1
|
Ishioka H, Ghose A, Kingston HW, Plewes K, Leopold SJ, Srinamon K, Charunwatthana P, Ahmed M, Alam AKMS, Tuip-de Boer A, Hossain MA, Dondorp AM, Schultz MJ. The predictive capacity of biomarkers for clinical pulmonary oedema in patients with severe falciparum malaria is low: a prospective observational study. Malar J 2024; 23:320. [PMID: 39448997 PMCID: PMC11515577 DOI: 10.1186/s12936-024-05142-3] [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: 07/08/2024] [Accepted: 10/12/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Pulmonary oedema is a feared and difficult to predict complication of severe malaria that can emerge after start of antimalarial treatment. Proinflammatory mediators are thought to play a central role in its pathogenesis. METHODS An exploratory study was conducted to evaluate the predictive capacity of biomarkers for development of clinical pulmonary oedema in patients with severe falciparum malaria at two hospitals in Bangladesh. Plasma concentrations of interleukin-6 (IL-6), IL-8, tumour necrosis factor (TNF), soluble Receptor of Advanced Glycation End-products (sRAGE), surfactant protein-D (SP-D), club cell secretory protein (CC16), and Krebs von den Lungen-6 (KL-6) on admission were compared with healthy controls. Correlations between these biomarker and plasma lactate and Plasmodium falciparum histidine-rich protein 2 (PfHRP2) levels were evaluated. Receiver Operating Characteristic (ROC) curves were constructed to assess the predictive capacity for clinical pulmonary oedema of the biomarkers of interest. RESULTS Of 106 screened patients with falciparum malaria, 56 were classified as having severe malaria with a mortality rate of 29%. Nine (16%) patients developed clinical pulmonary oedema after admission. Plasma levels of the biomarkers of interest were higher in patients compared to healthy controls. IL-6, IL-8, TNF, sRAGE, and CC16 levels correlated well with plasma PfHRP2 levels (rs = 0.39; P = 0.004, rs = 0.43; P = 0.001, rs = 0.54; P < 0.001, rs = 0.44; P < 0.001, rs = 0.43; P = 0.001, respectively). Furthermore, IL-6 and IL-8 levels correlated well with plasma lactate levels (rs = 0.37; P = 0.005, rs = 0.47; P < 0.001, respectively). None of the biomarkers of interest had predictive capacity for development of clinical pulmonary oedema. CONCLUSIONS IL-6, IL-8, TNF, sRAGE, SP-D, CC16 and KL-6 cannot be used in predicting clinical pulmonary oedema in severe malaria patients.
Collapse
Affiliation(s)
- Haruhiko Ishioka
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
- Division of Infectious Diseases, Jichi Medical University Hospital, 3311-1 Yakushiji Shimotsuke-shi, Tochigi, 329-0498, Japan.
| | | | - Hugh W Kingston
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Katherine Plewes
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Stije J Leopold
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Ketsanee Srinamon
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Prakaykaew Charunwatthana
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Maswood Ahmed
- Chittagong Medical College Hospital, Chattogram, Bangladesh
| | | | - Anita Tuip-de Boer
- Department of Intensive Care, Amsterdam University Medical Center, Amsterdam, Netherlands
| | | | - Arjen M Dondorp
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Marcus J Schultz
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Intensive Care, Amsterdam University Medical Center, Amsterdam, Netherlands
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
2
|
Yin Y, Zeng Z, Wei S, Shen Z, Cong Z, Zhu X. Using the sympathetic system, beta blockers and alpha-2 agonists, to address acute respiratory distress syndrome. Int Immunopharmacol 2024; 139:112670. [PMID: 39018694 DOI: 10.1016/j.intimp.2024.112670] [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: 05/31/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024]
Abstract
Acute Respiratory Distress Syndrome (ARDS) manifests as an acute inflammatory lung injury characterized by persistent hypoxemia, featuring a swift onset, high mortality, and predominantly supportive care as the current therapeutic approach, while effective treatments remain an area of active investigation. Adrenergic receptors (AR) play a pivotal role as stress hormone receptors, extensively participating in various inflammatory processes by initiating downstream signaling pathways. Advancements in molecular biology and pharmacology continually unveil the physiological significance of distinct AR subtypes. Interventions targeting these subtypes have the potential to induce specific alterations in cellular and organismal functions, presenting a promising avenue as a therapeutic target for managing ARDS. This article elucidates the pathogenesis of ARDS and the basic structure and function of AR. It also explores the relationship between AR and ARDS from the perspective of different AR subtypes, aiming to provide new insights for the improvement of ARDS.
Collapse
Affiliation(s)
- Yiyuan Yin
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Zhaojin Zeng
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Senhao Wei
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China
| | - Ziyuan Shen
- Department of Anaesthesiology, Peking University Third Hospital, Beijing, China
| | - Zhukai Cong
- Department of Anaesthesiology, Peking University Third Hospital, Beijing, China.
| | - Xi Zhu
- Department of Intensive Care Unit, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
3
|
Alladina JW, Giacona FL, Haring AM, Hibbert KA, Medoff BD, Schmidt EP, Thompson T, Maron BA, Alba GA. Circulating Biomarkers of Endothelial Dysfunction Associated With Ventilatory Ratio and Mortality in ARDS Resulting From SARS-CoV-2 Infection Treated With Antiinflammatory Therapies. CHEST CRITICAL CARE 2024; 2:100054. [PMID: 39035722 PMCID: PMC11259037 DOI: 10.1016/j.chstcc.2024.100054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
BACKGROUND The association of plasma biomarkers and clinical outcomes in ARDS resulting from SARS-CoV-2 infection predate the evidence-based use of immunomodulators. RESEARCH QUESTION Which plasma biomarkers are associated with clinical outcomes in patients with ARDS resulting from SARS-CoV-2 infection treated routinely with immunomodulators? STUDY DESIGN AND METHODS We collected plasma from patients with ARDS resulting from SARS-CoV-2 infection within 24 h of admission to the ICU between December 2020 and March 2021 (N = 69). We associated 16 total biomarkers of inflammation (eg, IL-6), coagulation (eg, D-dimer), epithelial injury (eg, surfactant protein D), and endothelial injury (eg, angiopoietin-2) with the primary outcome of in-hospital mortality and secondary outcome of ventilatory ratio (at baseline and day 3). RESULTS Thirty patients (43.5%) died within 60 days. All patients received corticosteroids and 6% also received tocilizumab. Compared with survivors, nonsurvivors demonstrated a higher baseline modified Sequential Organ Failure Assessment score (median, 8.5 [interquartile range (IQR), 7-9] vs 7 [IQR, 5-8]); P = .004), lower Pao2 to Fio2 ratio (median, 153 [IQR, 118-182] vs 184 [IQR, 142-247]; P = .04), and higher ventilatory ratio (median, 2.0 [IQR, 1.9-2.3] vs 1.5 [IQR, 1.4-1.9]; P < .001). No difference was found in inflammatory, coagulation, or epithelial biomarkers between groups. Nonsurvivors showed higher median neural precursor cell expressed, developmentally down-regulated 9 (NEDD9) levels (median, 8.4 ng/mL [IQR, 7.0-11.2 ng/mL] vs 6.9 ng/mL [IQR, 5.5-8.0 ng/mL]; P = .0025), von Willebrand factor domain A2 levels (8.7 ng/mL [IQR, 7.9-9.7 ng/mL] vs 6.5 ng/mL [IQR, 5.7-8.7 ng/mL]; P = .007), angiopoietin-2 levels (9.0 ng/mL [IQR, 7.9-14.1 ng/mL] vs 7.0 ng/mL [IQR, 5.6-10.6 ng/mL]; P = .01), and syndecan-1 levels (15.9 ng/mL [IQR, 14.5-17.5 ng/mL] vs 12.6 ng/mL [IQR, 10.5-16.1 ng/mL]; P = .01). Only NEDD9 level met the adjusted threshold for significance (P < .003). Plasma NEDD9 level was associated with 60-day mortality (adjusted OR, 9.7; 95% CI, 1.6-60.4; P = .015). Syndecan-1 level correlated with both baseline (ρ = 0.4; P = .001) and day 3 ventilatory ratio (ρ = 0.5; P < .001). INTERPRETATION Biomarkers of inflammation, coagulation, and epithelial injury were not associated with clinical outcomes in a small cohort of patients with ARDS uniformly treated with immunomodulators. However, endothelial biomarkers, including plasma NEDD9, were associated with 60-day mortality.
Collapse
Affiliation(s)
- Jehan W Alladina
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Francesca L Giacona
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Alexis M Haring
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Kathryn A Hibbert
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Benjamin D Medoff
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Eric P Schmidt
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Bradley A Maron
- Department of Medicine; University of Maryland School of Medicine, Baltimore, University of Maryland-Institute for Health Computing, Bethesda, MD
| | - George A Alba
- Division of Pulmonary and Critical Care Medicine, Bethesda, MD, Department of Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
4
|
Côté A, Lee CH, Metwaly SM, Doig CJ, Andonegui G, Yipp BG, Parhar KKS, Winston BW. Endotyping in ARDS: one step forward in precision medicine. Eur J Med Res 2024; 29:284. [PMID: 38745261 PMCID: PMC11092098 DOI: 10.1186/s40001-024-01876-7] [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: 04/06/2023] [Accepted: 04/30/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND The Berlin definition of acute respiratory distress syndrome (ARDS) includes only clinical characteristics. Understanding unique patient pathobiology may allow personalized treatment. We aimed to define and describe ARDS phenotypes/endotypes combining clinical and pathophysiologic parameters from a Canadian ARDS cohort. METHODS A cohort of adult ARDS patients from multiple sites in Calgary, Canada, had plasma cytokine levels and clinical parameters measured in the first 24 h of ICU admission. We used a latent class model (LCM) to group the patients into several ARDS subgroups and identified the features differentiating those subgroups. We then discuss the subgroup effect on 30 day mortality. RESULTS The LCM suggested three subgroups (n1 = 64, n2 = 86, and n3 = 30), and 23 out of 69 features made these subgroups distinct. The top five discriminating features were IL-8, IL-6, IL-10, TNF-a, and serum lactate. Mortality distinctively varied between subgroups. Individual clinical characteristics within the subgroup associated with mortality included mean PaO2/FiO2 ratio, pneumonia, platelet count, and bicarbonate negatively associated with mortality, while lactate, creatinine, shock, chronic kidney disease, vasopressor/ionotropic use, low GCS at admission, and sepsis were positively associated. IL-8 and Apache II were individual markers strongly associated with mortality (Area Under the Curve = 0.84). PERSPECTIVE ARDS subgrouping using biomarkers and clinical characteristics is useful for categorizing a heterogeneous condition into several homogenous patient groups. This study found three ARDS subgroups using LCM; each subgroup has a different level of mortality. This model may also apply to developing further trial design, prognostication, and treatment selection.
Collapse
Affiliation(s)
- Andréanne Côté
- Department of Medicine, Institut Universitaire de Cardiologie et de Pneumologie de Quebec-Université Laval, Quebec, Canada
- Department of Critical Care Medicine, Medicine and Biochemistry and Molecular Biology, Health Research Innovation Center (HRIC), University of Calgary, Room 4C64, 3280 Hospital Drive N.W., Calgary, AB, T2N 4Z6, Canada
| | - Chel Hee Lee
- Department of Critical Care Medicine, Medicine and Biochemistry and Molecular Biology, Health Research Innovation Center (HRIC), University of Calgary, Room 4C64, 3280 Hospital Drive N.W., Calgary, AB, T2N 4Z6, Canada
- Department of Mathematics and Statistics, University of Calgary, Calgary, Canada
| | - Sayed M Metwaly
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
- Division of Internal Medicine, Aberdeen Royal Infirmary, NHS Scotland, Aberdeen, UK
| | - Christopher J Doig
- Department of Critical Care Medicine, Medicine and Biochemistry and Molecular Biology, Health Research Innovation Center (HRIC), University of Calgary, Room 4C64, 3280 Hospital Drive N.W., Calgary, AB, T2N 4Z6, Canada
| | | | - Bryan G Yipp
- Department of Critical Care Medicine, Medicine and Biochemistry and Molecular Biology, Health Research Innovation Center (HRIC), University of Calgary, Room 4C64, 3280 Hospital Drive N.W., Calgary, AB, T2N 4Z6, Canada
| | - Ken Kuljit S Parhar
- Department of Critical Care Medicine, Medicine and Biochemistry and Molecular Biology, Health Research Innovation Center (HRIC), University of Calgary, Room 4C64, 3280 Hospital Drive N.W., Calgary, AB, T2N 4Z6, Canada
| | - Brent W Winston
- Department of Critical Care Medicine, Medicine and Biochemistry and Molecular Biology, Health Research Innovation Center (HRIC), University of Calgary, Room 4C64, 3280 Hospital Drive N.W., Calgary, AB, T2N 4Z6, Canada.
- Depatments of Medicine, University of Calgary, Calgary, Canada.
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada.
| |
Collapse
|
5
|
Qiao X, Yin J, Zheng Z, Li L, Feng X. Endothelial cell dynamics in sepsis-induced acute lung injury and acute respiratory distress syndrome: pathogenesis and therapeutic implications. Cell Commun Signal 2024; 22:241. [PMID: 38664775 PMCID: PMC11046830 DOI: 10.1186/s12964-024-01620-y] [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: 02/28/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Sepsis, a prevalent critical condition in clinics, continues to be the leading cause of death from infections and a global healthcare issue. Among the organs susceptible to the harmful effects of sepsis, the lungs are notably the most frequently affected. Consequently, patients with sepsis are predisposed to developing acute lung injury (ALI), and in severe cases, acute respiratory distress syndrome (ARDS). Nevertheless, the precise mechanisms associated with the onset of ALI/ARDS remain elusive. In recent years, there has been a growing emphasis on the role of endothelial cells (ECs), a cell type integral to lung barrier function, and their interactions with various stromal cells in sepsis-induced ALI/ARDS. In this comprehensive review, we summarize the involvement of endothelial cells and their intricate interplay with immune cells and stromal cells, including pulmonary epithelial cells and fibroblasts, in the pathogenesis of sepsis-induced ALI/ARDS, with particular emphasis placed on discussing the several pivotal pathways implicated in this process. Furthermore, we discuss the potential therapeutic interventions for modulating the functions of endothelial cells, their interactions with immune cells and stromal cells, and relevant pathways associated with ALI/ARDS to present a potential therapeutic strategy for managing sepsis and sepsis-induced ALI/ARDS.
Collapse
Affiliation(s)
- Xinyu Qiao
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Junhao Yin
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Zhihuan Zheng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Liangge Li
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Xiujing Feng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.
- School of Clinical and Basic Medical Sciences, Shandong First Medical University& Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China.
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
6
|
Gazzaniga G, Voltini M, Carletti A, Lenta E, Meloni F, Briganti DF, Avanzini MA, Comoli P, Belliato M. Potential application of mesenchymal stromal cells as a new therapeutic approach in acute respiratory distress syndrome and pulmonary fibrosis. Respir Res 2024; 25:170. [PMID: 38637860 PMCID: PMC11027419 DOI: 10.1186/s12931-024-02795-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 03/29/2024] [Indexed: 04/20/2024] Open
Abstract
While the COVID-19 outbreak and its complications are still under investigation, post-inflammatory pulmonary fibrosis (PF) has already been described as a long-term sequela of acute respiratory distress syndrome (ARDS) secondary to SARS-CoV2 infection. However, therapeutical strategies for patients with ARDS and PF are still limited and do not significantly extend lifespan. So far, lung transplantation remains the only definitive treatment for end-stage PF. Over the last years, numerous preclinical and clinical studies have shown that allogeneic mesenchymal stromal cells (MSCs) might represent a promising therapeutical approach in several lung disorders, and their potential for ARDS treatment and PF prevention has been investigated during the COVID-19 pandemic. From April 2020 to April 2022, we treated six adult patients with moderate COVID-19-related ARDS in a late proliferative stage with up to two same-dose infusions of third-party allogeneic bone marrow-derived MSCs (BM-MSCs), administered intravenously 15 days apart. No major adverse events were registered. Four patients completed the treatment and reached ICU discharge, while two received only one dose of MSCs due to multiorgan dysfunction syndrome (MODS) and subsequent death. All four survivors showed improved gas exchanges (PaO2/FiO2 ratio > 200), contrary to the others. Furthermore, LDH trends after MSCs significantly differed between survivors and the deceased. Although further investigations and shared protocols are still needed, the safety of MSC therapy has been recurrently shown, and its potential in treating ARDS and preventing PF might represent a new therapeutic strategy.
Collapse
Affiliation(s)
- Giulia Gazzaniga
- SC Anestesia e Rianimazione 2, Fondazione IRCCS Policlinico San Matteo, Viale Camillo Golgi 19, Pavia, PV, 27100, Italy.
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy.
- Cardio-Thoracic Surgery Department, Heart & Vascular Centre, Maastricht University Medical Centre (MUMC+), P. Debyelaan 25, Maastricht, 6229 HX, The Netherlands.
| | - Marta Voltini
- SC Anestesia e Rianimazione 2, Fondazione IRCCS Policlinico San Matteo, Viale Camillo Golgi 19, Pavia, PV, 27100, Italy
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Alessandro Carletti
- SC Anestesia e Rianimazione 3 - TIPO, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Elisa Lenta
- SSD Cell Factory and Center for Advanced Therapies, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Federica Meloni
- UOS Transplant Center, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Internal Medicine, University of Pavia, Pavia, Italy
| | - Domenica Federica Briganti
- UOS Transplant Center, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Internal Medicine, University of Pavia, Pavia, Italy
| | - Maria Antonietta Avanzini
- SSD Cell Factory and Center for Advanced Therapies, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Pediatric Hematology/Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Patrizia Comoli
- SSD Cell Factory and Center for Advanced Therapies, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Pediatric Hematology/Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Mirko Belliato
- SC Anestesia e Rianimazione 2, Fondazione IRCCS Policlinico San Matteo, Viale Camillo Golgi 19, Pavia, PV, 27100, Italy
| |
Collapse
|
7
|
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
|
8
|
Han Z, Li J, Yi X, Zhang T, Liao D, You J, Ai J. Diagnostic accuracy of interleukin-6 in multiple diseases: An umbrella review of meta-analyses. Heliyon 2024; 10:e27769. [PMID: 38515672 PMCID: PMC10955306 DOI: 10.1016/j.heliyon.2024.e27769] [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: 07/18/2023] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 03/23/2024] Open
Abstract
Objective This review aims to conduct a comprehensive study of the diagnostic accuracy of interleukin-6 (IL-6) for multiple diseases by utilizing existing systematic reviews and meta-analyses. Methods We performed a thorough search of Embase, Web of Science, PubMed, and Cochrane Database of Systematic Reviews up to April 2023 to gather meta-analyses that investigate the diagnostic accuracy of IL-6. To assess the methodological quality of the studies, we employed the Assessing the Methodological Quality of Systematic Reviews-2 and Grading of Recommendations, Assessment, Development and Evaluation criteria. Results We included 34 meta-analyses out of the 3024 articles retrieved from the search. These meta-analyses covered 9 categories of diseases of the International Classification of Diseases-11. Studies rated as "Critically Low" or "Very Low" in the quality assessment process were excluded, resulting in a total of 6 meta-analyses that encompassed sepsis, colorectal cancer, tuberculous pleural effusion (TPE), endometriosis, among others. Among these diseases, IL-6 demonstrated a relatively high diagnostic potential in accurately identifying TPE and endometriosis. Conclusions IL-6 exhibited favorable diagnostic accuracy across multiple diseases, suggesting its potential as a reliable diagnostic biomarker in the near future. Substantial evidence supported its high diagnostic accuracy, particularly in the cases of TPE and endometriosis.
Collapse
Affiliation(s)
| | | | | | - Tianyi Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu, 610041, PR China
| | - Dazhou Liao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu, 610041, PR China
| | - Jia You
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu, 610041, PR China
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, 88 South Keyuan Road, Chengdu, 610041, PR China
| |
Collapse
|
9
|
Spadaro S, Jimenez-Santana JD, La Rosa R, Spinazzola G, Argente Navarro P, Volta CA, Scaramuzzo G. Prone Positioning and Molecular Biomarkers in COVID and Non-COVID ARDS: A Narrative Review. J Clin Med 2024; 13:317. [PMID: 38256451 PMCID: PMC10816213 DOI: 10.3390/jcm13020317] [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: 11/29/2023] [Revised: 12/25/2023] [Accepted: 12/29/2023] [Indexed: 01/24/2024] Open
Abstract
Prone positioning (PP) represents a therapeutic intervention with the proven capacity of ameliorating gas exchanges and ventilatory mechanics indicated in acute respiratory distress syndrome (ARDS). When PP is selectively applied to moderate-severe cases of ARDS, it sensitively affects clinical outcomes, including mortality. After the COVID-19 outbreak, clinical application of PP peaked worldwide and was applied in 60% of treated cases, according to large reports. Research on this topic has revealed many physiological underpinnings of PP, focusing on regional ventilation redistribution and the reduction of parenchymal stress and strain. However, there is a lack of evidence on biomarkers behavior in different phases and phenotypes of ARDS. Patients response to PP are, to date, decided on PaO2/FiO2 ratio improvement, whereas scarce data exist on biomarker tracking during PP. The purpose of this review is to explore current evidence on the clinical relevance of biomarkers in the setting of moderate-severe ARDS of different etiologies (i.e., COVID and non-COVID-related ARDS). Moreover, this review focuses on how PP may modulate biomarkers and which biomarkers may have a role in outcome prediction in ARDS patients.
Collapse
Affiliation(s)
- Savino Spadaro
- Department of Translational Medicine, University of Ferrara, 44124 Ferrara, Italy; (R.L.R.); (C.A.V.); (G.S.)
- Anesthesia and Intensive Care Unit, Emergency Department, Azienda Ospedaliera Universitaria di Ferrara, 44124 Ferrara, Italy
| | - Jose Daniel Jimenez-Santana
- Department of Anaesthesiology, Hospital Universitari i Politécnic la Fe, 46026 Valencia, Spain; (J.D.J.-S.); (P.A.N.)
| | - Riccardo La Rosa
- Department of Translational Medicine, University of Ferrara, 44124 Ferrara, Italy; (R.L.R.); (C.A.V.); (G.S.)
- Anesthesia and Intensive Care Unit, Emergency Department, Azienda Ospedaliera Universitaria di Ferrara, 44124 Ferrara, Italy
| | - Giorgia Spinazzola
- Department of Emergency, Anesthesiologic and Reanimation Sciences, Fondazione Policlinico Universitario Gemelli, IRCSS, 00168 Rome, Italy;
| | - Pilar Argente Navarro
- Department of Anaesthesiology, Hospital Universitari i Politécnic la Fe, 46026 Valencia, Spain; (J.D.J.-S.); (P.A.N.)
| | - Carlo Alberto Volta
- Department of Translational Medicine, University of Ferrara, 44124 Ferrara, Italy; (R.L.R.); (C.A.V.); (G.S.)
- Anesthesia and Intensive Care Unit, Emergency Department, Azienda Ospedaliera Universitaria di Ferrara, 44124 Ferrara, Italy
| | - Gaetano Scaramuzzo
- Department of Translational Medicine, University of Ferrara, 44124 Ferrara, Italy; (R.L.R.); (C.A.V.); (G.S.)
- Anesthesia and Intensive Care Unit, Emergency Department, Azienda Ospedaliera Universitaria di Ferrara, 44124 Ferrara, Italy
| |
Collapse
|
10
|
Yang J, Wei A, Wu B, Deng J. Predictive value of combination of lung injury prediction score and receptor for advanced glycation end‑products for the occurrence of acute respiratory distress syndrome. Exp Ther Med 2024; 27:4. [PMID: 38223323 PMCID: PMC10785033 DOI: 10.3892/etm.2023.12291] [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: 08/31/2023] [Accepted: 10/20/2023] [Indexed: 01/16/2024] Open
Abstract
The present study evaluated the predictive value of the combination of the lung injury prediction score (LIPS) and receptor for advanced glycation end-products (RAGE) for the occurrence of acute respiratory distress syndrome (ARDS) in critically ill patients with ARDS risk factors. A total of 551 patients with risk factors of ARDS were divided into an ARDS group and a non-ARDS group. LIPS was computed within 6 h of admission into the ICU, and the plasma concentration of RAGE was detected within 24 h of admission. Multivariate analysis was performed to identify independent associations, and the predictive values for ARDS occurrence were assessed with receiver operating characteristic (ROC) curve. Within 7 days after admission into the ICU, ARDS occurred in 176 patients (31.9%). Multivariate analysis demonstrated that LIPS [odds ratio (OR), 1.282; 95% confidence interval (CI), 1.108-1.604], RAGE levels (OR, 2.359; 95% CI, 1.351-4.813) and Acute Physiology and Chronic Health Evaluation II score (OR, 1.167; 95% CI, 1.074-1.485) were independently associated with ARDS occurrence. ROC curves demonstrated that the area under curve (AUC) of LIPS, RAGE levels and their combination was 0.714 [standard error (SE), 0.023; 95% CI, 0.670-0.759], 0.709 (SE, 0.025; 95% CI, 0.660-0.758) and 0.889 (SE, 0.014; 95% CI, 0.861-0.917), respectively. The AUC of LIPS combined with RAGE levels was significantly higher compared with those of LIPS (0.889 vs. 0.714; Z=6.499; P<0.001) and RAGE (0.889 vs. 0.709; Z=6.282; P<0.001) levels alone. In conclusion, both LIPS and RAGE levels were independently associated with ARDS occurrence in critically ill patients with ARDS risk factors, and had medium predictive values for ARDS occurrence. Combination of LIPS with RAGE levels increased the predictive value for ARDS occurrence.
Collapse
Affiliation(s)
- Jun Yang
- Department of Critical Care Medicine, Chongqing University Jiangjin Hospital, Chongqing 402260, P.R. China
| | - Ai Wei
- Department of Critical Care Medicine, Chongqing University Jiangjin Hospital, Chongqing 402260, P.R. China
| | - Bing Wu
- Department of Critical Care Medicine, Chongqing University Jiangjin Hospital, Chongqing 402260, P.R. China
| | - Jialin Deng
- Department of Nursing, Chongqing University Jiangjin Hospital, Chongqing 402260, P.R. China
| |
Collapse
|
11
|
Brunialti MKC, Leite GGF, Eburneo GS, de Araujo OR, Peçanha-Pietrobom PM, Ferreira PRA, Bellei NCJ, Arakaki JSO, Medina-Pestana J, Requião-Moura L, Salomao R. Patterns of Circulating Cytokines and Vascular Markers' Response in the Presence of COVID-19 in Kidney Transplant Recipients Compared with Non-Transplanted Patients. Viruses 2023; 15:2166. [PMID: 38005844 PMCID: PMC10675241 DOI: 10.3390/v15112166] [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: 08/13/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
COVID-19's severity has been associated with a possible imbalance in the cross-regulation of cytokines and vascular mediators. Since the beginning of the pandemic, kidney transplant recipients (KTRs) have been identified as patients of high vulnerability to more severe diseases. Thus, aiming to describe the patterns of cytokines and vascular mediators and to trace patients' differences according to their KTR status, this prospective study enrolled 67 COVID-19 patients (20 KTRs) and 29 non-COVID-19 controls before vaccination. A panel comprising 17 circulating cytokines and vascular mediators was run on samples collected at different time points. The cytokine and mediator patterns were investigated via principal component analysis (PCA) and correlation-based network (CBN). In both groups, compared to their respective controls, COVID-19 was associated with higher levels of cytokines and vascular mediators. Differentiating between the KTRs and non-KTRs, the number of correlations was much higher in the non-KTRs (44 vs. 14), and the node analysis showed the highest interactions of NGAL and sVCAM-1 in the non-KTRs and KTRs (9 vs. 4), respectively. In the PCA, while the non-KTRs with COVID-19 were differentiated from their controls in their IL-10, IFN-α, and TNF-α, this pattern was marked in the NGAL, sVCAM-1, and IL-8 of the KTRs.
Collapse
Affiliation(s)
- Milena Karina Coló Brunialti
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
| | - Giuseppe G. F. Leite
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
| | - Gabriela Strafolino Eburneo
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
| | - Orlei Ribeiro de Araujo
- Intensive Care Unit, GRAACC, Pediatric Institute of Oncology, Universidade Federal de São Paulo, São Paulo 04039-001, Brazil;
| | - Paula M. Peçanha-Pietrobom
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
| | - Paulo Roberto Abrão Ferreira
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
| | - Nancy C. Junqueira Bellei
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
| | - Jaquelina Sonoe Ota Arakaki
- Division of Respiratory Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04020-050, Brazil;
| | - José Medina-Pestana
- Division of Nephrology, Universidade Federal de São Paulo, São Paulo 04038-031, Brazil;
- Hospital do Rim, Fundação Oswalado Ramos, São Paulo 04038-002, Brazil
| | - Lúcio Requião-Moura
- Division of Nephrology, Universidade Federal de São Paulo, São Paulo 04038-031, Brazil;
- Hospital do Rim, Fundação Oswalado Ramos, São Paulo 04038-002, Brazil
| | - Reinaldo Salomao
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-900, Brazil; (M.K.C.B.); (G.G.F.L.); (G.S.E.); (P.M.P.-P.); (P.R.A.F.); (N.C.J.B.)
- Hospital São Paulo, São Paulo 04024-002, Brazil
| |
Collapse
|
12
|
Papadopoulos D, Skopas V, Trakas N, Papaefstathiou E, Tzogas N, Makris D, Daniil Z, Gourgoulianis K. Serum lactate dehydrogenase and its isoenzymes as predictors of clinical outcomes in acute exacerbation of chronic obstructive pulmonary disease: a retrospective analysis of a hospitalized cohort. Monaldi Arch Chest Dis 2023; 94. [PMID: 37284765 DOI: 10.4081/monaldi.2023.2543] [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: 02/06/2023] [Accepted: 05/25/2023] [Indexed: 06/08/2023] Open
Abstract
We aimed to test the relationship between serum lactate dehydrogenase (LDH) and its isoenzymes and treatment outcomes during hospitalization for acute exacerbations of chronic obstructive pulmonary disease (AECOPD). Thirty-eight AECOPD patients were recruited from a tertiary hospital from December 2017 to June 2018. Serum LDH and LDH isoenzymes were measured on venous blood collected at admission. Treatment outcomes included duration of hospital stay, initiation of mechanical or non-invasive ventilation (NIV), initiation of antipseudomonal antibiotics, change in empirical antibiotic treatment, need for intravenous corticosteroids or methylxanthines, and percentage of change in C-reactive protein from admission to the third day. Multivariate linear and binary logistic regression analyses were used to test the study's objectives. We found that, after adjusting for age, gender, comorbidities, COPD severity, level of hypoxemia, and inflammation markers, each 10 U/L increase in serum LDH was associated with prolongation of the hospital stay by 0.25 (0.03, 0.46) days, 42% higher odds [odds ratio (OR) 1.42 (1.00, 2.03)] for need of NIV, and 25% higher odds [OR 1.25 (1.04, 1.49)] for initiation of antipseudomonal treatment. LDH1 and LDH2 were the LDH isoenzymes that mainly drove these relationships. LDH release in the context of an AECOPD could originate from lung, muscle, or heart tissue damage due to airway inflammation, respiratory muscle recruitment, and myocardial stress. Myocardial injury and aerobic adaptation in respiratory muscles may explain the predominance of LDH1 and LDH2 isoenzymes in these associations.
Collapse
Affiliation(s)
| | - Vlasios Skopas
- 2nd Pulmonary Medicine Department, "Sismanogleion" General Hospital of Attica, Marousi; Respiratory Medicine Department, School of Medicine, University of Thessaly, University Hospital of Larissa.
| | - Nikolaos Trakas
- Clinical Chemistry Department, "Sismanogleion" General Hospital of Attica, Marousi.
| | - Eleni Papaefstathiou
- Clinical Chemistry Department, "Sismanogleion" General Hospital of Attica, Marousi.
| | - Nikolaos Tzogas
- 3rd Pulmonary Medicine Department, Henry Dunant Hospital Center, Athens.
| | - Demosthenes Makris
- Critical Care Department, School of Medicine, University of Thessaly, University Hospital of Larissa.
| | - Zoe Daniil
- Respiratory Medicine Department, School of Medicine, University of Thessaly, University Hospital of Larissa.
| | - Konstantinos Gourgoulianis
- Respiratory Medicine Department, School of Medicine, University of Thessaly, University Hospital of Larissa.
| |
Collapse
|
13
|
Peukert K, Sauer A, Seeliger B, Feuerborn C, Fox M, Schulz S, Wild L, Borger V, Schuss P, Schneider M, Güresir E, Coburn M, Putensen C, Wilhelm C, Bode C. Increased Alveolar Epithelial Damage Markers and Inflammasome-Regulated Cytokines Are Associated with Pulmonary Superinfection in ARDS. J Clin Med 2023; 12:jcm12113649. [PMID: 37297845 DOI: 10.3390/jcm12113649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/09/2023] [Accepted: 05/18/2023] [Indexed: 06/12/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening form of respiratory failure defined by dysregulated immune homeostasis and alveolar epithelial and endothelial damage. Up to 40% of ARDS patients develop pulmonary superinfections, contributing to poor prognosis and increasing mortality. Understanding what renders ARDS patients highly susceptible to pulmonary superinfections is therefore essential. We hypothesized that ARDS patients who develop pulmonary superinfections display a distinct pulmonary injury and pro-inflammatory response pattern. Serum and BALF samples from 52 patients were collected simultaneously within 24 h of ARDS onset. The incidence of pulmonary superinfections was determined retrospectively, and the patients were classified accordingly. Serum concentrations of the epithelial markers soluble receptor for advanced glycation end-products (sRAGE) and surfactant protein D (SP-D) and the endothelial markers vascular endothelial growth factor (VEGF) and angiopoetin-2 (Ang-2) as well as bronchoalveolar lavage fluid concentrations of the pro-inflammatory cytokines interleukin 1ß (IL-1ß), interleukin 18 (IL-18), interleukin 6 (IL-6), and tumor necrosis factor-alpha (TNF-a) were analyzed via multiplex immunoassay. Inflammasome-regulated cytokine IL-18 and the epithelial damage markers SP-D and sRAGE were significantly increased in ARDS patients who developed pulmonary superinfections. In contrast, endothelial markers and inflammasome-independent cytokines did not differ between the groups. The current findings reveal a distinct biomarker pattern that indicates inflammasome activation and alveolar epithelial injury. This pattern may potentially be used in future studies to identify high-risk patients, enabling targeted preventive strategies and personalized treatment approaches.
Collapse
Affiliation(s)
- Konrad Peukert
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Andrea Sauer
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Benjamin Seeliger
- Department of Respiratory Medicine and German Centre of Lung Research (DZL), Hannover Medical School, Carl-Neuberg-Str. 1, 30635 Hannover, Germany
| | - Caroline Feuerborn
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Mario Fox
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Susanne Schulz
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Lennart Wild
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Valeri Borger
- Department of Neurosurgery, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Patrick Schuss
- Department of Neurosurgery, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department of Neurosurgery, BG Klinikum Unfallkrankenhaus Berlin gGmbH, Warener Str. 7, 12683 Berlin, Germany
| | - Matthias Schneider
- Department of Neurosurgery, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Erdem Güresir
- Department of Neurosurgery, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department of Neurosurgery, University Hospital Leipzig, Liebig Str. 20, Haus 4, 04103 Leipzig, Germany
| | - Mark Coburn
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Christian Putensen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Christoph Wilhelm
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Christian Bode
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| |
Collapse
|
14
|
Lin H, Liu Q, Zhao L, Liu Z, Cui H, Li P, Fan H, Guo L. Circulating Pulmonary-Originated Epithelial Biomarkers for Acute Respiratory Distress Syndrome: A Systematic Review and Meta-Analysis. Int J Mol Sci 2023; 24:ijms24076090. [PMID: 37047065 PMCID: PMC10093822 DOI: 10.3390/ijms24076090] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/07/2023] [Accepted: 03/20/2023] [Indexed: 04/14/2023] Open
Abstract
Previous studies have found several biomarkers for acute respiratory distress syndrome (ARDS), but the accuracy of most biomarkers is still in doubt due to the occurrence of other comorbidities. In this systematic review and meta-analysis, we aimed to explore ideal ARDS biomarkers which can reflect pathophysiology features precisely and better identify at-risk patients and predict mortality. Web of Science, PubMed, Embase, OVID, and the Cochrane Library were systematically searched for studies assessing the reliability of pulmonary-originated epithelial proteins in ARDS. A total of 32 studies appeared eligible for meta-analysis, including 2654 ARDS/ALI patients in this study. In the at-risk patients' identification group, the highest pooled effect size was observed in Krebs von den Lungren-6 (KL-6) (SMD: 1.17 [95% CI: 0.55, 1.79]), followed by club cell proteins 16 (CC16) (SMD: 0.74 [95% CI: 0.01, 1.46]), and surfactant proteins-D (SP-D) (SMD: 0.71 [95% CI: 0.57, 0.84]). For the mortality prediction group, CC16 exhibited the largest effect size with SMD of 0.92 (95% CI: 0.42, 1.43). Meanwhile, the summary receiver operating characteristic (SROC) of CC16 for ARDS diagnosis reached an AUC of 0.80 (95% CI: 0.76, 0.83). In conclusion, this study provides a ranking system for pulmonary-originated epithelial biomarkers according to their association with distinguishing at-risk patients and predicting mortality. In addition, the study provides evidence for the advantage of biomarkers over traditional diagnostic criteria. The performance of biomarkers may help to clinically improve the ARDS diagnosis and mortality prediction.
Collapse
Affiliation(s)
- Huishu Lin
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou 325000, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Qisijing Liu
- Research Institute of Public Health, School of Medicine, Nankai University, Tianjin 300381, China
| | - Lei Zhao
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou 325000, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Ziquan Liu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou 325000, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Huanhuan Cui
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou 325000, China
| | - Penghui Li
- School of Environmental Science and Safety Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Haojun Fan
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou 325000, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Liqiong Guo
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China
- Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou 325000, China
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| |
Collapse
|
15
|
Odish M, Pollema T, Meier A, Hepokoski M, Yi C, Spragg R, Patel HH, Alexander LEC, Sun XS, Jain S, Simonson TS, Malhotra A, Owens RL. Very Low Driving-Pressure Ventilation in Patients With COVID-19 Acute Respiratory Distress Syndrome on Extracorporeal Membrane Oxygenation: A Physiologic Study. J Cardiothorac Vasc Anesth 2023; 37:423-431. [PMID: 36567221 PMCID: PMC9701579 DOI: 10.1053/j.jvca.2022.11.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/01/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To determine in patients with acute respiratory distress syndrome (ARDS) on venovenous extracorporeal membrane oxygenation (VV ECMO) whether reducing driving pressure (ΔP) would decrease plasma biomarkers of inflammation and lung injury (interleukin-6 [IL-6], IL-8, and the soluble receptor for advanced glycation end-products sRAGE). DESIGN A single-center prospective physiologic study. SETTING At a single university medical center. PARTICIPANTS Adult patients with severe COVID-19 ARDS on VV ECMO. INTERVENTIONS Participants on VV ECMO had the following biomarkers measured: (1) pre-ECMO with low-tidal-volume ventilation (LTVV), (2) post-ECMO with LTVV, (3) during low-driving-pressure ventilation (LDPV), (4) after 2 hours of very low driving-pressure ventilation (V-LDPV, main intervention ΔP = 1 cmH2O), and (5) 2 hours after returning to LDPV. MAIN MEASUREMENTS AND RESULTS Twenty-six participants were enrolled; 21 underwent V-LDPV. There was no significant change in IL-6, IL-8, and sRAGE from LDPV to V-LDPV and from V-LDPV to LDPV. Only participants (9 of 21) with nonspontaneous breaths had significant change (p < 0.001) in their tidal volumes (Vt) (mean ± SD), 1.9 ± 0.5, 0.1 ± 0.2, and 2.0 ± 0.7 mL/kg predicted body weight (PBW). Participants with spontaneous breathing, Vt were unchanged-4.5 ± 3.1, 4.7 ± 3.1, and 5.6 ± 2.9 mL/kg PBW (p = 0.481 and p = 0.065, respectively). There was no relationship found when accounting for Vt changes and biomarkers. CONCLUSIONS Biomarkers did not significantly change with decreased ΔPs or Vt changes during the first 24 hours post-ECMO. Despite deep sedation, reductions in Vt during V-LDPV were not reliably achieved due to spontaneous breaths. Thus, patients on VV ECMO for ARDS may have higher Vt (ie, transpulmonary pressure) than desired despite low ΔPs or Vt.
Collapse
Affiliation(s)
- Mazen Odish
- UC San Diego Department of Medicine, Division of Pulmonary, Critical Care, Sleep Medicine, and Physiology, La Jolla, CA.
| | - Travis Pollema
- UC San Diego Department of Surgery, Division of Cardiovascular and Thoracic Surgery, La Jolla, CA
| | - Angela Meier
- UC San Diego Department of Anesthesiology, Division of Critical Care, La Jolla, CA
| | - Mark Hepokoski
- UC San Diego Department of Medicine, Division of Pulmonary, Critical Care, Sleep Medicine, and Physiology, La Jolla, CA; VA San Diego Healthcare System, Pulmonary Critical Care Section, San Diego, CA
| | - Cassia Yi
- UC San Diego Health Department of Nursing, La Jolla, CA
| | - Roger Spragg
- UC San Diego Department of Medicine, Division of Pulmonary, Critical Care, Sleep Medicine, and Physiology, La Jolla, CA
| | - Hemal H Patel
- UC San Diego Department of Anesthesiology, Division of Critical Care, La Jolla, CA; VA San Diego Healthcare System, Pulmonary Critical Care Section, San Diego, CA
| | - Laura E Crotty Alexander
- UC San Diego Department of Medicine, Division of Pulmonary, Critical Care, Sleep Medicine, and Physiology, La Jolla, CA; VA San Diego Healthcare System, Pulmonary Critical Care Section, San Diego, CA
| | - Xiaoying Shelly Sun
- UC San Diego, Herbert Wertheim School of Public Health and Human Longevity Science, La Jolla, CA
| | - Sonia Jain
- UC San Diego, Herbert Wertheim School of Public Health and Human Longevity Science, La Jolla, CA
| | - Tatum S Simonson
- UC San Diego Department of Medicine, Division of Pulmonary, Critical Care, Sleep Medicine, and Physiology, La Jolla, CA
| | - Atul Malhotra
- UC San Diego Department of Medicine, Division of Pulmonary, Critical Care, Sleep Medicine, and Physiology, La Jolla, CA
| | - Robert L Owens
- UC San Diego Department of Medicine, Division of Pulmonary, Critical Care, Sleep Medicine, and Physiology, La Jolla, CA
| |
Collapse
|
16
|
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
|
17
|
Almuntashiri S, Jones TW, Wang X, Sikora A, Zhang D. Plasma TIMP-1 as a sex-specific biomarker for acute lung injury. Biol Sex Differ 2022; 13:70. [PMID: 36482481 PMCID: PMC9733313 DOI: 10.1186/s13293-022-00481-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) confers high morbidity and mortality, with a death rate reaching 40%. Pre-clinical and clinical studies have cited sex-specific sex hormones as a critical contributor to divergent immunologic responses. Therefore, exploration of sex and sex hormone roles following lung injury and ARDS development is needed. Tissue inhibitor of metalloproteinase-1 (TIMP-1) was the first-discovered natural collagenase inhibitor and is located exclusively on the X chromosome. This study aimed to evaluate the prognostic role of circulating TIMP-1, and if concentration differences between males and females correlate with the mortality of ARDS patients. METHODS Human plasma samples from 100 ARDS patients enrolled in Albuterol to Treat Acute Lung Injury (ALTA) trial on the day of randomization were evaluated. The amount of TIMP-1 was measured using an enzyme-linked immunoassay (ELISA). Area under the receiver operating characteristic (AUROC) was computed to assess the predictive power of TIMP-1 for 30 and 90-day mortality. Chi-squared tests and Kaplan-Meier curves were computed to assess different variables and survival. RESULTS AUROC analysis of TIMP-1 and 30-day mortality among females showed that TIMP-1 exhibited an AUC of 0.87 (95% confidence interval [CI] 0.78 to 0.97; P = 0.0014) with an optimal cut-off value of 159.7 ng/mL producing a 100% sensitivity and 74% specificity. For 90-day mortality, AUROC analysis showed an AUC of 0.82 (95% confidence interval [CI] 0.67 to 0.97; P = 0.0016) with a similar cut-off value producing a 90% sensitivity and 76.47% specificity. Stratifying subjects by TIMP-1 concentration as high (≥ 159.7 ng/mL) or low (< 159.7 ng/mL) indicated that high TIMP-1 was associated with increased 30 and 90-day mortality rates (all P < 0.0001). Lastly, high TIMP-1 group was associated with worse other outcomes including ventilator-free days (VFDs) and ICU-free days (all P < 0.05). CONCLUSION Circulating TIMP-1 appeared to be a promising biomarker for mortality among females with ARDS. The high TIMP-1 group showed worse VFDs and ICU-free days. Circulating TIMP-1 may be a sex-specific biomarker in the setting of ARDS and could improve ARDS phenotyping as well as provide a novel therapeutic target in females.
Collapse
Affiliation(s)
- Sultan Almuntashiri
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA.,Department of Clinical Pharmacy, College of Pharmacy, University of Hail, Hail, 55473, Saudi Arabia
| | - Timothy W Jones
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
| | - Xiaoyun Wang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
| | - Andrea Sikora
- Department of Clinical and Administrative Pharmacy, College of Pharmacy, University of Georgia, Augusta, GA, 30901, USA.,Department of Pharmacy, Augusta University Medical Center, Augusta, GA, 30912, USA
| | - Duo Zhang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA. .,Department of Medicine, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
18
|
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
|
19
|
Imai R, Yamada D, Tomishima Y, Nakamura T, So C, Ro S, Okafuji K, Kitamura A, Jinta T, Nishimura N. Elevated plasma levels of Krebs von den Lungen-6 and geographic appearance on high-resolution computed tomography are associated with diffuse alveolar damage in autopsy cases of acute respiratory distress syndrome: a retrospective study. BMC Pulm Med 2022; 22:308. [PMID: 35953795 PMCID: PMC9367020 DOI: 10.1186/s12890-022-02102-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/05/2022] [Indexed: 11/25/2022] Open
Abstract
Background Although diffuse alveolar damage (DAD) is a histopathological hallmark of acute respiratory distress syndrome (ARDS), its detection without lung biopsy is challenging. In patients with ARDS, the specificity of the Berlin definition to diagnose DAD as a reference standard is not adequately high, making it difficult to adequately diagnose DAD. The purpose of this study was to investigate the relationship between DAD and clinical findings, including KL-6 and geographic appearance, in ARDS patients and to identify more specific diagnostic criteria for DAD. Methods Among all adult autopsy cases at a tertiary hospital in Japan between January 2006 and March 2021, patients with ARDS who met the Berlin definition criteria were included. The patients’ conditions were classified according to histopathological patterns as DAD or non-DAD, and clinical characteristics, laboratory data, and high-resolution computed tomography (HRCT) findings were compared between the two groups. Results During the study period, 27 met the Berlin definition (median age: 79 years, 19 men), of whom 18 (67%) had DAD and 9 (33%) did not. In the non-DAD group, histopathologic findings revealed organizing pneumonia in seven patients and pulmonary hemorrhage in two patients. On HRCT at onset, patients with DAD had more geographic appearance than those without DAD (89% vs. 44%). In patients with geographic appearance and elevated KL-6 (> 500 U/mL), the sensitivity and specificity for DAD diagnosis were 56% and 100%, respectively. All three patients with no geographic appearance and normal KL-6 did not have DAD. Conclusions Geographic appearance on HRCT combined with KL-6 levels may predict the presence of DAD in patients with ARDS.
Collapse
Affiliation(s)
- Ryosuke Imai
- Department of Pulmonary Medicine, Thoracic Center, St. Luke's International Hospital, 9-1, Akashi-cho, Chuo City, Tokyo, 104-8560, Japan.
| | - Daisuke Yamada
- Department of Radiology, St. Luke's International Hospital, Tokyo, Japan
| | - Yutaka Tomishima
- Department of Pulmonary Medicine, Thoracic Center, St. Luke's International Hospital, 9-1, Akashi-cho, Chuo City, Tokyo, 104-8560, Japan
| | - Tomoaki Nakamura
- Department of Pulmonary Medicine, Thoracic Center, St. Luke's International Hospital, 9-1, Akashi-cho, Chuo City, Tokyo, 104-8560, Japan
| | - Clara So
- Department of Pulmonary Medicine, Thoracic Center, St. Luke's International Hospital, 9-1, Akashi-cho, Chuo City, Tokyo, 104-8560, Japan
| | - Shosei Ro
- Department of Pulmonary Medicine, Thoracic Center, St. Luke's International Hospital, 9-1, Akashi-cho, Chuo City, Tokyo, 104-8560, Japan
| | - Kohei Okafuji
- Department of Pulmonary Medicine, Thoracic Center, St. Luke's International Hospital, 9-1, Akashi-cho, Chuo City, Tokyo, 104-8560, Japan
| | - Atsushi Kitamura
- Department of Pulmonary Medicine, Thoracic Center, St. Luke's International Hospital, 9-1, Akashi-cho, Chuo City, Tokyo, 104-8560, Japan
| | - Torahiko Jinta
- Department of Pulmonary Medicine, Thoracic Center, St. Luke's International Hospital, 9-1, Akashi-cho, Chuo City, Tokyo, 104-8560, Japan
| | - Naoki Nishimura
- Department of Pulmonary Medicine, Thoracic Center, St. Luke's International Hospital, 9-1, Akashi-cho, Chuo City, Tokyo, 104-8560, Japan
| |
Collapse
|
20
|
Cheng J, Yang J, Ma A, Dong M, Yang J, Wang P, Xue Y, Zhou Y, Kang Y. The Effects of Airway Pressure Release Ventilation on Pulmonary Permeability in Severe Acute Respiratory Distress Syndrome Pig Models. Front Physiol 2022; 13:927507. [PMID: 35936889 PMCID: PMC9354663 DOI: 10.3389/fphys.2022.927507] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: The aim of the study was to compare the effects of APRV and LTV ventilation on pulmonary permeability in severe ARDS.Methods: Mini Bama adult pigs were randomized into the APRV group (n = 5) and LTV group (n = 5). A severe ARDS animal model was induced by the whole lung saline lavage. Pigs were ventilated and monitored continuously for 48 h.Results: Compared with the LTV group, CStat was significantly better (p < 0.05), and the PaO2/FiO2 ratio showed a trend to be higher throughout the period of the experiment in the APRV group. The extravascular lung water index and pulmonary vascular permeability index showed a trend to be lower in the APRV group. APRV also significantly mitigates lung histopathologic injury determined by the lung histopathological injury score (p < 0.05) and gross pathological changes of lung tissues. The protein contents of occludin (p < 0.05), claudin-5 (p < 0.05), E-cadherin (p < 0.05), and VE-cadherin (p < 0.05) in the middle lobe of the right lung were higher in the APRV group than in the LTV group; among them, the contents of occludin (p < 0.05) and E-cadherin (p < 0.05) of the whole lung were higher in the APRV group. Transmission electron microscopy showed that alveolar–capillary barrier damage was more severe in the middle lobe of lungs in the LTV group.Conclusion: In comparison with LTV, APRV could preserve the alveolar–capillary barrier architecture, mitigate lung histopathologic injury, increase the expression of cell junction protein, improve respiratory system compliance, and showed a trend to reduce extravascular lung water and improve oxygenation. These findings indicated that APRV might lead to more profound beneficial effects on the integrity of the alveolar–capillary barrier architecture and on the expression of biomarkers related to pulmonary permeability.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yan Kang
- *Correspondence: Yongfang Zhou, ; Yan Kang,
| |
Collapse
|
21
|
Zhan B, Shen J. Mitochondria and their potential role in acute lung injury (Review). Exp Ther Med 2022; 24:479. [PMID: 35761815 PMCID: PMC9214601 DOI: 10.3892/etm.2022.11406] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/16/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Biao Zhan
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai 201508, P.R. China
| | - Jie Shen
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai 201508, P.R. China
| |
Collapse
|
22
|
Luo J, Liang J, Wang S, Huang S, Zhou L, Shi Y, Zhang J, Wang Y, Wu BQ, Li L. Serum human epididymis secretory protein 4 correlates with sepsis-associated acute respiratory distress syndrome and 28-day mortality in critically ill patients. Ann Clin Biochem 2022; 59:338-346. [PMID: 35549539 DOI: 10.1177/00045632221103805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a severe disease with high mortality, and its primary cause is sepsis. The aim of this study was to detect and evaluate the role of Human epididymis protein 4 (HE4) in sepsis-related ARDS. METHODS One hundred and twenty-three critically ill sepsis patients with/without ARDS and 102 healthy controls were enrolled in this study. Blood samples were collected upon admission for quantitative testing of HE4 by chemiluminescent microparticle immunoassay (CMIA). ROC curve analysis and Spearman's correlation analysis were conducted to determine the diagnostic and prognostic value of HE4. RESULTS Compared with controls, the serum HE4 concentrations of sepsis patients were elevated, and levels in sepsis patients with ARDS were significantly higher (all p < 0.0001). Moreover, HE4 concentrations were strongly correlated with the clinical severity characteristics of sepsis patients, and ROC curve suggested that the AUC of HE4 applied to discriminate sepsis-ARDS patients from sepsis patients was 0.903. HE4 was also found to be a prognostic biomarker of clinical severity and 28-day mortality among critically ill sepsis patients. Logistic regression analysis showed that HE4 was an independent factor for diagnosis of ARDS. Meanwhile, ROC curve analysis showed that the cut-off value of serum HE4 to discriminate 28-day mortality from sepsis patients (AUC: 0.782) was 646.5 pmol/L. CONCLUSIONS The concentration of serum HE4 in patients with sepsis-related ARDS was markedly increased and was significantly correlated with mortality, which suggests that serum HE4 could be a promising diagnostic and prognostic biomarker for ARDS in sepsis patients.
Collapse
Affiliation(s)
- Jinmei Luo
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, 144991The Third Affiliated Hospital of Sun Yat-sen University, People's Republic of China
| | - Jingjing Liang
- Department of Emergency, 144991The Third Affiliated Hospital of Sun Yat-sen University, People's Republic of China
| | - Shaofang Wang
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, 144991The Third Affiliated Hospital of Sun Yat-sen University, People's Republic of China
| | - Shaoqiong Huang
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, 144991The Third Affiliated Hospital of Sun Yat-sen University, People's Republic of China
| | - Laizhi Zhou
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, 144991The Third Affiliated Hospital of Sun Yat-sen University, People's Republic of China
| | - Yunfeng Shi
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, 144991The Third Affiliated Hospital of Sun Yat-sen University, People's Republic of China
| | - Jingcong Zhang
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, 144991The Third Affiliated Hospital of Sun Yat-sen University, People's Republic of China
| | - Yanhong Wang
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, 144991The Third Affiliated Hospital of Sun Yat-sen University, People's Republic of China
| | - Ben-Quan Wu
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, 144991The Third Affiliated Hospital of Sun Yat-sen University, People's Republic of China
| | - Laisheng Li
- Department of Laboratory Medicine, 71068The First Affiliated Hospital of Sun Yat-sen University, People's Republic of China
| |
Collapse
|
23
|
Gu ZY, Liu WJ, Huang DL, Liu YJ, He HY, Yang C, Liu YM, Xu M, Rong RM, Zhu DM, Luo Z, Ju MJ. Preliminary Study on the Combination Effect of Clindamycin and Low Dose Trimethoprim-Sulfamethoxazole on Severe Pneumocystis Pneumonia After Renal Transplantation. Front Med (Lausanne) 2022; 9:827850. [PMID: 35602475 PMCID: PMC9120531 DOI: 10.3389/fmed.2022.827850] [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] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/11/2022] [Indexed: 11/15/2022] Open
Abstract
Objective Evaluate the effect of the combination of clindamycin with low-dose trimethoprim-sulfamethoxazole (TMP/SMX) regimen on sever Pneumocystis pneumonia (PCP) after renal transplantation. Method 20 severe PCP patients after renal transplantation were included in this historical-control, retrospective study. A 10 patients were treated with the standard dose of TMP/SMX (T group), the other 10 patients were treated with the combination of clindamycin and low dose TMP/SMX (CT group). Results Although there was no significant difference in the hospital survival between the two groups, the CT protocol improved the PaO2/FiO2 ratio more significantly and rapidly after the 6th ICU day (1.51 vs. 0.38, P = 0.014). CT protocol also ameliorated the pulmonary infiltration and the lactate dehydrogenase level more effectively. Moreover, the CT protocol reduced the incidence of pneumomediastinum (0 vs. 50%, P = 0.008), the length of hospital staying (26.5 vs. 39.0 days, P = 0.011) and ICU staying (12.5 vs. 22.5 days, P = 0.008). Furthermore, more thrombocytopenia (9/10 vs. 3/10, P = 0.020) was emerged in the T group than in the CT group. The total adverse reaction rate was much lower in the CT group than in the T group (8/80 vs. 27/80, P < 0.001). Consequently, the dosage of TMP/SMX was reduced in 8 patients, while only 2 patients in the CT group received TMP/SMX decrement (P = 0.023). Conclusion The current study proposed that clindamycin combined with low-dose TMP/SMX was more effective and safer the than single use of TMP/SMX for severe PCP patients after renal transplantation (NCT04328688).
Collapse
Affiliation(s)
- Zhun-Yong Gu
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wen-Jun Liu
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dan-Lei Huang
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu-Jing Liu
- Department of Nursing, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hong-Yu He
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng Yang
- Department of Urology Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yi-Mei Liu
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ming Xu
- Department of Urology Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rui-Ming Rong
- Department of Urology Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Du-Ming Zhu
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhe Luo
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Min-Jie Ju
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Matsui M, Makimoto A, Nishio N, Takahashi Y, Urashima M, Yuza Y. Predictive factors of acute respiratory events during initial induction chemotherapy in patients with advanced neuroblastoma. Cancer Rep (Hoboken) 2022; 5:e1499. [PMID: 34255936 PMCID: PMC9124507 DOI: 10.1002/cnr2.1499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 06/15/2021] [Accepted: 06/22/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Acute respiratory events (ARE) occasionally occur during induction chemotherapy as a complication in patients with advanced neuroblastoma. AIMS The present study aimed to identify the predictive factors of ARE, defined as severe hypoxia, during initial induction chemotherapy in patients with newly diagnosed advanced neuroblastoma. METHODS AND RESULTS The medical records of 75 consecutive patients in whom stage III or IV neuroblastoma was newly diagnosed between January 2003 and December 2018 at two medical institutions were retrospectively reviewed. The outcome was ARE, which were assessed by measuring oxygen saturation between days 1 and 14 of initial induction chemotherapy. Severe hypoxia was defined as grade 3 or higher according to the Common Terminology Criteria for Adverse Events version 4 (CTCAE v4.0) or decreased oxygen saturation at rest (e.g., pulse oximeter <88% or PaO2 ≤55 mmHg). Possible predictive factors on admission were first screened for using univariate analyses with P = .05, then models of the predictive power of the outcome were evaluated by generating receiver operating characteristic (ROC) curves. Eleven patients (14.7%) had the outcome, including three (4.0%) who required respiratory support in the intensive care unit. The area under the curve of the ROC for the predictive factors screened by univariate analyses was 0.84 (95% confidence interval [CI]: 0.73-0.95) for lactate dehydrogenase (LDH) and 0.90 (95% CI: 0.82-0.98) for the disseminated intravascular coagulation (DIC) score. CONCLUSION The LDH value and DIC score on admission may be clinically useful predictors of ARE during initial induction chemotherapy in patients with advanced neuroblastoma.
Collapse
Affiliation(s)
- Motohiro Matsui
- Department of Hematology/OncologyTokyo Metropolitan Children's Medical CenterTokyoJapan
- Division of Molecular EpidemiologyJikei University School of MedicineTokyoJapan
| | - Atsushi Makimoto
- Department of Hematology/OncologyTokyo Metropolitan Children's Medical CenterTokyoJapan
| | - Nobuhiro Nishio
- Department of PediatricsNagoya University Graduate School of MedicineNagoyaJapan
| | - Yoshiyuki Takahashi
- Department of PediatricsNagoya University Graduate School of MedicineNagoyaJapan
| | - Mitsuyoshi Urashima
- Division of Molecular EpidemiologyJikei University School of MedicineTokyoJapan
| | - Yuki Yuza
- Department of Hematology/OncologyTokyo Metropolitan Children's Medical CenterTokyoJapan
| |
Collapse
|
25
|
Wang R, Dai H. Association of platelet count with all-cause mortality from acute respiratory distress syndrome: A cohort study. J Clin Lab Anal 2022; 36:e24378. [PMID: 35358347 PMCID: PMC9102613 DOI: 10.1002/jcla.24378] [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: 03/05/2022] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
Background The purpose of this study was to investigate whether platelet count was associated with mortality in acute respiratory distress syndrome (ARDS) patients. Methods We analyzed patients with ARDS from Multi‐parameter Intelligent Monitoring in Intensive Care Database III (MIMIC‐III). Platelet count was measured at the time of intensive care unit (ICU) admission. The cox proportional hazard model and subgroup analysis were used to determine the relationship between the platelet count and mortality of ARDS, as well as the consistency of its association. The primary outcome of this study was 365‐day mortality from the date of ICU admission. Result This study enrolled a total of 395 critically ill patients with ARDS. After adjustment for age, gender and ethnicity, the multivariate cox regression model showed that the hazard ratios (HRs) (95% confidence intervals [CIs]) of platelet count <192 × 109/L and >296 × 109/L were 2.08 (1.43, 3.04) and 1.35 (0.91, 2.01), respectively, compared with the reference (192–296 ×109/L). After adjusting for confounding factors, lower platelet count (<192 × 109/L) was associated with increased mortality (adjusted HR, 1.71; 95% CI 1.06–2.76, p = 0.0284). However, there was no similar trend in the 30‐day (adjusted HR,1.02; 95% CI 0.54–1.94) or 90‐day (adjusted HR, 1.65; 95% CI 0.94–2.89) mortality. In the subgroup analysis, lower platelet count showed significant interactions with specific populations (p interaction = 0.0413), especially in patients with atrial fibrillation. Conclusion Taken together, our analysis showed that platelet count is an independent predictor of mortality in critically ill patients with ARDS.
Collapse
Affiliation(s)
- Rennv Wang
- Emergency Department, Affiliated Zhejiang Hospital of Zhejiang University School of Medical, Hangzhou, Zhejiang, China
| | - Haiwen Dai
- Emergency Department, Affiliated Zhejiang Hospital of Zhejiang University School of Medical, Hangzhou, Zhejiang, China
| |
Collapse
|
26
|
Alipanah N, Calfee CS. Phenotyping in acute respiratory distress syndrome: state of the art and clinical implications. Curr Opin Crit Care 2022; 28:1-8. [PMID: 34670998 PMCID: PMC8782441 DOI: 10.1097/mcc.0000000000000903] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE OF REVIEW Decades of research in acute respiratory distress syndrome (ARDS) have led to few interventions that impact clinical outcomes. The pandemic of patients with ARDS due to the novel SARS-CoV-2 infection has stressed the need for more effective therapies in ARDS. Phenotyping may enable successful trials and precision therapeutics in this patient population. RECENT FINDINGS Clinical phenotypes that group patients by shared cause, time-course or radiographic presentation are of prognostic value, but their use is limited by misclassification. Physiological phenotypes, including the P/F ratio, ventilatory ratio and dead space fraction, predict poor outcomes but can rapidly change, making them unstable over time. Biologic phenotypes have prognostic value with composite clinical and biomarker sub-phenotypes additionally impacting treatment response but are yet to be prospectively validated. SUMMARY Although much progress has been made in ARDS phenotyping, implementation of precision medicine practices will depend on conducting phenotype-aware trials using rapid point of care assays or machine learning algorithms. Omics studies will enhance our understanding of biologic determinants of clinical outcomes in ARDS sub-phenotypes. Whether biologic ARDS sub-phenotypes are specific to this syndrome or rather more broadly identify endotypes of critical illness remains to be determined.
Collapse
Affiliation(s)
- Narges Alipanah
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco
| | - Carolyn S. Calfee
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco
- Department of Anesthesia, University of California San Francisco
| |
Collapse
|
27
|
Tu C, Wang Z, Xiang E, Zhang Q, Zhang Y, Wu P, Li C, Wu D. Human Umbilical Cord Mesenchymal Stem Cells Promote Macrophage PD-L1 Expression and Attenuate Acute Lung Injury in Mice. Curr Stem Cell Res Ther 2022; 17:564-575. [PMID: 35086457 DOI: 10.2174/1574888x17666220127110332] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/11/2021] [Accepted: 12/19/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) remains a serious clinical problem but has no approved pharmacotherapy. Mesenchymal stem cells (MSCs) represent an attractive therapeutic tool for tissue damage and inflammation owing to their unique immunomodulatory properties. The present study aims to explore the therapeutic effect and underlying mechanisms of human umbilical cord MSCs (UC-MSCs) in ALI mice. OBJECTIVE In this study, we identify a novel mechanism for human umbilical cord-derived MSCs (UC-MSCs)-mediated immunomodulation through PGE2-dependent reprogramming of host macrophages to promote their PD-L1 expression. Our study suggests that UC-MSCs or primed-UC-MSCs offer new therapeutic approaches for lung inflammatory diseases. METHODS Lipopolysaccharide (LPS)-induced ALI mice were injected with 5×105 UC-MSCs via the tail vein after 4 hours of LPS exposure. After 24 hours of UC-MSC administration, the total protein concentration and cell number in the bronchoalveolar lavage fluid (BALF), and cytokine levels in the lung tissue were measured. Lung pathological changes and macrophage infiltration after UC-MSC treatment were analyzed. Moreover, in vitro co-culture experiments were performed to analyze cytokine levels of RAW264.7 cells and Jurkat T cells. RESULTS UC-MSC treatment significantly improved LPS-induced ALI, as indicated by decreased total protein exudation concentration and cell number in BALF, and reduced pathological damage in ALI mice. UC-MSCs could inhibit pro-inflammatory cytokine levels (IL-1β, TNF-α, MCP-1, IL-2, and IFN-γ), whereas enhancing anti-inflammatory cytokine IL-10 expression, as well as reduced macrophage infiltration into the injured lung tissue. Importantly, UC-MSC administration increased programmed cell death protein ligand 1 (PD-L1) expression in the lung macrophages. Mechanistically, UC-MSCs upregulated cyclooxygenase-2 (COX2) expression and prostaglandin E2 (PGE2) secretion in response to LPS stimulation. UC-MSCs reduced the inflammatory cytokine levels in murine macrophage Raw264.7 through the COX2/PGE2 axis. Furthermore, UC-MSC-derived PGE2 enhanced PD-L1 expression in RAW264.7 cells, which in turn promoted programmed cell death protein 1 (PD-1) expression and reduced IL-2 and IFN-γ production in Jurkat T cells. CONCLUSION Our results suggest that UC-MSCs attenuate ALI via PGE2-dependent reprogramming of macrophages to promote their PD-L1 expression.
Collapse
Affiliation(s)
- Chengshu Tu
- Department of Pathophysiology, Tongji Medical College, Huazhong Science and Technology University, Wuhan, China
| | | | - E Xiang
- Wuhan Hamilton Biotechnology-Co., Ltd, Wuhan, China
- Department of Biochemistry and Molecular Biology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Quan Zhang
- Wuhan Hamilton Biotechnology-Co., Ltd, Wuhan, China
| | - Yaqi Zhang
- Wuhan Hamilton Biotechnology-Co., Ltd, Wuhan, China
- Department of Biochemistry and Molecular Biology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Ping Wu
- Department of Pathophysiology, Tongji Medical College, Huazhong Science and Technology University, Wuhan, China
| | - Changyong Li
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Dongcheng Wu
- Wuhan Hamilton Biotechnology-Co., Ltd, Wuhan, China
- Department of Biochemistry and Molecular Biology, Wuhan University School of Basic Medical Sciences, Wuhan, China
- Guangzhou Hamilton Biotechnology-Co., Ltd, Guangzhou, China
| |
Collapse
|
28
|
Hussain M, Khurram Syed S, Fatima M, Shaukat S, Saadullah M, Alqahtani AM, Alqahtani T, Bin Emran T, Alamri AH, Barkat MQ, Wu X. Acute Respiratory Distress Syndrome and COVID-19: A Literature Review. J Inflamm Res 2022; 14:7225-7242. [PMID: 34992415 PMCID: PMC8710428 DOI: 10.2147/jir.s334043] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is an overwhelming inflammatory disorder of the lung due to direct and indirect insults to the lungs. ARDS is characterized by increased vascular permeability, protein-rich edema, diffuse alveolar infiltrate, and loss of aerated lung tissue, leading to decreased lung compliance, tachypnea, and severe hypoxemia. COVID-19 is generally associated with ARDS, and it has gained prime importance since it started. The mortality rate is alarmingly high in COVID-19-related ARDS patients regardless of advances in mechanical ventilation. Several pharmacological agents, including corticosteroids, nitric oxide, neuromuscular blocker, anti-TNF, statins, and exogenous surfactant, have been studied and some are under investigation, like ketoconazole, lisofylline, N-acetylcysteine, prostaglandins, prostacyclin, and fish oil. The purpose of this review is to appraise the understanding of the pathophysiology of ARDS, biomarkers, and clinical trials of pharmacological therapies of ARDS and COVID-19-related ARDS.
Collapse
Affiliation(s)
- Musaddique Hussain
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Shahzada Khurram Syed
- Department of Basic Medical Sciences, School of Health Sciences, University of Management and Technology Lahore, Lahore, 54000, Pakistan
| | - Mobeen Fatima
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Saira Shaukat
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| | - Malik Saadullah
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, 38000, Pakistan
| | - Ali M Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Taha Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh
| | - Ali H Alamri
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Muhammad Qasim Barkat
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City, 310000, People's Republic of China
| | - Ximei Wu
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City, 310000, People's Republic of China
| |
Collapse
|
29
|
Masumoto A, Kitai T, Matsumoto S, Kuroda S, Kohsaka S, Tachikawa R, Seo R, Doi A, Tomii K, Yonetsu T, Torii S, Komuro I, Hirata KI, Node K, Matsue Y, Furukawa Y. Impact of serum lactate dehydrogenase on the short-term prognosis of COVID-19 with pre-existing cardiovascular diseases. J Cardiol 2021; 79:501-508. [PMID: 35000825 PMCID: PMC8712257 DOI: 10.1016/j.jjcc.2021.12.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/28/2021] [Accepted: 12/19/2021] [Indexed: 12/25/2022]
Abstract
Background Patients with coronavirus disease 2019 (COVID-19) and underlying cardiovascular comorbidities have poor prognoses. Our aim was to identify the impact of serum lactate dehydrogenase (LDH), which is associated with mortality in acute respiratory distress syndrome, on the prognoses of patients with COVID-19 and underlying cardiovascular comorbidities. Methods Among 1518 patients hospitalized with COVID-19 enrolled in the CLAVIS-COVID (Clinical Outcomes of COVID-19 Infection in Hospitalized Patients with Cardiovascular Diseases and/or Risk Factors study), 515 patients with cardiovascular comorbidities were analyzed. Patients were divided into tertiles based on LDH levels at admission [tertile 1 (T1), <235 U/L; tertile 2 (T2), 235–355 U/L; and tertile 3 (T3); ≥356 U/L]. We investigated the impact of LDH levels on the in-hospital mortality. Results The mean age was 70.4 ± 30.0 years, and 65.3% were male. There were significantly more in-hospital deaths in T3 than in T1 and T2 [n = 50 (29.2%) vs. n = 15 (8.7%), and n = 24 (14.0%), respectively; p < 0.001]. Multivariable analysis adjusted for age, comorbidities, vital signs, and laboratory data including D-dimer and high-sensitivity troponin showed T3 was associated with an increased risk of in-hospital mortality (adjusted hazard ratio, 3.04; 95% confidence interval, 1.50–6.13; p = 0.002). Conclusions High serum LDH levels at the time of admission are associated with an increased risk of in-hospital death in patients with COVID-19 and known cardiovascular disease and may aid in triage of these patients.
Collapse
Affiliation(s)
- Akiko Masumoto
- Department of Cardiovascular Medicine, Kobe City Medical Center General Hospital, Kobe, Japan; Division of Cardiovascular Medicine, Hyogo Brain and Heart Center, Himeji, Japan
| | - Takeshi Kitai
- Department of Cardiovascular Medicine, Kobe City Medical Center General Hospital, Kobe, Japan; Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Medicine Center, 6-1 Kishibe-shinmachi, Osaka, Japan.
| | - Shingo Matsumoto
- Department of Cardiovascular Medicine, Department of Internal Medicine, Toho University Faculty of Medicine, Tokyo, Japan
| | - Shunsuke Kuroda
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, United States
| | - Shun Kohsaka
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Ryo Tachikawa
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Ryutaro Seo
- Department of Intensive Care, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Asako Doi
- Department of Infectious Diseases, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Keisuke Tomii
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Taishi Yonetsu
- Department of Interventional Cardiology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sho Torii
- Department of Cardiology, Tokai University School of Medicine, Isehara, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Ken-Ichi Hirata
- Department of Internal Medicine, Division of Cardiovascular Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Koichi Node
- Department of Cardiovascular Medicine, Saga University, Saga, Japan
| | - Yuya Matsue
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan; Cardiovascular Respiratory Sleep Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yutaka Furukawa
- Department of Cardiovascular Medicine, Kobe City Medical Center General Hospital, Kobe, Japan
| |
Collapse
|
30
|
Liu Z, Liu D, Wang Z, Zou Y, Wang H, Li X, Zheng D, Zhou G. Association between inflammatory biomarkers and acute respiratory distress syndrome or acute lung injury risk : A systematic review and meta-analysis. Wien Klin Wochenschr 2021; 134:24-38. [PMID: 34860273 PMCID: PMC8813738 DOI: 10.1007/s00508-021-01971-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 10/16/2021] [Indexed: 11/29/2022]
Abstract
Background The relationship between acute respiratory distress syndrome (ARDS)/acute lung injury (ALI) and levels of certain inflammatory factors remains controversial. The purpose of this meta-analysis was to summarize the available studies evaluating the association between levels of inflammatory factors and ARDS/ALI incidence. Methods We searched the PubMed, EmBase, and Cochrane databases for studies published up to July 2017. For each inflammatory factor, a random effects model was employed to pool results from different studies. Results We identified 63 studies that included 6243 patients in our meta-analysis. Overall, the results indicated that the levels of angiopoietin (ANG)-2 (standard mean difference, SMD: 1.34; P < 0.001), interleukin (IL)-1β (SMD: 0.92; P = 0.012), IL‑6 (SMD: 0.66; P = 0.005), and tumor necrosis factor (TNF)-α (SMD: 0.98; P = 0.001) were significantly higher in patients with ARDS/ALI than in unaffected individuals. No significant differences were observed between patients with ARDS/ALI and unaffected individuals in terms of the levels of IL‑8 (SMD: 0.61; P = 0.159), IL-10 (SMD: 1.10; P = 0.231), and plasminogen activator inhibitor (PAI)-1 (SMD: 0.70; P = 0.060). Conclusions ARDS/ALI is associated with a significantly elevated levels of ANG‑2, IL-1β, IL‑6, and TNF‑α, but not with IL‑8, IL-10, and PAI‑1 levels. Supplementary Information The online version of this article (10.1007/s00508-021-01971-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhenfeng Liu
- Department of Respiratory Medicine, Zunyi Honghuagang District People's Hospital, 185 Wanli Road, HongHuagang District, 563000, Guizhou, China.,Department of Respiratory Medicine, the Third Affiliated Hospital of Zunyi Medical University, 98 Fenghuang Road, Huichuan District, 563000, Guizhou, China
| | - Daishun Liu
- Department of Respiratory Medicine, the Third Affiliated Hospital of Zunyi Medical University, 98 Fenghuang Road, Huichuan District, 563000, Guizhou, China
| | - Zhihua Wang
- Department of Respiratory Medicine, Teaching Hospital of Zunyi Medical College, 134 LinJiapo Road, HongHuagang District, 563000, Guizhou, China
| | - Yugang Zou
- Department of Respiratory Medicine, the Third Affiliated Hospital of Zunyi Medical University, 98 Fenghuang Road, Huichuan District, 563000, Guizhou, China
| | - Haixia Wang
- Department of Respiratory Medicine, Suzhou Science & Technology Town Hospital, 215153, Jiangsu, China
| | - Xiao Li
- Department of Respiratory Medicine, Teaching Hospital of Zunyi Medical College, 134 LinJiapo Road, HongHuagang District, 563000, Guizhou, China
| | - Deliang Zheng
- Department of Respiratory Medicine, Teaching Hospital of Zunyi Medical College, 134 LinJiapo Road, HongHuagang District, 563000, Guizhou, China
| | - Guoqi Zhou
- Department of Respiratory Medicine, Teaching Hospital of Zunyi Medical College, 134 LinJiapo Road, HongHuagang District, 563000, Guizhou, China.
| |
Collapse
|
31
|
Das KM, Singh R, Al Dossari K, Subramanya S, Ojha SK, AlMansoori T, Alkoteesh JA. Chest radiographic score and lactate dehydrogenase are independent risk factors linked to mortality in Middle East Respiratory Syndrome Coronavirus (MERS-CoV) patients. THE EGYPTIAN JOURNAL OF RADIOLOGY AND NUCLEAR MEDICINE 2021. [PMCID: PMC8491180 DOI: 10.1186/s43055-021-00635-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Background Despite the dominance of Covid-19 in the current situation, MERS-CoV is found infrequently in the Middle East. When coupled with the chest radiographic score, serum biochemical parameters may be utilized to assess serum biochemical changes in individuals with different degrees of MERS-CoV infection and to predict death. The purpose of this study was to examine the association between increased LDH levels and severe MERS-CoV outcomes utilizing ventilation days and an elevated chest radiographic score. Results Fifty-seven patients were included in the retrospective cohort. The mean age was 44.9 ± 13.5 years, while the range was between 12 and 73 years. With an average age of 53.3 ± 16.5 years, 18 of 57 (31.6%) patients were classified as deceased. The deceased group showed a substantially greater amount of LDH than the recovery group (280.18 ± 150.79 vs. 1241.72 ± 1327.77, p = 0.007). A cut-off value of > 512 LDH was established with a C-statistic of 0.96 (95% CI 0.92–1.00) and was 94% sensitive and 93% specific for mortality. Multivariate cox regression analysis revealed that loge (LDH) (adjusted HR: 9.91, 95% CI: 2.44–40.3, p = 0.001) and chest radiographic score (adjusted HR: 1.24, 95% CI: 1.05–1.47, p = 0.01) were risk factors for mortality, whereas ventilation days were a protective factor (adjusted HR: 0.84, 95% CI: 0.76–0.93, p = 0.001). Conclusion According to our results, blood LDH levels of > 512 had a 94% sensitivity and 93% specificity for predicting in-hospital mortality in patients infected with MERS-CoV. The chest radiographic score of 11.34 ± 5.4 was the risk factor for the mortality (adjusted Hazard ratio HR: 1.24, 95% CI: 1.05–1.47, p = 0.01). Thus, threshold may aid in the identification of individuals with MERS-CoV infection who die in hospital.
Collapse
|
32
|
Clinical and biological markers for predicting ARDS and outcome in septic patients. Sci Rep 2021; 11:22702. [PMID: 34811434 PMCID: PMC8608812 DOI: 10.1038/s41598-021-02100-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/08/2021] [Indexed: 12/25/2022] Open
Abstract
Sepsis is a common cause of acute respiratory distress syndrome (ARDS) associated with a high mortality. A panel of biomarkers (BMs) to identify septic patients at risk for developing ARDS, or at high risk of death, would be of interest for selecting patients for therapeutic trials, which could improve ARDS diagnosis and treatment, and survival chances in sepsis and ARDS. We measured nine protein BMs by ELISA in serum from 232 adult septic patients at diagnosis (152 required invasive mechanical ventilation and 72 had ARDS). A panel including the BMs RAGE, CXCL16 and Ang-2, plus PaO2/FiO2, was good in predicting ARDS (area under the curve = 0.88 in total septic patients). Best performing panels for ICU death are related to the presence of ARDS, need for invasive mechanical ventilation, and pulmonary/extrapulmonary origin of sepsis. In all cases, the use of BMs improved the prediction by clinical markers. Our study confirms the relevance of RAGE, Ang-2, IL-1RA and SP-D, and is novel supporting the inclusion of CXCL16, in BMs panels for predicting ARDS diagnosis and ARDS and sepsis outcome.
Collapse
|
33
|
Jadaun PK, Chatterjee S. COVID-19 and dys-regulation of pulmonary endothelium: implications for vascular remodeling. Cytokine Growth Factor Rev 2021; 63:69-77. [PMID: 34728151 PMCID: PMC9611904 DOI: 10.1016/j.cytogfr.2021.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/05/2021] [Accepted: 10/13/2021] [Indexed: 01/08/2023]
Abstract
Coronavirus disease-2019 (COVID-19),
the disease caused by severe acute respiratory syndrome-coronavirus-2,
has claimed more than 4.4 million lives worldwide (as of 20 August 2021).
Severe cases of the disease often result in respiratory distress due to
cytokine storm, and mechanical ventilation is required. Although, the
lungs are the primary organs affected by the disease, more evidence on
damage to the heart, kidney, and liver is emerging. A common link in
these connections is the cardiovascular network. Inner lining of the
blood vessels, called endothelium, is formed by a single layer of
endothelial cells. Several clinical manifestations involving the
endothelium have been reported, such as its activation via
immunomodulation, endotheliitis, thrombosis, vasoconstriction, and
distinct intussusceptive angiogenesis (IA), a unique and rapid process of
blood-vessel formation by splitting a vessel into two lumens. In fact,
the virus directly infects the endothelium via TMPRSS2 spike glycoprotein
priming to facilitate ACE-2-mediated viral entry. Recent studies have
indicated a significant increase in remodeling of the pulmonary vascular
bed via intussusception in patients with COVID-19. However, the lack of
circulatory biomarkers for IA limits its detection in COVID-19
pathogenesis. In this review, we describe the implications of
angiogenesis in COVID-19, unique features of the pulmonary vascular bed
and its remodeling, and a rapid and non-invasive assessment of IA to
overcome the technical limitations in patients with
COVID-19.
Collapse
Affiliation(s)
- Pavitra K Jadaun
- Hepatology, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Suvro Chatterjee
- Department of Biotechnology, University of Burdwan, Golap Bag Campus, Burdwan, India.
| |
Collapse
|
34
|
Tabassum T, Rahman A, Araf Y, Ullah MA, Hosen MJ. Prospective selected biomarkers in COVID-19 diagnosis and treatment. Biomark Med 2021; 15:1435-1449. [PMID: 34538093 PMCID: PMC8454595 DOI: 10.2217/bmm-2021-0038] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 06/28/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 has become a global health concern, due to the high transmissible nature of its causal agent and lack of proper treatment. Early diagnosis and nonspecific medical supports of the patients appeared to be effective strategy so far to combat the pandemic caused by COVID-19 outbreak. Biomarkers can play pivotal roles in timely and proper diagnosis of COVID-19 patients, as well as for distinguishing them from other pulmonary infections. Besides, biomarkers can help in reducing the rate of mortality and evaluating viral pathogenesis with disease prognosis. This article intends to provide a broader overview of the roles and uses of different biomarkers in the early diagnosis of COVID-19, as well as in the classification of COVID-19 patients into multiple risk groups.
Collapse
Affiliation(s)
- Tahani Tabassum
- Department of Mathematics & Natural Sciences, Biotechnology Program, School of Data & Sciences, Brac University, Dhaka, Bangladesh
| | - Ahsab Rahman
- Department of Mathematics & Natural Sciences, Biotechnology Program, School of Data & Sciences, Brac University, Dhaka, Bangladesh
| | - Yusha Araf
- Department of Genetic Engineering & Biotechnology, School of Life Sciences, Shahjalal University of Science & Technology, Sylhet, Bangladesh
| | - Md A Ullah
- Department of Biotechnology & Genetic Engineering, Faculty of Biological Sciences, Jahangirnagar University, Dhaka, Bangladesh
| | - Mohammad J Hosen
- Department of Genetic Engineering & Biotechnology, School of Life Sciences, Shahjalal University of Science & Technology, Sylhet, Bangladesh
| |
Collapse
|
35
|
The misunderstood link between SARS-CoV-2 and angiogenesis. A narrative review. Pulmonology 2021:S2531-0437(21)00160-4. [PMID: 34593362 PMCID: PMC8390375 DOI: 10.1016/j.pulmoe.2021.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/30/2021] [Accepted: 08/02/2021] [Indexed: 02/06/2023] Open
Abstract
Novel Coronavirus Disease 2019 (Covid-19) is associated with multi-systemic derangement, including circulatory dysfunction with features of endothelial dysfunction, microangiopathic thrombosis and angiocentric inflammation. Recently, intussusceptive angiogenesis has been implicated in the pathogenesis of the disease. Herein, we conducted a narrative review according to the SANRA guidelines to review and discuss data regarding splitting angiogenesis including mechanisms, drivers, regulators and putative roles. Relevant angiogenic features in Covid-19, including their potential role in inflammation, endothelial dysfunction and permeability, as well as their use as prognostic markers and therapeutic roles are reviewed. Splitting angiogenesis in Covid-19 involve hypoxia, hypoxia-inducible factors, classic angiogenic mediators, such as the Vascular Endothelial Growth Factor (VEGF), Angiopoietins, hyperinflammation and cytokine storm, and dysregulation of the Renin-Angiotensin-Aldosterone System, which combined, interact to promote intussusception. Data regarding the use of angiogenic mediators as prognostic tools is summarized and suggest that angiopoietins and VEGF are elevated in Covid-19 patients and predictors of adverse outcomes. Finally, we reviewed the scarce data regarding angiogenic mediators as therapeutic targets. These preliminary findings suggest a potential benefit of bevacizumab as an add-on therapy.
Collapse
|
36
|
Drohan CM, Nouraie SM, Bain W, Shah FA, Evankovich J, Zhang Y, Morris A, McVerry BJ, Kitsios GD. Biomarker-Based Classification of Patients With Acute Respiratory Failure Into Inflammatory Subphenotypes: A Single-Center Exploratory Study. Crit Care Explor 2021; 3:e0518. [PMID: 34476405 PMCID: PMC8378789 DOI: 10.1097/cce.0000000000000518] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVES Hyper- and hypoinflammatory subphenotypes discovered in patients with acute respiratory distress syndrome predict clinical outcomes and therapeutic responses. These subphenotypes may be important in broader critically ill patient populations with acute respiratory failure regardless of clinical diagnosis. We investigated subphenotyping with latent class analysis in an inclusive population of acute respiratory failure, derived a parsimonious model for subphenotypic predictions based on a small set of variables, and examined associations with clinical outcomes. DESIGN Prospective, observational cohort study. SETTING Single-center, academic medical ICU. PATIENTS Mechanically ventilated patients with acute respiratory failure. MEASUREMENTS AND MAIN RESULTS We included 498 patients with acute respiratory failure (acute respiratory distress syndrome: 143, at-risk for acute respiratory distress syndrome: 198, congestive heart failure: 37, acute on chronic respiratory failure: 23, airway protection: 61, and multifactorial: 35) in our derivation cohort and measured 10 baseline plasma biomarkers. Latent class analysis considering clinical variables and biomarkers determined that a two-class model offered optimal fit (23% hyperinflammatory subphenotype). Distribution of hyperinflammatory subphenotype varied among acute respiratory failure etiologies (acute respiratory distress syndrome: 31%, at-risk for acute respiratory distress syndrome: 27%, congestive heart failure: 22%, acute on chronic respiratory failure 0%, airway protection: 5%, and multifactorial: 14%). Hyperinflammatory patients had higher Sequential Organ Failure Assessment scores, fewer ventilator-free days, and higher 30- and 90-day mortality (all p < 0.001). We derived a parsimonious model consisting of angiopoietin-2, soluble tumor necrosis factor receptor-1, procalcitonin, and bicarbonate and classified subphenotypes in a validation cohort (n = 139). Hyperinflammatory patients (19%) demonstrated higher levels of inflammatory biomarkers not included in the model (p < 0.01) and worse outcomes. CONCLUSIONS Host-response subphenotypes are observable in a heterogeneous population with acute respiratory failure and predict clinical outcomes. Simple, biomarker-based models can offer prognostic enrichment in patients with acute respiratory failure. The differential distribution of subphenotypes by specific etiologies of acute respiratory failure indicates that subphenotyping may be more relevant in patients with hypoxemic causes of acute respiratory failure and not in patients intubated for airway protection or acute on chronic decompensation.
Collapse
Affiliation(s)
- Callie M Drohan
- Division of General Internal Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - S Mehdi Nouraie
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - William Bain
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Staff Physician, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA
| | - Faraaz A Shah
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Staff Physician, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA
| | - John Evankovich
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Yingze Zhang
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Alison Morris
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Center for Medicine and the Microbiome, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Bryan J McVerry
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Center for Medicine and the Microbiome, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Georgios D Kitsios
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Center for Medicine and the Microbiome, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
37
|
Dembinski R. [Is it really an acute respiratory distress syndrome? : Current definitions, pathophysiology and differentiated diagnoses]. Anaesthesist 2021; 69:439-450. [PMID: 32430536 DOI: 10.1007/s00101-020-00789-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although the Berlin definition of the acute respiratory distress syndrome (ARDS) is generally recognized, the differentiation from other diseases with severe gas exchange disturbances is often difficult in clinical practice. In particular, the assessment of radiological findings and identification of primary noncardiogenic lung edema pose problems. In ARDS typical inflammatory processes can be found with involvement of activated neutrophilic granulocytes. Anti-inflammatory treatment strategies were unsuccessful. Lung protective ventilation strategies and prone positioning are the only evidence-based treatment options. Identifying ARDS phenotypes according to the etiology or disease progression can possibly provide a targeted individualized treatment option. The control of various biomarkers for assessment and treatment is the main focus of scientific interest. The results of appropriate studies remain to be seen.
Collapse
Affiliation(s)
- R Dembinski
- Klinik für Intensivmedizin und Notfallmedizin, Klinikum Bremen Mitte, St. Jürgen-Str. 1, 28177, Bremen, Deutschland.
| |
Collapse
|
38
|
Bihari S, Bersten A, Paul E, McGuinness S, Dixon D, Sinha P, Calfee CS, Nichol A, Hodgson C. Acute respiratory distress syndrome phenotypes with distinct clinical outcomes in PHARLAP trial cohort. CRIT CARE RESUSC 2021; 23:163-170. [PMID: 38045528 PMCID: PMC10692525 DOI: 10.51893/2021.2.oa3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: The Permissive Hypercapnia, Alveolar Recruitment and Low Airway Pressure (PHARLAP) randomised controlled trial compared an open lung ventilation strategy with control ventilation, and found that open lung ventilation did not reduce the number of ventilatorfree days (VFDs) or mortality in patients with moderate-to-severe acute respiratory distress syndrome (ARDS). Parsimonious models can identify distinct phenotypes of ARDS (hypo-inflammatory and hyperinflammatory) which are associated with different outcomes and treatment responses. Objective: To test the hypothesis that a parsimonious model would identify patients with distinctly different clinical outcomes in the PHARLAP study. Design, setting and participants: Blood and lung lavage samples were collected in a subset of PHARLAP patients who were recruited in Australian and New Zealand centres. A previously validated parsimonious model (interleukin-8, soluble tumour necrosis factor receptor-1 and bicarbonate) was used to classify patients with blood samples into hypo-inflammatory and hyperinflammatory groups. Generalised linear modelling was used to examine the interaction between inflammatory phenotype and treatment group (intervention or control). Main outcome measure: The primary outcome was number of VFDs at Day 28. Results: Data for the parsimonious model were available for 56 of 115 patients (49%). Within this subset, 38 patients (68%) and 18 patients (32%) were classified as having hypo-inflammatory and hyperinflammatory phenotypes, respectively. Patients with the hypo- inflammatory phenotype had more VFDs at Day 28 when compared with those with the hyperinflammatory phenotype (median [IQR], 19.5[11-24] versus 8[0-21];P= 0.03). Patients with the hyperinflammatory phenotype had numerically fewer VFDs when managed with an open lung strategy than when managed with control "protective" ventilation (median [IQR], 0 [0-19] versus 16 [8-22]). Conclusion: In the PHARLAP trial, ARDS patients classified as having a hyperinflammatory phenotype, with a parsimonious three-variable model, had fewer VFDs at Day 28 compared with patients classified as having a hypo-inflammatory phenotype. Future clinical studies of ventilatory strategies should consider incorporating distinct ARDS phenotypes into their trial design.
Collapse
Affiliation(s)
- Shailesh Bihari
- College of Medicine and Public Health- Flinders University-, Adelaide, - SA-, Australia
- Intensive and Critical Care Unit- Flinders Medical Centre-, Adelaide, - SA-, Australia
| | - Andrew Bersten
- College of Medicine and Public Health- Flinders University-, Adelaide, - SA-, Australia
- Intensive and Critical Care Unit- Flinders Medical Centre-, Adelaide, - SA-, Australia
| | - Eldho Paul
- Australian and New Zealand Intensive Care Research Centre, Monash University-, Melbourne, - VIC-, Australia
| | - Shay McGuinness
- Australian and New Zealand Intensive Care Research Centre, Monash University-, Melbourne, - VIC-, Australia
- Cardiothoracic and Vascular Intensive Care Unit-, Auckland, City Hospital- Auckland- New Zealand
- Medical Research Institute of New Zealand-, Wellington- New Zealand
| | - Dani Dixon
- College of Medicine and Public Health- Flinders University-, Adelaide, - SA-, Australia
- Intensive and Critical Care Unit- Flinders Medical Centre-, Adelaide, - SA-, Australia
| | - Pratik Sinha
- Division of Pulmonary- Critical Care- Allergy and Sleep Medicine Department of Medicine- University of California San Francisco-, San Francisco, - Calif-, USA
| | - Carolyn S. Calfee
- Division of Pulmonary- Critical Care- Allergy and Sleep Medicine Department of Medicine- University of California San Francisco-, San Francisco, - Calif-, USA
| | - Alistair Nichol
- Australian and New Zealand Intensive Care Research Centre, Monash University-, Melbourne, - VIC-, Australia
- Intensive Care Unit- The Alfred-, Melbourne, - VIC-, Australia
- University College Dublin Clinical Research Centre- St Vincent's University Hospital-, Dublin- Ireland
| | - Carol Hodgson
- Australian and New Zealand Intensive Care Research Centre, Monash University-, Melbourne, - VIC-, Australia
- Intensive Care Unit- The Alfred-, Melbourne, - VIC-, Australia
- Contributed equally to the manuscript
| | - for the PHARLAP Study Investigators
- College of Medicine and Public Health- Flinders University-, Adelaide, - SA-, Australia
- Intensive and Critical Care Unit- Flinders Medical Centre-, Adelaide, - SA-, Australia
- Australian and New Zealand Intensive Care Research Centre, Monash University-, Melbourne, - VIC-, Australia
- Cardiothoracic and Vascular Intensive Care Unit-, Auckland, City Hospital- Auckland- New Zealand
- Medical Research Institute of New Zealand-, Wellington- New Zealand
- Division of Pulmonary- Critical Care- Allergy and Sleep Medicine Department of Medicine- University of California San Francisco-, San Francisco, - Calif-, USA
- Intensive Care Unit- The Alfred-, Melbourne, - VIC-, Australia
- University College Dublin Clinical Research Centre- St Vincent's University Hospital-, Dublin- Ireland
- Contributed equally to the manuscript
| |
Collapse
|
39
|
Effect of Driving Pressure Change During Extracorporeal Membrane Oxygenation in Adults With Acute Respiratory Distress Syndrome: A Randomized Crossover Physiologic Study. Crit Care Med 2021; 48:1771-1778. [PMID: 33044283 DOI: 10.1097/ccm.0000000000004637] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Venovenous extracorporeal membrane oxygenation is an effective intervention to improve gas exchange in patients with severe acute respiratory distress syndrome. However, the mortality of patients with severe acute respiratory distress syndrome supported with venovenous extracorporeal membrane oxygenation remains high, and this may be due in part to a lack of standardized mechanical ventilation strategies aimed at further minimizing ventilator-induced lung injury. We tested whether a continuous positive airway pressure ventilation strategy mitigates ventilator-induced lung injury in patients with severe acute respiratory distress syndrome on venovenous extracorporeal membrane oxygenation, compared with current ventilation practice that employs tidal ventilation with limited driving pressure. We used plasma biomarkers as a surrogate outcome for ventilator-induced lung injury. DESIGN Randomized crossover physiologic study. SETTING Single-center ICU. PATIENTS Ten patients with severe acute respiratory distress syndrome supported on venovenous extracorporeal membrane oxygenation. INTERVENTIONS The study included four phases. After receiving pressure-controlled ventilation with driving pressure of 10 cm H2O for 1 hour (phase 1), patients were randomly assigned to receive first either pressure-controlled ventilation 20 cm H2O for 2 hours (phase 2) or continuous positive airway pressure for 2 hours (phase 3), and then crossover to the other phase for 2 hours; during phase 4 ventilation settings returned to baseline (pressure-controlled ventilation 10 cm H2O) for 4 hours. MEASUREMENTS AND MAIN RESULTS There was a linear relationship between the change in driving pressure and the plasma concentration of interleukin-6, soluble receptor for advanced glycation end products, interleukin-1ra, tumor necrosis factor alpha, surfactant protein D, and interleukin-10. CONCLUSIONS Ventilator-induced lung injury may occur in acute respiratory distress syndrome patients on venovenous extracorporeal membrane oxygenation despite the delivery of volume- and pressure-limited mechanical ventilation. Reducing driving pressure to zero may provide more protective mechanical ventilation in acute respiratory distress syndrome patients supported with venovenous extracorporeal membrane oxygenation. However, the risks versus benefits of such an approach need to be confirmed in studies that are designed to test patient centered outcomes.
Collapse
|
40
|
Liu Y, Liu J, Huang L. A Simple-to-Use Web-Based Calculator for Survival Prediction in Acute Respiratory Distress Syndrome. Front Med (Lausanne) 2021; 8:604694. [PMID: 33665197 PMCID: PMC7921740 DOI: 10.3389/fmed.2021.604694] [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: 09/10/2020] [Accepted: 01/29/2021] [Indexed: 11/15/2022] Open
Abstract
Background: The aim of this study was to construct and validate a simple-to-use model to predict the survival of patients with acute respiratory distress syndrome. Methods: A total of 197 patients with acute respiratory distress syndrome were selected from the Dryad Digital Repository. All eligible individuals were randomly stratified into the training set (n=133) and the validation set (n=64) as 2: 1 ratio. LASSO regression analysis was used to select the optimal predictors, and receiver operating characteristic and calibration curves were used to evaluate accuracy and discrimination of the model. Clinical usefulness of the model was also assessed using decision curve analysis and Kaplan-Meier analysis. Results: Age, albumin, platelet count, PaO2/FiO2, lactate dehydrogenase, high-resolution computed tomography score, and etiology were identified as independent prognostic factors based on LASSO regression analysis; these factors were integrated for the construction of the nomogram. Results of calibration plots, decision curve analysis, and receiver operating characteristic analysis showed that this model has good predictive ability of patient survival in acute respiratory distress syndrome. Moreover, a significant difference in the 28-day survival was shown between the patients stratified into different risk groups (P < 0.001). For convenient application, we also established a web-based calculator (https://huangl.shinyapps.io/ARDSprognosis/). Conclusions: We satisfactorily constructed a simple-to-use model based on seven relevant factors to predict survival and prognosis of patients with acute respiratory distress syndrome. This model can aid personalized treatment and clinical decision-making.
Collapse
Affiliation(s)
- Yong Liu
- Department of Emergency, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jian Liu
- Department of Emergency, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Liang Huang
- Department of Emergency, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW This article provides an overview of protein biomarkers for acute respiratory distress syndrome (ARDS) and their potential use in future clinical trials. RECENT FINDINGS The protein biomarkers studied as indices of biological processes involved in the pathogenesis of ARDS may have diagnostic and/or prognostic value. Recently, they also proved useful for identifying ARDS phenotypes and assessing heterogeneity of treatment effect in retrospective analyses of completed clinical trials. SUMMARY This article summarizes the current research on ARDS biomarkers and provides insights into how they should be integrated as prognostic and predictive enrichment tools in future clinical trials.
Collapse
Affiliation(s)
- Matthieu Jabaudon
- Department of Perioperative Medicine, CHU Clermont-Ferrand
- GReD, CNRS, INSERM, Université Clermont Auvergne, Clermont-Ferrand, France
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine
| | - Raiko Blondonnet
- Department of Perioperative Medicine, CHU Clermont-Ferrand
- GReD, CNRS, INSERM, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Lorraine B Ware
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
42
|
Understanding Restrictive Versus Liberal Fluid Therapy for Major Abdominal Surgery Trial Results: Did Liberal Fluids Associate With Increased Endothelial Injury Markers? Crit Care Explor 2021; 3:e0316. [PMID: 33521643 PMCID: PMC7838007 DOI: 10.1097/cce.0000000000000316] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Supplemental Digital Content is available in the text. Liberal fluid strategies in critically ill patients are associated with harm, thought to be due to endothelial and glycocalyx injury. As the restrictive versus liberal fluid therapy for major abdominal surgery trial not only failed to report survival benefit with restrictive fluids but was associated with a higher rate of acute kidney injury, we hypothesized that factors other than endothelial and glycocalyx injury were likely to account for these findings. Consequently, we measured injury biomarkers in a cohort of the restrictive versus liberal fluid therapy for major abdominal surgery trial.
Collapse
|
43
|
Zhou B, Kojima S, Kawamoto A, Fukushima M. COVID-19 pathogenesis, prognostic factors, and treatment strategy: Urgent recommendations. J Med Virol 2021; 93:2694-2704. [PMID: 33368358 DOI: 10.1002/jmv.26754] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/16/2020] [Accepted: 12/21/2020] [Indexed: 01/08/2023]
Abstract
The pandemic of novel coronavirus disease (COVID-19) is not yet close to being over, more than 8 months after the first cases, but researchers are making great progress in fighting the disease. We have conducted a brief review of the geographic differences in the prevalence of COVID-19, the updated pathological findings, prognostic factors, and treatments for disease prevention and improvement of prognosis. Although hydroxychloroquine and tocilizumab have been recommended by some researchers, many clinical trials have failed to confirm any beneficial effect of these and other drugs on COVID-19, in terms of improved clinical status or reduced patient mortality. Currently, glucocorticoid is the only drug that reduces the mortality of COVID-19 in a randomized controlled trial; however, it is still necessary to establish the optimal timing of administration. It is also urgent to set up an international or national cohort to address the risk factors associated with infection, the natural history of COVID-19, including the disease type, surrogate markers for critically ill, long-term sequelae, and reinfection after exposure, identify responders to glucocorticoid, and establish optimal treatment strategies for disease control.
Collapse
Affiliation(s)
- Bin Zhou
- Department of Medscience, Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation, Kobe, Japan
| | - Shinsuke Kojima
- Department of Medscience, Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation, Kobe, Japan
| | - Atsuhiko Kawamoto
- Department of Medscience, Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation, Kobe, Japan
| | - Masanori Fukushima
- Department of Medscience, Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation, Kobe, Japan
| |
Collapse
|
44
|
Bülow Anderberg S, Luther T, Berglund M, Larsson R, Rubertsson S, Lipcsey M, Larsson A, Frithiof R, Hultström M. Increased levels of plasma cytokines and correlations to organ failure and 30-day mortality in critically ill Covid-19 patients. Cytokine 2020; 138:155389. [PMID: 33348065 PMCID: PMC7833204 DOI: 10.1016/j.cyto.2020.155389] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/22/2020] [Accepted: 11/23/2020] [Indexed: 02/09/2023]
Abstract
Fifteen cytokines were increased at ICU admission in Covid-19 patients. A relatively moderate increase in cytokine concentrations was observed. IL-1ra, IL-6 and IP-10 correlated with respiratory failure and acute kidney injury. IL-8 may be a future biomarker due to its correlation with 30-day mortality.
Background The infection caused by SARS CoV-2 has been postulated to induce a cytokine storm syndrome that results in organ failure and even death in a considerable number of patients. However, the inflammatory response in Corona virus disease-19 (Covid-19) and its potential to cause collateral organ damage has not been fully elucidated to date. This study aims to characterize the acute cytokine response in a cohort of critically ill Covid-19 patients. Method 24 adults with PCR-confirmed Covid-19 were included at time of admission to intensive care a median of eleven days after initial symptoms. Eleven adult patients admitted for elective abdominal surgery with preoperative plasma samples served as controls. All patients were included after informed consent was obtained. 27 cytokines were quantified in plasma. The expression of inflammatory mediators was then related to routine inflammatory markers, SAPS3, SOFA score, organ failure and 30-day mortality. Results A general increase in cytokine expression was observed in all Covid-19 patients. A strong correlation between respiratory failure and IL-1ra, IL-4, IL-6, IL-8 and IP-10 expression was observed. Acute kidney injury development correlated well with increased levels of IL-1ra, IL-6, IL-8, IL-17a, IP-10 and MCP-1. Generally, the cohort demonstrated weaker correlations between cytokine expression and 30-day mortality out of which IL-8 showed the strongest signal in terms of mortality. Conclusion The present study found that respiratory failure, acute kidney injury and 30-day mortality in critically ill Covid-19 patients are associated with moderate increases of a broad range of inflammatory mediators at time of admission.
Collapse
Affiliation(s)
- Sara Bülow Anderberg
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden.
| | - Tomas Luther
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Malin Berglund
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Rolf Larsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Sten Rubertsson
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Miklos Lipcsey
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden; Hedenstierna Laboratory, Anesthesiology and Intensive Care, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Anders Larsson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Robert Frithiof
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Michael Hultström
- Department of Surgical Sciences, Anesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden; Department of Medical Cell Biology, Integrative Physiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
45
|
Bime C, Camp SM, Casanova N, Oita RC, Ndukum J, Lynn H, Garcia JGN. The acute respiratory distress syndrome biomarker pipeline: crippling gaps between discovery and clinical utility. Transl Res 2020; 226:105-115. [PMID: 32599095 PMCID: PMC7319618 DOI: 10.1016/j.trsl.2020.06.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/17/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022]
Abstract
Recent innovations in translational research have ushered an exponential increase in the discovery of novel biomarkers, thereby elevating the hope for deeper insights into "personalized" medicine approaches to disease phenotyping and care. However, a critical gap exists between the fast pace of biomarker discovery and the successful translation to clinical use. This gap underscores the fundamental biomarker conundrum across various acute and chronic disorders: how does a biomarker address a specific unmet need? Additionally, the gap highlights the need to shift the paradigm from a focus on biomarker discovery to greater translational impact and the need for a more streamlined drug approval process. The unmet need for biomarkers in acute respiratory distress syndrome (ARDS) is for reliable and validated biomarkers that minimize heterogeneity and allow for stratification of subject selection for enrollment in clinical trials of tailored therapies. This unmet need is particularly highlighted by the ongoing SARS-CoV-2/COVID-19 pandemic. The unprecedented numbers of COVID-19-induced ARDS cases has strained health care systems across the world and exposed the need for biomarkers that would accelerate drug development and the successful phenotyping of COVID-19-infected patients at risk for development of ARDS and ARDS mortality. Accordingly, this review discusses the current state of ARDS biomarkers in the context of the drug development pipeline and highlight gaps between biomarker discovery and clinical implementation while proposing potential paths forward. We discuss potential ARDS biomarkers by category and by context of use, highlighting progress in the development continuum. We conclude by discussing challenges to successful translation of biomarker candidates to clinical impact and proposing possible novel strategies.
Collapse
Affiliation(s)
- Christian Bime
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona.
| | - Sara M Camp
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Nancy Casanova
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Radu C Oita
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Juliet Ndukum
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Heather Lynn
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Joe G N Garcia
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| |
Collapse
|
46
|
Skopas V, Papadopoulos D, Trakas N, Papaefstathiou E, Koufopoulos C, Makris D, Daniil Z, Gourgoulianis K. Lactate dehydrogenase isoenzymes in patients with acute exacerbation of chronic obstructive pulmonary disease: An exploratory cross-sectional study. Respir Physiol Neurobiol 2020; 283:103562. [PMID: 33038523 DOI: 10.1016/j.resp.2020.103562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 09/18/2020] [Accepted: 10/02/2020] [Indexed: 10/23/2022]
Abstract
We aimed to evaluate differences in serum lactate dehydrogenase (LDH) isoenzymes between patients hospitalized for acute exacerbation of chronic obstructive pulmonary disease (AECOPD) and other lower respiratory tract infections (LRTIs). Based on self-reported COPD diagnosis, 71 participants were divided into AECOPD (n = 38, 29 males, mean age 70.5 years) and LRTI (n = 33, 12 males, mean age 70.4 years) groups. Information on demographics, comorbidities, and COPD severity markers, as well as arterial blood gases and laboratory data were collected, while serum LDH electrophoresis was performed to examine the LDH isoenzymes. Adjusting for sex, age, comorbidities, degree of hypoxemia, inflammation markers, muscle and myocardial enzymes, and total serum LDH, the mean differences (95 % confidence intervals) in the ratios of serum LDH isoenzymes to total serum LDH between groups (LDHxAECOPD - LDHxLRTI) were statistically significant for LDH1 [4.9 (1.4 to 8.3)], LDH2 [3.0 (0.1 to 5.8)], LDH3 [-4.3 (-6.3 to -2.3)], and LDH4 [-3.2 (-4.9 to -1.5)]. A sum of LDH3 and LDH4 ratios below 29 % had the highest discriminative ability to classify a subject in the AECOPD group (AUC 0.841, sensitivity 76 %, specificity 87 %). Aerobic metabolic adaptive mechanisms in respiratory muscles during AECOPD could explain the above differences.
Collapse
Affiliation(s)
- Vlasios Skopas
- 2nd Pulmonary Department, "Sismanogleion" General Hospital of Attica, Marousi, Greece; Respiratory Medicine Department, School of Medicine, University of Thessaly, University Hospital of Larissa, Larissa, Greece.
| | | | - Nikolaos Trakas
- Clinical Chemistry Department, "Sismanogleion" General Hospital of Attica, Marousi, Greece
| | - Eleni Papaefstathiou
- Clinical Chemistry Department, "Sismanogleion" General Hospital of Attica, Marousi, Greece
| | | | - Demosthenes Makris
- Critical Care Department, School of Medicine, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - Zoe Daniil
- Respiratory Medicine Department, School of Medicine, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - Konstantinos Gourgoulianis
- Respiratory Medicine Department, School of Medicine, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| |
Collapse
|
47
|
Saha A, Anirvan P. Cancer progression in COVID-19: integrating the roles of renin angiotensin aldosterone system, angiopoietin-2, heat shock protein-27 and epithelial mesenchymal transition. Ecancermedicalscience 2020; 14:1099. [PMID: 33082849 PMCID: PMC7532023 DOI: 10.3332/ecancer.2020.1099] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Indexed: 12/12/2022] Open
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic has affected millions worldwide and has been found to cause severe disease in patients with underlying comorbidities. In patients with known malignancies, in addition to constraints in routine healthcare, the risk of being susceptible to developing severe forms of the disease is of grave concern. While follow-up studies on survivors of the severe acute respiratory syndrome (SARS) 2003 outbreak revealed increased susceptibility to infections, tumours and cardiovascular abnormalities, recent studies implicating angiopoietin 2 in induction of inflammatory intussusceptive angiogenesis and diffuse alveolar damage in COVID-19 patients raises the possibility of progression of carcinogenetic processes in patients with known malignancies. Angiotensin converting enzyme-2 (ACE-2) mediated cellular entry of SARS-Cov2 leads to receptor shedding of ACE-2 and disrupts the renin angiotensin aldosterone axis (RAAS). This augments the pro-inflammatory and proliferative effects of RAAS, while attenuating the anti-inflammatory and anti-proliferative angiotensin 1-7 /Mas pathway. Angiopoietin-2, a molecule responsible for angiogenesis and cancer progression which corelates with tumour load in certain cancers, is upregulated by angiotensin 2-AT1 Receptor axis. Tumour microenvironment-comprising of various cells, blood vessels and extra cellular matrix which express the RAAS peptides-plays a key role in cancer initiation, progression and metastasis. Angiotensin 2 induces the formation of a desmoplastic environment, favouring cancer cell growth. ACE-2 downregulation causes bradykinin accumulation which may exert its proliferative action via mitogen activated protein kinase pathways which has established roles in cancers of breast and kidney. In addition to cytokine storm causing organ damage, acute inflammation in COVID-19 may also cause epithelial mesenchymal transition and heat shock protein 27 phosphorylation, both of which are key mediators in cancer signalling pathways. We hypothesise that SARS-Cov2, by impacting the RAAS and immune system, has the potential to cause tumour cell proliferation, apoptosis evasion and metastasis, thereby increasing the possibility of cancer progression in patients with known malignancies.
Collapse
Affiliation(s)
- Aritra Saha
- Department of Internal Medicine, Assam Medical College and Hospital, Dibrugarh 786002, Assam, India
- https://orcid.org/0000-0003-4705-7453
| | - Prajna Anirvan
- Department of Gastroenterology, Sriram Chandra Bhanj Medical College and Hospital, Cuttack 753007, Odisha, India
- https://orcid.org/0000-0003-4494-0865
| |
Collapse
|
48
|
Patregnani JT, Brooks BA, Chorvinsky E, Pillai DK. High BAL sRAGE is Associated with Low Serum Eosinophils and IgE in Children with Asthma. CHILDREN (BASEL, SWITZERLAND) 2020; 7:E110. [PMID: 32846877 PMCID: PMC7552609 DOI: 10.3390/children7090110] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022]
Abstract
Asthma remains the most common chronic lung disease in childhood in the United States. The receptor for advanced glycation end products (RAGE) has been recognized as both a marker of and participant in pulmonary pathophysiology. While membrane-bound RAGE (mRAGE) perpetuates the type 2 immune response, the soluble form (sRAGE) may act as a decoy receptor for pro-inflammatory ligands. Bronchoalveolar samples from 45 pediatric patients with asthma were obtained. Patients were divided into high and low BAL sRAGE groups using median sRAGE. Descriptive statistical analysis and non-parametric testing were applied. Children in the "high" sRAGE group had a lower median serum eosinophil (0.27 [SE ± 0.04] vs. 0.57 [± 0.06] K/mcl, adjusted p = 0.003) and lower serum IgE level (194.4 [± 60.7] vs. 676.2 ± 140.5) IU/mL, adjusted p = 0.004) as compared to the "low" sRAGE group. When controlling for age and body mass index percentile, absolute eosinophil count (p = 0.03) and serum IgE (p = 0.043) remained significantly lower in the "high" sRAGE group. Children with asthma and high levels of BAL sRAGE have lower serum eosinophil and IgE levels. These findings are consistent with the hypothesis that sRAGE may act as a decoy receptor by binding ligands that normally interact with mRAGE.
Collapse
Affiliation(s)
- Jason T. Patregnani
- Division of Cardiac Critical Care Medicine, Children’s National Hospital, Washington, DC 20010, USA
- Department of Genomics and Precision Medicine, The George Washington University, Washington, DC 20052, USA; (E.C.); (D.K.P.)
| | - Bonnie A. Brooks
- Division of Critical Care Medicine, Children’s National Hospital, Washington, DC 20010, USA;
| | - Elizabeth Chorvinsky
- Department of Genomics and Precision Medicine, The George Washington University, Washington, DC 20052, USA; (E.C.); (D.K.P.)
| | - Dinesh K. Pillai
- Department of Genomics and Precision Medicine, The George Washington University, Washington, DC 20052, USA; (E.C.); (D.K.P.)
- Division of Pulmonology, Children’s National Hospital, Washington, DC 20010, USA
| |
Collapse
|
49
|
IL-6 trans-signaling induces plasminogen activator inhibitor-1 from vascular endothelial cells in cytokine release syndrome. Proc Natl Acad Sci U S A 2020; 117:22351-22356. [PMID: 32826331 PMCID: PMC7486751 DOI: 10.1073/pnas.2010229117] [Citation(s) in RCA: 202] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Cytokine release syndrome (CRS) is a life-threatening complication induced by systemic inflammatory responses to infections, including bacteria and chimeric antigen receptor T cell therapy. There are currently no immunotherapies with proven clinical efficacy and understanding of the molecular mechanisms of CRS pathogenesis is limited. Here, we found that patients diagnosed with CRS from sepsis, acute respiratory distress syndrome (ARDS), or burns showed common manifestations: strikingly elevated levels of the four proinflammatory cytokines interleukin (IL)-6, IL-8, monocyte chemotactic protein-1 (MCP-1), and IL-10 and the coagulation cascade activator plasminogen activator inhibitor-1 (PAI-1). Our in vitro data indicate that endothelial IL-6 trans-signaling formed an inflammation circuit for robust IL-6, IL-8, and MCP-1 production and promoted PAI-1 production; additionally, an IL-6 signaling blockade by the human monoclonal antibody tocilizumab blunted endothelial cell activation. Plasma from severe COVID-19 patients similarly exhibited increased IL-6, IL-10, and MCP-1 levels, but these levels were not as high as those in patients with CRS from other causes. In contrast, the PAI-1 levels in COVID-19 patients were as highly elevated as those in patients with bacterial sepsis or ARDS. Tocilizumab treatment decreased the PAI-1 levels and alleviated critical illness in severe COVID-19 patients. Our findings suggest that distinct levels of cytokine production are associated with CRS induced by bacterial infection and COVID-19, but both CRS types are accompanied by endotheliopathy through IL-6 trans-signaling. Thus, the present study highlights the crucial role of IL-6 signaling in endothelial dysfunction during bacterial infection and COVID-19.
Collapse
|
50
|
|