specific cAMP hydrolyzing activity. Therefore, we hypothesized that PDE4 would mediate IL-8-induced heterologous internalization of the β2 -AR and that PDE4 inhibition would restore beta-agonist-induced functions. We determined that CINC-1 (a functional IL-8 analog in rats) induces internalization of β2 -AR from the cell surface, and arrestin-2, PDE4, and β2 -AR form a complex during this process. Furthermore, we determined that cAMP associated with the plasma membrane was adversely affected by β2 -AR heterologous desensitization. Additionally, we determined that rolipram, a PDE4 inhibitor, reversed CINC-1-induced derangements of cAMP and also caused β2 -AR to successfully recycle back to the cell surface. Finally, we demonstrated that rolipram could reverse CINC-1-mediated inhibition of beta-agonist-induced alveolar fluid clearance in a murine model of trauma-shock. These results indicate that PDE4 plays a role in CINC-1-induced heterologous internalization of the β2 -AR; PDE4 inhibition reverses these effects and may be a useful adjunct in particular ARDS patients.
Collapse
Affiliation(s)
- Thomas C Rich
- Department of Pharmacology, University of South Alabama, Mobile, Alabama, USA.,Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA
| | - Silas J Leavesley
- Department of Pharmacology, University of South Alabama, Mobile, Alabama, USA.,Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA.,Department of Chemical and Biomolecular Engineering, University of South Alabama, Mobile, Alabama, USA
| | - Angela P Brandon
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Cilina A Evans
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - S Vamsee Raju
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.,UAB Lung Health Center, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Brant M Wagener
- Division of Molecular and Translational Biomedicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Division of Critical Care Medicine, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.,Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
47
|
Kotas ME, Thompson BT. Toward Optimal Acute Respiratory Distress Syndrome Outcomes: Recognizing the Syndrome and Identifying Its Causes. Crit Care Clin 2021; 37:733-748. [PMID: 34548131 PMCID: PMC8449137 DOI: 10.1016/j.ccc.2021.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Maya E Kotas
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California, San Francisco, 505 Parnassus Avenue, Box 0111, San Francisco, CA 94143, USA
| | - B Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA.
| |
Collapse
|
48
|
Affiliation(s)
- Jisoo Lee
- Division of Pulmonary, Critical Care & Sleep Medicine, Rhode Island Hospital, 593 Eddy Street, POB Suite 224, Room 222.1, Providence, RI 02903, USA.
| | - Keith Corl
- Division of Pulmonary, Critical Care & Sleep Medicine, Rhode Island Hospital, 593 Eddy Street, POB Suite 224, Room 222.1, Providence, RI 02903, USA
| | - Mitchell M Levy
- Division of Pulmonary, Critical Care & Sleep Medicine, Rhode Island Hospital, 593 Eddy Street, POB Suite 224, Room 222.1, Providence, RI 02903, USA.
| |
Collapse
|
49
|
Schwager E, Jansson K, Rahman A, Schiffer S, Chang Y, Boverman G, Gross B, Xu-Wilson M, Boehme P, Truebel H, Frassica JJ. Utilizing machine learning to improve clinical trial design for acute respiratory distress syndrome. NPJ Digit Med 2021; 4:133. [PMID: 34504281 PMCID: PMC8429640 DOI: 10.1038/s41746-021-00505-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 08/09/2021] [Indexed: 02/05/2023] Open
Abstract
Heterogeneous patient populations, complex pharmacology and low recruitment rates in the Intensive Care Unit (ICU) have led to the failure of many clinical trials. Recently, machine learning (ML) emerged as a new technology to process and identify big data relationships, enabling a new era in clinical trial design. In this study, we designed a ML model for predictively stratifying acute respiratory distress syndrome (ARDS) patients, ultimately reducing the required number of patients by increasing statistical power through cohort homogeneity. From the Philips eICU Research Institute (eRI) database, no less than 51,555 ARDS patients were extracted. We defined three subpopulations by outcome: (1) rapid death, (2) spontaneous recovery, and (3) long-stay patients. A retrospective univariate analysis identified highly predictive variables for each outcome. All 220 variables were used to determine the most accurate and generalizable model to predict long-stay patients. Multiclass gradient boosting was identified as the best-performing ML model. Whereas alterations in pH, bicarbonate or lactate proved to be strong predictors for rapid death in the univariate analysis, only the multivariate ML model was able to reliably differentiate the disease course of the long-stay outcome population (AUC of 0.77). We demonstrate the feasibility of prospective patient stratification using ML algorithms in the by far largest ARDS cohort reported to date. Our algorithm can identify patients with sufficiently long ARDS episodes to allow time for patients to respond to therapy, increasing statistical power. Further, early enrollment alerts may increase recruitment rate.
Collapse
Affiliation(s)
- E Schwager
- Philips Research North America, Cambridge, MA, USA
| | - K Jansson
- Research & Development, Pharmaceuticals, Bayer AG, Wuppertal, Germany
| | - A Rahman
- Philips Research North America, Cambridge, MA, USA
| | - S Schiffer
- Research & Development, Pharmaceuticals, Bayer AG, Wuppertal, Germany
| | - Y Chang
- Philips Research North America, Cambridge, MA, USA
| | - G Boverman
- Philips Research North America, Cambridge, MA, USA
| | - B Gross
- Philips Research North America, Cambridge, MA, USA
| | - M Xu-Wilson
- Philips Research North America, Cambridge, MA, USA
| | - P Boehme
- Research & Development, Pharmaceuticals, Bayer AG, Wuppertal, Germany.,Faculty of Health, Witten/Herdecke University, Witten, Germany
| | - H Truebel
- Research & Development, Pharmaceuticals, Bayer AG, Wuppertal, Germany. .,Faculty of Health, Witten/Herdecke University, Witten, Germany.
| | - J J Frassica
- Philips Research North America, Cambridge, MA, USA. .,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
50
|
Liu X, Jiang Y, Jia X, Ma X, Han C, Guo N, Peng Y, Liu H, Ju Y, Luo X, Li X, Bu Y, Zhang J, Liu Y, Gao Y, Zhao M, Wang H, Luo L, Yu K, Wang C. Identification of distinct clinical phenotypes of acute respiratory distress syndrome with differential responses to treatment. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2021; 25:320. [PMID: 34461969 PMCID: PMC8404019 DOI: 10.1186/s13054-021-03734-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/18/2021] [Indexed: 02/05/2023]
Abstract
Background Acute respiratory distress syndrome (ARDS) is a heterogeneous syndrome, and the identification of homogeneous subgroups and phenotypes is the first step toward precision critical care. We aimed to explore whether ARDS phenotypes can be identified using clinical data, are reproducible and are associated with clinical outcomes and treatment response. Methods This study is based on a retrospective analysis of data from the telehealth intensive care unit (eICU) collaborative research database and three ARDS randomized controlled trials (RCTs) (ALVEOLI, FACTT and SAILS trials). We derived phenotypes in the eICU by cluster analysis based on clinical data and compared the clinical characteristics and outcomes of each phenotype. The reproducibility of the derived phenotypes was tested using the data from three RCTs, and treatment effects were evaluated. Results Three clinical phenotypes were identified in the training cohort of 3875 ARDS patients. Of the three phenotypes identified, phenotype I (n = 1565; 40%) was associated with fewer laboratory abnormalities, less organ dysfunction and the lowest in-hospital mortality rate (8%). Phenotype II (n = 1232; 32%) was correlated with more inflammation and shock and had a higher mortality rate (18%). Phenotype III (n = 1078; 28%) was strongly correlated with renal dysfunction and acidosis and had the highest mortality rate (22%). These results were validated using the data from the validation cohort (n = 3670) and three RCTs (n = 2289) and had reproducibility. Patients with these ARDS phenotypes had different treatment responses to randomized interventions. Specifically, in the ALVEOLI cohort, the effects of ventilation strategy (high PEEP vs low PEEP) on ventilator-free days differed by phenotype (p = 0.001); in the FACTT cohort, there was a significant interaction between phenotype and fluid-management strategy for 60-day mortality (p = 0.01). The fluid-conservative strategy was associated with improved mortality in phenotype II but had the opposite effect in phenotype III. Conclusion Three clinical phenotypes of ARDS were identified and had different clinical characteristics and outcomes. The analysis shows evidence of a phenotype-specific treatment benefit in the ALVEOLI and FACTT trials. These findings may improve the identification of distinct subsets of ARDS patients for exploration in future RCTs. Supplementary Information The online version contains supplementary material available at 10.1186/s13054-021-03734-y.
Collapse
Affiliation(s)
- Xiaowei Liu
- Department of Critical Care Medicine, First Affiliated Hospital of Harbin Medical University, 23 Postal Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Yusheng Jiang
- LinkDoc AI Lab, LinkDoc Technology, Floor 11, Sinosteel Plaza, 8 Haidian Street, Haidian District, Beijing, 100080, China
| | - Xiaonan Jia
- Department of Critical Care Medicine, First Affiliated Hospital of Harbin Medical University, 23 Postal Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Xiaohui Ma
- Department of Critical Care Medicine, First Affiliated Hospital of Harbin Medical University, 23 Postal Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Ci Han
- Department of Critical Care Medicine, First Affiliated Hospital of Harbin Medical University, 23 Postal Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Nana Guo
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Yahui Peng
- Department of Critical Care Medicine, First Affiliated Hospital of Harbin Medical University, 23 Postal Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Haitao Liu
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Yingnan Ju
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Xiangfeng Luo
- LinkDoc AI Lab, LinkDoc Technology, Floor 11, Sinosteel Plaza, 8 Haidian Street, Haidian District, Beijing, 100080, China
| | - Xueting Li
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China
| | - Yue Bu
- Department of Critical Care Medicine, First Affiliated Hospital of Harbin Medical University, 23 Postal Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Jin Zhang
- Department of Critical Care Medicine, First Affiliated Hospital of Harbin Medical University, 23 Postal Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Yansong Liu
- Department of Critical Care Medicine, First Affiliated Hospital of Harbin Medical University, 23 Postal Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Yan Gao
- Department of Critical Care Medicine, Forth Affiliated Hospital of Harbin Medical University, 37 Yiyuan Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Mingyan Zhao
- Department of Critical Care Medicine, First Affiliated Hospital of Harbin Medical University, 23 Postal Street, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Hongliang Wang
- Department of Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Ligang Luo
- LinkDoc AI Lab, LinkDoc Technology, Floor 11, Sinosteel Plaza, 8 Haidian Street, Haidian District, Beijing, 100080, China
| | - Kaijiang Yu
- Department of Critical Care Medicine, First Affiliated Hospital of Harbin Medical University, 23 Postal Street, Nangang District, Harbin, 150001, Heilongjiang, China.
| | - Changsong Wang
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, Heilongjiang, China.
| |
Collapse
|