1
|
Scaravilli V, Turconi G, Colombo SM, Guzzardella A, Bosone M, Zanella A, Bos L, Grasselli G. Early serum biomarkers to characterise different phenotypes of primary graft dysfunction after lung transplantation: a systematic scoping review. ERJ Open Res 2024; 10:00121-2024. [PMID: 39104958 PMCID: PMC11298996 DOI: 10.1183/23120541.00121-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 03/12/2024] [Indexed: 08/07/2024] Open
Abstract
Background Lung transplantation (LUTX) is often complicated by primary graft dysfunction (PGD). Plasma biomarkers hold potential for PGD phenotyping and targeted therapy. This scoping review aims to collect the available literature in search of serum biomarkers for PGD phenotyping. Methods Following JBI and PRISMA guidelines, we conducted a systematic review searching MEDLINE, Web of Science, EMBASE and The Cochrane Library for papers reporting the association between serum biomarkers measured within 72 h of reperfusion and PGD, following International Society for Heart and Lung Transplantation (ISHLT) guidelines. We extracted study details, patient demographics, PGD definition and timing, biomarker concentration, and their performance in identifying PGD cases. Results Among the 1050 papers screened, 25 prospective observational studies were included, with only nine conducted in the last decade. These papers included 1793 unique adult patients (1195 double LUTX, median study size 100 (IQR 44-119)). Most (n=21) compared PGD grade 3 to less severe PGD, but only four adhered to 2016 PGD definitions. Enzyme-linked immunosorbent assays and the multiplex bead array technique were utilised in 23 and two papers, respectively. In total, 26 candidate biomarkers were identified, comprising 13 inflammatory, three endothelial activation, three epithelial injury, three cellular damage and two coagulation dysregulation markers. Only five biomarkers (sRAGE, ICAM-1, PAI-1, SP-D, FSTL-1) underwent area under the receiver operating characteristic curve analysis, yielding a median value of 0.58 (0.51-0.78) in 406 patients (276 double LUTX). Conclusions Several biomarkers exhibit promise for future studies aimed at PGD phenotyping after LUTX. To uncover the significant existing knowledge gaps, further international prospective studies incorporating updated diagnostic criteria, modern platforms and advanced statistical approaches are essential.
Collapse
Affiliation(s)
- Vittorio Scaravilli
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca’ Granda – Ospedale Maggiore Policlinico, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Gloria Turconi
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Sebastiano Maria Colombo
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca’ Granda – Ospedale Maggiore Policlinico, Milan, Italy
| | - Amedeo Guzzardella
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Marco Bosone
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Alberto Zanella
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca’ Granda – Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Lieuwe Bos
- Department of Intensive Care, University of Amsterdam, Amsterdam, Netherlands
| | - Giacomo Grasselli
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca’ Granda – Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
2
|
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
|
3
|
Delrue C, Speeckaert R, Delanghe JR, Speeckaert MM. Breath of fresh air: Investigating the link between AGEs, sRAGE, and lung diseases. VITAMINS AND HORMONES 2024; 125:311-365. [PMID: 38997169 DOI: 10.1016/bs.vh.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Advanced glycation end products (AGEs) are compounds formed via non-enzymatic reactions between reducing sugars and amino acids or proteins. AGEs can accumulate in various tissues and organs and have been implicated in the development and progression of various diseases, including lung diseases. The receptor of advanced glycation end products (RAGE) is a receptor that can bind to advanced AGEs and induce several cellular processes such as inflammation and oxidative stress. Several studies have shown that both AGEs and RAGE play a role in the pathogenesis of lung diseases, such as chronic obstructive pulmonary disease, asthma, idiopathic pulmonary fibrosis, cystic fibrosis, and acute lung injury. Moreover, the soluble form of the receptor for advanced glycation end products (sRAGE) has demonstrated its ability to function as a decoy receptor, possessing beneficial characteristics such as anti-inflammatory, antioxidant, and anti-fibrotic properties. These qualities make it an encouraging focus for therapeutic intervention in managing pulmonary disorders. This review highlights the current understanding of the roles of AGEs and (s)RAGE in pulmonary diseases and their potential as biomarkers and therapeutic targets for preventing and treating these pathologies.
Collapse
Affiliation(s)
- Charlotte Delrue
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium
| | | | - Joris R Delanghe
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Marijn M Speeckaert
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium; Research Foundation-Flanders (FWO), Brussels, Belgium.
| |
Collapse
|
4
|
de Souza Xavier Costa N, da Costa Sigrist G, Schalch AS, Belotti L, Dolhnikoff M, da Silva LFF. Lung tissue expression of epithelial injury markers is associated with acute lung injury severity but does not discriminate sepsis from ARDS. Respir Res 2024; 25:129. [PMID: 38500106 PMCID: PMC10949726 DOI: 10.1186/s12931-024-02761-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 03/08/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a common cause of respiratory failure in critically ill patients, and diffuse alveolar damage (DAD) is considered its histological hallmark. Sepsis is one of the most common aetiology of ARDS with the highest case-fatality rate. Identifying ARDS patients and differentiate them from other causes of acute respiratory failure remains a challenge. To address this, many studies have focused on identifying biomarkers that can help assess lung epithelial injury. However, there is scarce information available regarding the tissue expression of these markers. Evaluating the expression of elafin, RAGE, and SP-D in lung tissue offers a potential bridge between serological markers and the underlying histopathological changes. Therefore, we hypothesize that the expression of epithelial injury markers varies between sepsis and ARDS as well as according to its severity. METHODS We compared the post-mortem lung tissue expression of the epithelial injury markers RAGE, SP-D, and elafin of patients that died of sepsis, ARDS, and controls that died from non-pulmonary causes. Lung tissue was collected during routine autopsy and protein expression was assessed by immunohistochemistry. We also assessed the lung injury by a semi-quantitative analysis. RESULTS We observed that all features of DAD were milder in septic group compared to ARDS group. Elafin tissue expression was increased and SP-D was decreased in the sepsis and ARDS groups. Severe ARDS expressed higher levels of elafin and RAGE, and they were negatively correlated with PaO2/FiO2 ratio, and positively correlated with bronchopneumonia percentage and hyaline membrane score. RAGE tissue expression was negatively correlated with mechanical ventilation duration in both ARDS and septic groups. In septic patients, elafin was positively correlated with ICU admission length, SP-D was positively correlated with serum lactate and RAGE was correlated with C-reactive protein. CONCLUSIONS Lung tissue expression of elafin and RAGE, but not SP-D, is associated with ARDS severity, but does not discriminate sepsis patients from ARDS patients.
Collapse
Affiliation(s)
| | - Giovana da Costa Sigrist
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, LIM-05, Brazil
| | - Alexandre Santos Schalch
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, LIM-05, Brazil
| | - Luciano Belotti
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, LIM-05, Brazil
| | - Marisa Dolhnikoff
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, LIM-05, Brazil
| | - Luiz Fernando Ferraz da Silva
- Departamento de Patologia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, LIM-05, Brazil
- Serviço de Verificação de Óbitos da Capital, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
5
|
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
|
6
|
Levine AR, Calfee CS. Subphenotypes of Acute Respiratory Distress Syndrome: Advancing Towards Precision Medicine. Tuberc Respir Dis (Seoul) 2024; 87:1-11. [PMID: 37675452 PMCID: PMC10758309 DOI: 10.4046/trd.2023.0104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/25/2023] [Accepted: 09/06/2023] [Indexed: 09/08/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common cause of severe hypoxemia defined by the acute onset of bilateral non-cardiogenic pulmonary edema. The diagnosis is made by defined consensus criteria. Supportive care, including prevention of further injury to the lungs, is the only treatment that conclusively improves outcomes. The inability to find more advanced therapies is due, in part, to the highly sensitive but relatively non-specific current syndromic consensus criteria, combining a heterogenous population of patients under the umbrella of ARDS. With few effective therapies, the morality rate remains 30% to 40%. Many subphenotypes of ARDS have been proposed to cluster patients with shared combinations of observable or measurable traits. Subphenotyping patients is a strategy to overcome heterogeneity to advance clinical research and eventually identify treatable traits. Subphenotypes of ARDS have been proposed based on radiographic patterns, protein biomarkers, transcriptomics, and/or machine-based clustering of clinical and biological variables. Some of these strategies have been reproducible across patient cohorts, but at present all have practical limitations to their implementation. Furthermore, there is no agreement on which strategy is the most appropriate. This review will discuss the current strategies for subphenotyping patients with ARDS, including the strengths and limitations, and the future directions of ARDS subphenotyping.
Collapse
Affiliation(s)
- Andrea R. Levine
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Carolyn S. Calfee
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
7
|
Blot PL, Chousterman BG, Santafè M, Cartailler J, Pacheco A, Magret M, Masclans JR, Artigas A, Roca O, García-de-Acilu M. Subphenotypes in patients with acute respiratory distress syndrome treated with high-flow oxygen. Crit Care 2023; 27:419. [PMID: 37915062 PMCID: PMC10619276 DOI: 10.1186/s13054-023-04687-0] [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: 08/23/2023] [Accepted: 10/14/2023] [Indexed: 11/03/2023] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) subphenotypes differ in outcomes and treatment responses. Subphenotypes in high-flow nasal oxygen (HFNO)-treated ARDS patients have not been investigated. OBJECTIVES To identify biological subphenotypes in HFNO-treated ARDS patients. METHODS Secondary analysis of a prospective multicenter observational study including ARDS patients supported with HFNO. Plasma inflammation markers (interleukin [IL]-6, IL-8, and IL-33 and soluble suppression of tumorigenicity-2 [sST2]) and lung epithelial (receptor for advanced glycation end products [RAGE] and surfactant protein D [SP-D]) and endothelial (angiopoietin-2 [Ang-2]) injury were measured. These biomarkers and bicarbonate were used in K-means cluster analysis to identify subphenotypes. Logistic regression was performed on biomarker combinations to predict clustering. We chose the model with the best AUROC and the lowest number of variables. This model was used to describe the HAIS (High-flow ARDS Inflammatory Subphenotype) score. RESULTS Among 41 HFNO patients, two subphenotypes were identified. Hyperinflammatory subphenotype (n = 17) showed higher biomarker levels than hypoinflammatory (n = 24). Despite similar baseline characteristics, the hyperinflammatory subphenotype had higher 60-day mortality (47 vs 8.3% p = 0.014) and longer ICU length of stay (22.0 days [18.0-30.0] vs 39.5 [25.5-60.0], p = 0.034). The HAIS score, based on IL-8 and sST2, accurately distinguished subphenotypes (AUROC 0.96 [95%CI: 0.90-1.00]). A HAIS score ≥ 7.45 was predictor of hyperinflammatory subphenotype. CONCLUSION ARDS patients treated with HFNO exhibit two biological subphenotypes that have similar clinical characteristics, but hyperinflammatory patients have worse outcomes. The HAIS score may identify patients with hyperinflammatory subphenotype and might be used for enrichment strategies in future clinical trials.
Collapse
Affiliation(s)
- Pierre-Louis Blot
- Département d'anesthésie-Réanimation, Hôpital Lariboisière, Paris, France
- INSERM UMRS-942 MASCOT, Hôpital Lariboisière, Paris, France
| | - Benjamin G Chousterman
- Département d'anesthésie-Réanimation, Hôpital Lariboisière, Paris, France
- INSERM UMRS-942 MASCOT, Hôpital Lariboisière, Paris, France
| | - Manel Santafè
- Servei de Medicina Intensiva, Parc Taulí Hospital Universitari, Institut de Recerca Part Taulí (I3PT-CERCA), Parc del Taulí 1, 08028, Sabadell, Spain
| | | | - Andrés Pacheco
- Servei de Medicina Intensiva, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Mònica Magret
- Servei de Medicina Intensiva, Hospital Universitari Joan XXIII, Tarragona, Spain
| | - Joan R Masclans
- Critical Care Department, Hospital del Mar-Parc de Salut MAR. GREPAC-Group Recerca Departamento de Medicina y Ciencias de la Vida Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Director de Docencia PSMAR, Intensive Care Unit Hospital del Mar. Professor of Medicine Universitat Pompeu Fabra (UPF) IMIM (GREPAC - Group Recerca Patologia Critica) Departamento de Medicina Y Ciencias de la Vida (MELIS), Universitat Pompeu Fabra, Barcelona, Spain
| | - Antoni Artigas
- Servei de Medicina Intensiva, Parc Taulí Hospital Universitari, Institut de Recerca Part Taulí (I3PT-CERCA), Parc del Taulí 1, 08028, Sabadell, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
- CIBER de Enfermedades Respiratorias, Insituto de Salud Carlos III, Madrid, Spain
| | - Oriol Roca
- Servei de Medicina Intensiva, Parc Taulí Hospital Universitari, Institut de Recerca Part Taulí (I3PT-CERCA), Parc del Taulí 1, 08028, Sabadell, Spain.
- Departament de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| | - Marina García-de-Acilu
- Servei de Medicina Intensiva, Parc Taulí Hospital Universitari, Institut de Recerca Part Taulí (I3PT-CERCA), Parc del Taulí 1, 08028, Sabadell, Spain
| |
Collapse
|
8
|
Ishaque S, Famularo ST, Saleem AF, Siddiqui NUR, Kazi Z, Parkar S, Hotwani A, Thomas NJ, Thompson JM, Lahni P, Varisco B, Yehya N. Biomarker-Based Risk Stratification in Pediatric Sepsis From a Low-Middle Income Country. Pediatr Crit Care Med 2023; 24:563-573. [PMID: 37092821 PMCID: PMC10317305 DOI: 10.1097/pcc.0000000000003244] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
OBJECTIVES Most biomarker studies of sepsis originate from high-income countries, whereas mortality risk is higher in low- and middle-income countries. The second version of the Pediatric Sepsis Biomarker Risk Model (PERSEVERE-II) has been validated in multiple North American PICUs for prognosis. Given differences in epidemiology, we assessed the performance of PERSEVERE-II in septic children from Pakistan, a low-middle income country. Due to uncertainty regarding how well PERSEVERE-II would perform, we also assessed the utility of other select biomarkers reflecting endotheliopathy, coagulopathy, and lung injury. DESIGN Prospective cohort study. SETTING PICU in Aga Khan University Hospital in Karachi, Pakistan. PATIENTS Children (< 18 yr old) meeting pediatric modifications of adult Sepsis-3 criteria between November 2020 and February 2022 were eligible. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Plasma was collected within 24 hours of admission and biomarkers quantified. The area under the receiver operating characteristic curve for PERSEVERE-II to discriminate 28-day mortality was determined. Additional biomarkers were compared between survivors and nonsurvivors and between subjects with and without acute respiratory distress syndrome. In 86 subjects (20 nonsurvivors, 23%), PERSEVERE-II discriminated mortality (area under the receiver operating characteristic curve, 0.83; 95% CI, 0.72-0.94) and stratified the cohort into low-, medium-, and high-risk of mortality. Biomarkers reflecting endotheliopathy (angiopoietin 2, intracellular adhesion molecule 1) increased across worsening risk strata. Angiopoietin 2, soluble thrombomodulin, and plasminogen activator inhibitor 1 were higher in nonsurvivors, and soluble receptor for advanced glycation end-products and surfactant protein D were higher in children meeting acute respiratory distress syndrome criteria. CONCLUSIONS PERSEVERE-II performs well in septic children from Aga Khan University Hospital, representing the first validation of PERSEVERE-II in a low-middle income country. Patients possessed a biomarker profile comparable to that of sepsis from high-income countries, suggesting that biomarker-based enrichment strategies may be effective in this setting.
Collapse
Affiliation(s)
- Sidra Ishaque
- Department of Pediatrics and Child Health, The Aga Khan University Hospital, Karachi, Pakistan
| | - Stephen Thomas Famularo
- Division of Pediatric Critical Care Medicine, Department of Anesthesiology and Critical Care, University of Pennsylvania and Children's Hospital of Philadelphia, Philadelphia, PA
| | - Ali Faisal Saleem
- Department of Pediatrics and Child Health, The Aga Khan University Hospital, Karachi, Pakistan
| | | | - Zaubina Kazi
- Department of Pediatrics and Child Health, The Aga Khan University Hospital, Karachi, Pakistan
| | - Sadia Parkar
- Department of Pediatrics and Child Health, The Aga Khan University Hospital, Karachi, Pakistan
| | - Aneeta Hotwani
- Department of Pediatrics and Child Health, The Aga Khan University Hospital, Karachi, Pakistan
| | - Neal J Thomas
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Penn State University College of Medicine, Hershey, PA
| | - Jill Marie Thompson
- Division of Pediatric Critical Care Medicine, Department of Anesthesiology and Critical Care, University of Pennsylvania and Children's Hospital of Philadelphia, Philadelphia, PA
| | - Patrick Lahni
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Brian Varisco
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- University of Cincinnati College of Medicine, Cincinnati, OH
| | - Nadir Yehya
- Division of Pediatric Critical Care Medicine, Department of Anesthesiology and Critical Care, University of Pennsylvania and Children's Hospital of Philadelphia, Philadelphia, PA
| |
Collapse
|
9
|
Zhao J, Zhen N, Zhou Q, Lou J, Cui W, Zhang G, Tian B. NETs Promote Inflammatory Injury by Activating cGAS-STING Pathway in Acute Lung Injury. Int J Mol Sci 2023; 24:ijms24065125. [PMID: 36982193 PMCID: PMC10049640 DOI: 10.3390/ijms24065125] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/12/2023] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) threatens the survival of critically ill patients, the mechanisms of which are still unclear. Neutrophil extracellular traps (NETs) released by activated neutrophils play a critical role in inflammatory injury. We investigated the role of NETs and the underlying mechanism involved in acute lung injury (ALI). We found a higher expression of NETs and cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) in the airways, which was reduced by Deoxyribonuclease I (DNase I) in ALI. The administration of the STING inhibitor H-151 also significantly relieved inflammatory lung injury, but failed to affect the high expression of NETs in ALI. We isolated murine neutrophils from bone marrow and acquired human neutrophils by inducing HL-60 to differentiate. After the PMA interventions, exogenous NETs were obtained from such extracted neutrophils. Exogenous NETs intervention in vitro and in vivo resulted in airway injury, and such inflammatory lung injury was reversed upon degrading NETs with or inhibiting cGAS-STING with H-151 as well as siRNA STING. In conclusion, cGAS-STING participates in regulating NETs-mediated inflammatory pulmonary injury, which is expected to be a new therapeutic target for ARDS/ALI.
Collapse
Affiliation(s)
- Jie Zhao
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd., Hangzhou 310009, China
| | - Ningxin Zhen
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd., Hangzhou 310009, China
| | - Qichao Zhou
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd., Hangzhou 310009, China
| | - Jian Lou
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Wei Cui
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd., Hangzhou 310009, China
| | - Gensheng Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd., Hangzhou 310009, China
- Correspondence: (G.Z.); (B.T.)
| | - Baoping Tian
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd., Hangzhou 310009, China
- Correspondence: (G.Z.); (B.T.)
| |
Collapse
|
10
|
Schnabel C, Harnisch LO, Walter D, Blaurock-Möller N, Bauer M, Quintel M, Kiehntopf M. Association of the C-terminal 42-peptide fragment of alpha-1 antitrypsin with the severity of ARDS: A pilot study. Clin Biochem 2023; 111:41-46. [PMID: 36244468 DOI: 10.1016/j.clinbiochem.2022.10.005] [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: 04/20/2022] [Revised: 08/01/2022] [Accepted: 10/10/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Acute respiratory distress syndrome is a life-threatening condition with a hospital mortality rate of up to 40%. Biomarkers related to the pathophysiology of ARDS may not only identify patients at risk but may also serve as potential therapeutic targets. This study examined the association between the proteolytic C-terminal 42-peptide fragment of alpha-1 antitrypsin and ARDS severity. METHODS The 42-peptide fragment and interleukin-6 levels were measured in 21 patients with mild-to-moderate ARDS and 47 patients with moderate-to-severe ARDS on days 1, 3, and 5 after diagnosis/admission to the intensive care unit. To elucidate the association between both biomarkers and the PaO2/FiO2 ratio, the concentrations of both biomarkers were compared between the two groups, and a multivariate regression analysis was performed. RESULTS The concentrations of both biomarkers were higher in patients with moderate-to-severe ARDS. While the PaO2/FiO2 ratio increased from day 1 to day 3, the concentrations of both biomarkers decreased. Multivariate regression analysis revealed negative associations between the PaO2/FiO2 ratio and both the C-terminal 42-peptide of alpha-1 antitrypsin and interleukin-6 on day 1 (beta: -0.138, p = 0.052; beta: -0.096, p = 0.004) and on day 3 (beta: -0.157, p = 0.045; beta: -0.106, p = 0.043). INTERPRETATION The C-terminal 42-peptide of alpha-1 antitrypsin is a new biomarker associated with ARDS severity. Its predictive value in identifying patients at risk of developing moderate-to-severe ARDS must be investigated in additional, independent prospective studies.
Collapse
Affiliation(s)
- Claudia Schnabel
- Department of Clinical Chemistry and Laboratory Medicine, Jena University Hospital, Am Klinikum 1, Jena 07747, Germany; Laboratory Dr. Fenner and Collegues, Bergstrasse 14, Hamburg 20095, Germany; Semmelweis University, Asklepios Medical School Hamburg, Lohmühlenstrasse 1, Hamburg 20099, Germany.
| | - Lars-Olav Harnisch
- Department of Anesthesiology, University of Göttingen Medical School, Robert-Koch-Strasse 40, Göttingen 37075, Germany.
| | - Dominic Walter
- Department of Clinical Chemistry and Laboratory Medicine, Jena University Hospital, Am Klinikum 1, Jena 07747, Germany.
| | - Nancy Blaurock-Möller
- Department of Clinical Chemistry and Laboratory Medicine, Jena University Hospital, Am Klinikum 1, Jena 07747, Germany.
| | - Michael Bauer
- Department of Anesthesiology, Jena University Hospital, Am Klinikum 1, Jena 07747, Germany.
| | - Michael Quintel
- Department of Anesthesiology, University of Göttingen Medical School, Robert-Koch-Strasse 40, Göttingen 37075, Germany.
| | - Michael Kiehntopf
- Department of Clinical Chemistry and Laboratory Medicine, Jena University Hospital, Am Klinikum 1, Jena 07747, Germany.
| |
Collapse
|
11
|
Blanchard F, James A, Assefi M, Kapandji N, Constantin JM. Personalized medicine targeting different ARDS phenotypes: The future of pharmacotherapy for ARDS? Expert Rev Respir Med 2023; 17:41-52. [PMID: 36724878 DOI: 10.1080/17476348.2023.2176302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Acute respiratory distress syndrome (ARDS) still represents a major challenge with high mortality rates and altered quality of life. Many well-designed studies have failed to improve ARDS outcomes. Heterogeneity of etiologies, mechanisms of lung damage, different lung mechanics, and different treatment approaches may explain these failures. At the era of personalized medicine, ARDS phenotyping is not only a field of research, but a bedside consideration when implementing therapy. ARDS has moved from being a simple syndrome to a more complex area of subgrouping. Intensivists must understand these phenotypes and therapies associated with a better outcome. AREAS COVERED After a brief sum-up of the different type of ARDS phenotypes, we will present some relevant therapy that may be impacted by phenotyping. A focus on pharmacotherapy will be realized before a section on non-pharmaceutical strategies. Eventually, we will highlight the limits of our knowledge of phenotyping and the pitfalls of personalized medicine. EXPERT OPINION Biological and morphological ARDS phenotypes are now well studied. The future of ARDS therapy will go through phenotyping that allows a personalized medication for each patient. However, a better assessment of these phenotypes is required, and clinical trials should be conducted with an ad-hoc phenotyping before randomization.
Collapse
Affiliation(s)
- Florian Blanchard
- Department of Anesthesiology and Critical Care, Pitié-Salpêtrière Hospital, Paris, France.,Antimicrobial Stewardship Team GH Paris Centre, Cochin Hospital, APHP, Paris, France
| | - Arthur James
- Department of Anesthesiology and Critical Care, Pitié-Salpêtrière Hospital, Paris, France
| | - Mona Assefi
- Department of Anesthesiology and Critical Care, Pitié-Salpêtrière Hospital, Paris, France
| | - Natacha Kapandji
- Department of Anesthesiology and Critical Care, Pitié-Salpêtrière Hospital, Paris, France
| | - Jean-Michel Constantin
- Department of Anesthesiology and Critical Care, Pitié-Salpêtrière Hospital, Paris, France
| |
Collapse
|
12
|
Xiong X, Dou J, Shi J, Ren Y, Wang C, Zhang Y, Cui Y. RAGE inhibition alleviates lipopolysaccharides-induced lung injury via directly suppressing autophagic apoptosis of type II alveolar epithelial cells. Respir Res 2023; 24:24. [PMID: 36691012 PMCID: PMC9872382 DOI: 10.1186/s12931-023-02332-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 01/17/2023] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Advanced glycation end product receptor (RAGE) acts as a receptor of pro-inflammatory ligands and is highly expressed in alveolar epithelial cells (AECs). Autophagy in AECs has received much attention recently. However, the roles of autophagy and RAGE in the pathogenesis of acute lung injury remain unclear. Therefore, this study aimed to explore whether RAGE activation signals take part in the dysfunction of alveolar epithelial barrier through autophagic death. METHODS Acute lung injury animal models were established using C57BL/6 and Ager gene knockout (Ager -/- mice) mice in this study. A549 cells and primary type II alveolar epithelial (ATII) cells were treated with siRNA to reduce Ager gene expression. Autophagy was inhibited by 3-methyladenine (3-MA). Lung injury was assessed by histopathological examination. Cell viability was estimated by cell counting kit-8 (CCK-8) assay. The serum and bronchoalveolar lavage fluid (BALF) levels of interleukin (IL)-6, IL-8 and soluble RAGE (sRAGE) were evaluated by Enzyme-linked immunosorbent assay (ELISA). The involvement of RAGE signals, autophagy and apoptosis was assessed using western blots, immunohistochemistry, immunofluorescence, transmission electron microscopy and TUNEL test. RESULTS The expression of RAGE was promoted by lipopolysaccharide (LPS), which was associated with activation of autophagy both in mice lung tissues and A549 cells as well as primary ATII cells. sRAGE in BALF was positively correlated with IL-6 and IL-8 levels. Compared with the wild-type mice, inflammation and apoptosis in lung tissues were alleviated in Ager-/- mice. Persistently activated autophagy contributed to cell apoptosis, whereas the inhibition of autophagy by 3-MA protected lungs from damage. In addition, Ager knockdown inhibited LPS-induced autophagy activation and attenuated lung injury. In vitro, knockdown of RAGE significantly suppressed the activation of LPS-induced autophagy and apoptosis of A549 and primary ATII cells. Furthermore, RAGE activated the downstream STAT3 signaling pathway. CONCLUSION RAGE plays an essential role in the pathogenesis of ATII cells injury. Our results suggested that RAGE inhibition alleviated LPS-induced lung injury by directly suppressing autophagic apoptosis of alveolar epithelial cells.
Collapse
Affiliation(s)
- Xi Xiong
- grid.16821.3c0000 0004 0368 8293Department of Critical Care Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293Institute of Pediatric Infection, Immunity and Intensive Care Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China
| | - Jiaying Dou
- grid.16821.3c0000 0004 0368 8293Department of Critical Care Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293Institute of Pediatric Infection, Immunity and Intensive Care Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China
| | - Jingyi Shi
- grid.16821.3c0000 0004 0368 8293Department of Critical Care Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, 200062 China
| | - Yuqian Ren
- grid.16821.3c0000 0004 0368 8293Department of Critical Care Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, 200062 China
| | - Chunxia Wang
- grid.16821.3c0000 0004 0368 8293Institute of Pediatric Infection, Immunity and Intensive Care Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.415625.10000 0004 0467 3069Clinical Research Unit, Shanghai Children’s Hospital, Shanghai, 200062 China
| | - Yucai Zhang
- grid.16821.3c0000 0004 0368 8293Department of Critical Care Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293Institute of Pediatric Infection, Immunity and Intensive Care Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, 200062 China
| | - Yun Cui
- grid.16821.3c0000 0004 0368 8293Department of Critical Care Medicine, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293Institute of Pediatric Infection, Immunity and Intensive Care Medicine, Shanghai Jiao Tong University, Shanghai, 200062 China ,grid.16821.3c0000 0004 0368 8293Institute of Pediatric Critical Care, Shanghai Jiao Tong University, Shanghai, 200062 China
| |
Collapse
|
13
|
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
|
14
|
Hernández-Díazcouder A, González-Ramírez J, Sanchez F, Leija-Martínez JJ, Martínez-Coronilla G, Amezcua-Guerra LM, Sánchez-Muñoz F. Negative Effects of Chronic High Intake of Fructose on Lung Diseases. Nutrients 2022; 14:nu14194089. [PMID: 36235741 PMCID: PMC9571075 DOI: 10.3390/nu14194089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 11/09/2022] Open
Abstract
In the modern diet, excessive fructose intake (>50 g/day) had been driven by the increase, in recent decades, of the consumption of sugar-sweetened beverages. This phenomenon has dramatically increased within the Caribbean and Latin American regions. Epidemiological studies show that chronic high intake of fructose related to sugar-sweetened beverages increases the risk of developing several non-communicable diseases, such as chronic obstructive pulmonary disease and asthma, and may also contribute to the exacerbation of lung diseases, such as COVID-19. Evidence supports several mechanisms—such as dysregulation of the renin−angiotensin system, increased uric acid production, induction of aldose reductase activity, production of advanced glycation end-products, and activation of the mTORC1 pathway—that can be implicated in lung damage. This review addresses how these pathophysiologic and molecular mechanisms may explain the lung damage resulting from high intake of fructose.
Collapse
Affiliation(s)
| | - Javier González-Ramírez
- Cellular Biology Laboratory, Faculty of Nursing, Universidad Autónoma de Baja California Campus Mexicali, Mexicali 21100, Mexico
| | - Fausto Sanchez
- Department of Agricultural and Animal Production, Universidad Autónoma Metropolitana Xochimilco, Mexico City 04960, Mexico
| | - José J. Leija-Martínez
- Master and Doctorate Program in Medical, Dental, and Health Sciences, Faculty of Medicine, Universidad Nacional Autónoma de México Campus Ciudad Universitaria, Mexico City 04510, Mexico
- Research Laboratory of Pharmacology, Hospital Infantil de Mexico Federico Gómez, Mexico City 06720, Mexico
| | - Gustavo Martínez-Coronilla
- Histology Laboratory, Faculty of Medicine, Universidad Autónoma de Baja California Campus Mexicali, Mexicali 21100, Mexico
| | - Luis M. Amezcua-Guerra
- Department of Immunology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| | - Fausto Sánchez-Muñoz
- Department of Immunology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
- Correspondence: ; Tel.: +52-5573-2911 (ext. 21310)
| |
Collapse
|
15
|
Receptor for Advanced Glycation End-Products Promotes Activation of Alveolar Macrophages through the NLRP3 Inflammasome/TXNIP Axis in Acute Lung Injury. Int J Mol Sci 2022; 23:ijms231911659. [PMID: 36232959 PMCID: PMC9569658 DOI: 10.3390/ijms231911659] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022] Open
Abstract
The roles of thioredoxin-interacting protein (TXNIP) and receptor for advanced glycation end-products (RAGE)-dependent mechanisms of NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome-driven macrophage activation during acute lung injury are underinvestigated. Cultured THP-1 macrophages were treated with a RAGE agonist (S100A12), with or without a RAGE antagonist; cytokine release and intracytoplasmic production of reactive oxygen species (ROS) were assessed in response to small interfering RNA knockdowns of TXNIP and NLRP3. Lung expressions of TXNIP and NLRP3 and alveolar levels of IL-1β and S100A12 were measured in mice after acid-induced lung injury, with or without administration of RAGE inhibitors. Alveolar macrophages from patients with acute respiratory distress syndrome and from mechanically ventilated controls were analyzed using fluorescence-activated cell sorting. In vitro, RAGE promoted cytokine release and ROS production in macrophages and upregulated NLRP3 and TXNIP mRNA expression in response to S100A12. TXNIP inhibition downregulated NLRP3 gene expression and RAGE-mediated release of IL-1β by macrophages in vitro. In vivo, RAGE, NLRP3 and TXNIP lung expressions were upregulated during experimental acute lung injury, a phenomenon being reversed by RAGE inhibition. The numbers of cells expressing RAGE, NLRP3 and TXNIP among a specific subpopulation of CD16+CD14+CD206- (“pro-inflammatory”) alveolar macrophages were higher in patients with lung injury. This study provides a novel proof-of-concept of complex RAGE–TXNIP–NLRP3 interactions during macrophage activation in acute lung injury.
Collapse
|
16
|
Soluble Receptor of Advanced Glycation End-Products (sRAGE) in Pediatric Asthma: A Prospective Study in 68 Children Aged 7 Years. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12125926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Asthma is a chronic inflammatory disease of the airways common in children. Soluble advanced glycation end-product receptor (sRAGE) is a blood biomarker of lung damage and inflammation. We sought to determine whether it could also be a biomarker in childhood asthma. Methods: We conducted a prospective, observational, analytical study at Clermont-Ferrand University Hospital. We measured plasma sRAGE levels in asthmatic and healthy children aged 7 years. Results: Of the 68 children assessed, 15 (22.05%) presented asthma. All presented normal respiratory function. The mean plasma sRAGE level was 1875 pg/mL in the children with asthma and 1794 pg/mL in the healthy children (p = 0.525). The mean plasma sRAGE level was significantly decreased with tobacco exposure during pregnancy: 1478 pg/mL versus 1870 pg/mL without (p = 0.007). Lower levels were observed in children living in apartments (1557 pg/mL) than in those living in houses (1863 pg/mL) (p = 0.031). Conclusions: No difference was observed in plasma sRAGE levels in children with asthma in our well-treated and controlled population. Environmental exposure may affect these levels. Further studies are required to better characterize the role of sRAGE.
Collapse
|
17
|
Lim MJ, Zinter MS, Chen L, Wong KMY, Bhalla A, Gala K, Guglielmo M, Alkhouli M, Huard LL, Hanudel MR, Vangala S, Schwingshackl A, Matthay M, Sapru A. Beyond the Alveolar Epithelium: Plasma Soluble Receptor for Advanced Glycation End Products Is Associated With Oxygenation Impairment, Mortality, and Extrapulmonary Organ Failure in Children With Acute Respiratory Distress Syndrome. Crit Care Med 2022; 50:837-847. [PMID: 34678846 PMCID: PMC9035468 DOI: 10.1097/ccm.0000000000005373] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVES Soluble receptor for advanced glycation end products is a known plasma marker of alveolar epithelial injury. However, RAGE is also expressed on cell types beyond the lung, and its activation leads to up-regulation of pro-inflammatory mediators. We sought to examine the relationship between plasma soluble receptor for advanced glycation end products and primary pulmonary dysfunction, extrapulmonary organ dysfunction, and mortality in pediatric acute respiratory distress syndrome patients at two early time points following acute respiratory distress syndrome diagnosis and compare these results to plasma surfactant protein-D, a marker of pure alveolar epithelial injury. DESIGN Prospective observational study. SETTING Five academic PICUs. PATIENTS Two hundred fifty-eight pediatric patients 30 days to 18 years old meeting Berlin Criteria for acute respiratory distress syndrome. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Plasma was collected for soluble receptor for advanced glycation end products and surfactant protein-D measurements within 24 hours (day 1) and 48 to 72 hours (day 3) after acute respiratory distress syndrome diagnosis. Similar to surfactant protein-D, plasma soluble receptor for advanced glycation end products was associated with a higher oxygenation index (p < 0.01) and worse lung injury score (p < 0.001) at the time of acute respiratory distress syndrome diagnosis. However, unlike surfactant protein-D, plasma soluble receptor for advanced glycation end products was associated with worse extrapulmonary Pediatric Logistic Organ Dysfunction score during ICU stay (day 3; p < 0.01) and positively correlated with plasma levels of interleukin-6 (p < 0.01), tumor necrosis factor-α (p < 0.01), and angiopoietin-2 (p < 0.01). Among children with indirect lung injury, plasma soluble receptor for advanced glycation end products was associated with mortality independent of age, sex, race, cancer/bone marrow transplant, and Pediatric Risk of Mortality score (day 3; odds ratio, 3.14; 95% CI, 1.46-6.75; p < 0.01). CONCLUSIONS Unlike surfactant protein-D, which is primarily localized to the alveolar epithelium plasma soluble receptor for advanced glycation end products is systemically expressed and correlates with markers of inflammation, extrapulmonary multiple organ dysfunction, and death in pediatric acute respiratory distress syndrome with indirect lung injury. This suggests that unlike surfactant protein-D, soluble receptor for advanced glycation end products is a multifaceted marker of alveolar injury and increased inflammation and that receptor for advanced glycation end products activation may contribute to the pathogenesis of multiple organ failure among children with indirect acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Michelle J. Lim
- UC Davis School of Medicine, UC Davis Children’s Hospital, Department of Pediatrics, Division of Critical Care, Sacramento, CA, USA
| | - Matt S. Zinter
- UCSF School of Medicine, Benioff Children’s Hospital, Department of Pediatrics, Division of Critical Care, San Francisco, CA, USA
| | - Lucia Chen
- UCLA Geffen School of Medicine, Mattel Children’s Hospital, Department of Pediatrics, Division of Critical Care, Los Angeles, CA, USA
| | - Kayley Man Yee Wong
- UCLA Geffen School of Medicine, Mattel Children’s Hospital, Department of Pediatrics, Division of Critical Care, Los Angeles, CA, USA
| | - Anoopindar Bhalla
- USC Keck School of Medicine, Children’s Hospital Los Angeles, Department of Anesthesiology and Critical Care Medicine, Los Angeles, CA, USA
| | - Kinisha Gala
- UCLA Geffen School of Medicine, Mattel Children’s Hospital, Department of Pediatrics, Division of Critical Care, Los Angeles, CA, USA
| | - Mona Guglielmo
- Loma Linda University School of Medicine, Loma Linda University Children’s Hospital, Department of Pediatrics, Division of Critical Care, Loma Linda, CA, USA
| | - Mustafa Alkhouli
- UCSF School of Medicine, Benioff Children’s Hospital, Department of Pediatrics, Division of Critical Care, San Francisco, CA, USA
| | - Leanna L. Huard
- UCLA Geffen School of Medicine, Mattel Children’s Hospital, Department of Pediatrics, Division of Critical Care, Los Angeles, CA, USA
| | - Mark R. Hanudel
- UCLA Geffen School of Medicine, Mattel Children’s Hospital, Department of Pediatrics, Division of Nephrology, Los Angeles, CA, USA
| | - Sitaram Vangala
- UCLA Geffen School of Medicine, Mattel Children’s Hospital, Department of Pediatrics, Division of Critical Care, Los Angeles, CA, USA
| | - Andreas Schwingshackl
- UCLA Geffen School of Medicine, Mattel Children’s Hospital, Department of Pediatrics, Division of Critical Care, Los Angeles, CA, USA
| | - Michael Matthay
- Departments of Medicine and Anesthesia and the Cardiovascular Research Institute, UCSF, San Francisco, CA, USA
| | - Anil Sapru
- UCLA Geffen School of Medicine, Mattel Children’s Hospital, Department of Pediatrics, Division of Critical Care, Los Angeles, CA, USA
| |
Collapse
|
18
|
Ramin S, Arcelli M, Bouchdoug K, Laumon T, Duflos C, De Jong A, Jaber S, Capdevila X, Charbit J. Driving pressure is not predictive of ARDS outcome in chest trauma patients under mechanical ventilation. Anaesth Crit Care Pain Med 2022; 41:101095. [PMID: 35489710 DOI: 10.1016/j.accpm.2022.101095] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/27/2022] [Accepted: 03/09/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND The relationship between the driving pressure of the respiratory system (ΔPrs) under mechanical ventilation and worse outcome has never been studied specifically in chest trauma patients. The objective of the present study was to assess in cases of chest trauma the relationship between ΔPrs and severity of acute respiratory distress syndrome (ARDS) or death and length of stay. METHODS A retrospective analysis of severe trauma patients (ISS > 15) with chest injuries admitted to the Trauma Centre from January 2010 to December 2018 was performed. Patients who received mechanical ventilation were included in our analysis. Mechanical ventilation parameters and ΔPrs were recorded during the stay in the intensive care unit. Association of ΔPrs with mortality and outcomes was specifically studied at the onset of ARDS (ΔPrs-ARDS) by receiver operator characteristic curve analysis, Kaplan-Meier curves, and multivariate analysis. RESULTS Among the 266 chest trauma patients studied, 194 (73%) developed ARDS. ΔPrs was significantly higher in the ARDS group versus in the no ARDS group (11.6 ± 2.4 cm H2O vs. 10.9 ± 1.9 cm H2O, p = 0.04). Among the patients with ARDS, no difference according to the duration of mechanical ventilation was found between the high ΔPrs group (ΔPrs-ARDS > 14 cm H2O) and the low ΔPrs group (ΔPrs-ARDS ≤ 14 cm H2O), (p = 0.75). ΔPrs-ARDS was not independently associated with the duration of mechanical ventilation (hazard ratio [HR], 1.006; 95% CI, 0.95-1.07; p = 0.8) or mortality (HR, 1.07; 95% CI, 0.9-1.28; p = 0.45). High mechanical power (≥ 12 J/min) was associated with a lower time for weaning of mechanical ventilation in Kaplan-Meier curves but not in multivariate analysis (HR, 0.98; 95% CI, 0.94-1.02; p = 0.22). CONCLUSION A high ΔPrs-ARDS was not significantly associated with an increase in mechanical ventilation duration or mortality risk in ARDS patients with chest trauma in contrast with medical patients.
Collapse
Affiliation(s)
- Severin Ramin
- Department of Anaesthesiology and Critical Care Medicine, Hôpital Lapeyronie, Montpellier, France; OcciTRAUMA Network, Regional Network of Medical Organization and Management for Severe Trauma in Occitanie, France.
| | - Matteo Arcelli
- Department of Anaesthesiology and Critical Care Medicine, Hôpital Lapeyronie, Montpellier, France; OcciTRAUMA Network, Regional Network of Medical Organization and Management for Severe Trauma in Occitanie, France
| | - Karim Bouchdoug
- Department of Anaesthesiology and Critical Care Medicine, Hôpital Lapeyronie, Montpellier, France; OcciTRAUMA Network, Regional Network of Medical Organization and Management for Severe Trauma in Occitanie, France; Department of Statistical Analysis, Montpellier, France
| | - Thomas Laumon
- Department of Anaesthesiology and Critical Care Medicine, Hôpital Lapeyronie, Montpellier, France; OcciTRAUMA Network, Regional Network of Medical Organization and Management for Severe Trauma in Occitanie, France
| | | | - Audrey De Jong
- Department of Anaesthesiology and Critical Care Medicine, Saint Eloi University Hospital, CHU Montpellier, University of Montpellier, Montpellier, France
| | - Samir Jaber
- Department of Anaesthesiology and Critical Care Medicine, Saint Eloi University Hospital, CHU Montpellier, University of Montpellier, Montpellier, France
| | - Xavier Capdevila
- Department of Anaesthesiology and Critical Care Medicine, Hôpital Lapeyronie, Montpellier, France; OcciTRAUMA Network, Regional Network of Medical Organization and Management for Severe Trauma in Occitanie, France
| | - Jonathan Charbit
- Department of Anaesthesiology and Critical Care Medicine, Hôpital Lapeyronie, Montpellier, France; OcciTRAUMA Network, Regional Network of Medical Organization and Management for Severe Trauma in Occitanie, France
| |
Collapse
|
19
|
Wu HP, Chu CM, Liu PH, Leu SW, Lin SW, Hu HC, Kao KC, Li LF, Yu CC. Increased Production of Interleukin-10 and Tumor Necrosis Factor-Alpha in Stimulated Peripheral Blood Mononuclear Cells after Inhibition of S100A12. Curr Issues Mol Biol 2022; 44:1701-1712. [PMID: 35723375 PMCID: PMC9164026 DOI: 10.3390/cimb44040117] [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: 03/04/2022] [Revised: 04/02/2022] [Accepted: 04/06/2022] [Indexed: 11/24/2022] Open
Abstract
Sepsis may induce immunosuppression and result in death. S100A12 can bind to the receptor for advanced glycation end-products (RAGE) and Toll-like receptor (TLR)4 following induction of various inflammatory responses. It is unclear whether S100A12 significantly influences the immune system, which may be associated with sepsis-related mortality. We measured plasma S100A12 levels and cytokine responses (mean ± standard error mean) of lipopolysaccharide (LPS)-stimulated peripheral blood mononuclear cells (PBMCs) after S100A12 inhibition in healthy controls and patients with sepsis on days one and seven. Day one plasma soluble RAGE (sRAGE) and S100A12 levels in patients with sepsis were significantly higher than those in controls (2481.3 ± 295.0 vs. 1273.0 ± 108.2 pg/mL, p < 0.001; 530.3 ± 18.2 vs. 310.1 ± 28.1 pg/mL, p < 0.001, respectively). Day seven plasma S100A12 levels in non-survivors were significantly higher than those in survivors (593.1 ± 12.7 vs. 499.3 ± 23.8 pg/mL, p = 0.002, respectively). In survivors, plasma sRAGE levels were significantly decreased after 6 days (2297.3 ± 320.3 vs. 1530.1 ± 219.1 pg/mL, p = 0.009, respectively), but not in non-survivors. Inhibiting S100A12 increased the production of tumor necrosis factor (TNF)-α and interleukin (IL)-10 in stimulated PBMCs for both controls and patients. Therefore, S100A12 plays an important role in sepsis pathogenesis. S100A12 may competitively bind to TLR4 and RAGE, resulting in decreased IL-10 and TNF-α production.
Collapse
Affiliation(s)
- Huang-Pin Wu
- Division of Pulmonary, Critical Care and Sleep Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (H.-P.W.); (C.-M.C.); (L.-F.L.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-W.L.); (S.-W.L.); (H.-C.H.); (K.-C.K.)
| | - Chien-Ming Chu
- Division of Pulmonary, Critical Care and Sleep Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (H.-P.W.); (C.-M.C.); (L.-F.L.)
| | - Pi-Hua Liu
- Clinical Informatics and Medical Statistics Research Center, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Shaw-Woei Leu
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-W.L.); (S.-W.L.); (H.-C.H.); (K.-C.K.)
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Shih-Wei Lin
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-W.L.); (S.-W.L.); (H.-C.H.); (K.-C.K.)
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Han-Chung Hu
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-W.L.); (S.-W.L.); (H.-C.H.); (K.-C.K.)
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Kuo-Chin Kao
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-W.L.); (S.-W.L.); (H.-C.H.); (K.-C.K.)
- Department of Pulmonary and Critical Care Medicine, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Li-Fu Li
- Division of Pulmonary, Critical Care and Sleep Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (H.-P.W.); (C.-M.C.); (L.-F.L.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-W.L.); (S.-W.L.); (H.-C.H.); (K.-C.K.)
| | - Chung-Chieh Yu
- Division of Pulmonary, Critical Care and Sleep Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan; (H.-P.W.); (C.-M.C.); (L.-F.L.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (S.-W.L.); (S.-W.L.); (H.-C.H.); (K.-C.K.)
| |
Collapse
|
20
|
Wick KD, Siegel L, Neaton JD, Oldmixon C, Lundgren J, Dewar RL, Lane HC, Thompson BT, Matthay MA. RAGE has potential pathogenetic and prognostic value in non-intubated hospitalized patients with COVID-19. JCI Insight 2022; 7:157499. [PMID: 35298440 PMCID: PMC9090244 DOI: 10.1172/jci.insight.157499] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/16/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The value of the soluble receptor for advanced glycation end-products (sRAGE) as a biomarker in COVID-19 is not well understood. We tested the association between plasma sRAGE and illness severity, viral burden, and clinical outcomes in non-mechanically ventilated hospitalized COVID-19 patients. METHODS Baseline sRAGE was measured among participants enrolled in the ACTIV-3/TICO trial of bamlanivimab for hospitalized COVID-19. Spearman rank correlation was used to assess the relationship between sRAGE and other plasma biomarkers, including viral nucleocapsid antigen. Fine-Gray models adjusted for baseline supplemental oxygen requirement, antigen level, positive endogenous antibody response, gender, age, body mass index, diabetes mellitus, renal impairment, and log2-transformed IL-6 level were used to assess the association between baseline sRAGE and time to sustained recovery. Cox regression adjusted for the same factors was used to assess the association between sRAGE and mortality. RESULTS Among 277 participants, baseline sRAGE was strongly correlated with viral plasma antigen concentration (ρ = 0.57). There was a weaker correlation between sRAGE and biomarkers of systemic inflammation such as IL-6 (ρ = 0.36) and CRP (ρ = 0.20). Participants with plasma sRAGE in the highest quartile had a significantly lower rate of sustained recovery (adjusted recovery rate ratio 0.64 [95% CI 0.43-0.90]) and a higher unadjusted risk of death (HR 4.70 [95% CI 2.01-10.99]) compared with participants in the lower quartiles. CONCLUSIONS Elevated plasma sRAGE in hospitalized, non-ventilated patients with COVID-19 was an indicator of both clinical illness severity and plasma viral load and was associated with a lower likelihood of sustained recovery. These novel results indicate that plasma sRAGE may be a promising biomarker for COVID-19 prognostication and clinical trial enrichment.
Collapse
Affiliation(s)
- Katherine D Wick
- Cardiovascular Research Institute, UCSF, San Francisco, United States of America
| | - Lianne Siegel
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, United States of America
| | - James D Neaton
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, United States of America
| | - Cathryn Oldmixon
- Division of Biostatistics, Massachusetts General Hospital, Boston, United States of America
| | - Jens Lundgren
- CHIP (Centre of Excellence for Health, Immunity and Infections), Rigshospit, University of Copenhagen, Copenhagen, Denmark
| | - Robin L Dewar
- Virus Isolation and Serology Laboratory, Applied and Developmental Director, Frederick National Laboratory, Frederick, United States of America
| | - H Clifford Lane
- Division of Clinical Research, NIAID, NIH, Bethesda, United States of America
| | - B Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, United States of America
| | - Michael A Matthay
- Cardiovascular Research Institute, UCSF, San Francisco, United States of America
| |
Collapse
|
21
|
Bos LDJ, Laffey JG, Ware LB, Heijnen NFL, Sinha P, Patel B, Jabaudon M, Bastarache JA, McAuley DF, Summers C, Calfee CS, Shankar-Hari M. Towards a biological definition of ARDS: are treatable traits the solution? Intensive Care Med Exp 2022; 10:8. [PMID: 35274164 PMCID: PMC8913033 DOI: 10.1186/s40635-022-00435-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 03/01/2022] [Indexed: 02/07/2023] Open
Abstract
The pathophysiology of acute respiratory distress syndrome (ARDS) includes the accumulation of protein-rich pulmonary edema in the air spaces and interstitial areas of the lung, variable degrees of epithelial injury, variable degrees of endothelial barrier disruption, transmigration of leukocytes, alongside impaired fluid and ion clearance. These pathophysiological features are different between patients contributing to substantial biological heterogeneity. In this context, it is perhaps unsurprising that a wide range of pharmacological interventions targeting these pathophysiological processes have failed to improve patient outcomes. In this manuscript, our goal is to provide a narrative summary of the potential methods to capture the underlying biological heterogeneity of ARDS and discuss how this information could inform future ARDS redefinitions. We discuss what biological tests are available to identify patients with any of the following predominant biological patterns: (1) epithelial and/or endothelial injury, (2) protein rich pulmonary edema and (3) systemic or within lung inflammatory responses.
Collapse
Affiliation(s)
- Lieuwe D J Bos
- Intensive Care, Amsterdam UMC, Location AMC, 1105AZ, Amsterdam, The Netherlands.
| | - John G Laffey
- Anaesthesia and Intensive Care Medicine, Galway University Hospitals, National University of Ireland Galway, Galway, Ireland
| | - Lorraine B Ware
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nanon F L Heijnen
- Department of Intensive Care Medicine, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Pratik Sinha
- Department of Anesthesiology, School of Medicine, Washington University, St. Louis, USA
| | - Brijesh Patel
- Division of Anaesthetics, Pain Medicine, and Intensive Care, Department of Surgery and Cancer, Imperial College, London, UK
| | - Matthieu Jabaudon
- Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France.,GReD, Université Clermont Auvergne, CNRS, INSERM, Clermont-Ferrand, France
| | - Julie A Bastarache
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Daniel F McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Charlotte Summers
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Carolyn S Calfee
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Manu Shankar-Hari
- School of Immunology and Microbial Sciences, King's College London, London, UK.,Centre for Inflammation Research, The University of Edinburgh, Edinburgh, Scotland, UK
| |
Collapse
|
22
|
Hoste L, Roels L, Naesens L, Bosteels V, Vanhee S, Dupont S, Bosteels C, Browaeys R, Vandamme N, Verstaen K, Roels J, Van Damme KF, Maes B, De Leeuw E, Declercq J, Aegerter H, Seys L, Smole U, De Prijck S, Vanheerswynghels M, Claes K, Debacker V, Van Isterdael G, Backers L, Claes KB, Bastard P, Jouanguy E, Zhang SY, Mets G, Dehoorne J, Vandekerckhove K, Schelstraete P, Willems J, Stordeur P, Janssens S, Beyaert R, Saeys Y, Casanova JL, Lambrecht BN, Haerynck F, Tavernier SJ. TIM3+ TRBV11-2 T cells and IFNγ signature in patrolling monocytes and CD16+ NK cells delineate MIS-C. J Exp Med 2022; 219:e20211381. [PMID: 34914824 PMCID: PMC8685281 DOI: 10.1084/jem.20211381] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/01/2021] [Accepted: 11/23/2021] [Indexed: 12/24/2022] Open
Abstract
In rare instances, pediatric SARS-CoV-2 infection results in a novel immunodysregulation syndrome termed multisystem inflammatory syndrome in children (MIS-C). We compared MIS-C immunopathology with severe COVID-19 in adults. MIS-C does not result in pneumocyte damage but is associated with vascular endotheliitis and gastrointestinal epithelial injury. In MIS-C, the cytokine release syndrome is characterized by IFNγ and not type I interferon. Persistence of patrolling monocytes differentiates MIS-C from severe COVID-19, which is dominated by HLA-DRlo classical monocytes. IFNγ levels correlate with granzyme B production in CD16+ NK cells and TIM3 expression on CD38+/HLA-DR+ T cells. Single-cell TCR profiling reveals a skewed TCRβ repertoire enriched for TRBV11-2 and a superantigenic signature in TIM3+/CD38+/HLA-DR+ T cells. Using NicheNet, we confirm IFNγ as a central cytokine in the communication between TIM3+/CD38+/HLA-DR+ T cells, CD16+ NK cells, and patrolling monocytes. Normalization of IFNγ, loss of TIM3, quiescence of CD16+ NK cells, and contraction of patrolling monocytes upon clinical resolution highlight their potential role in MIS-C immunopathogenesis.
Collapse
Affiliation(s)
- Levi Hoste
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Division of Pediatric Pulmonology, Infectious Diseases and Inborn Errors of Immunity, Ghent University Hospital, Ghent, Belgium
| | - Lisa Roels
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Division of Pediatric Pulmonology, Infectious Diseases and Inborn Errors of Immunity, Ghent University Hospital, Ghent, Belgium
| | - Leslie Naesens
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Division of Pediatric Pulmonology, Infectious Diseases and Inborn Errors of Immunity, Ghent University Hospital, Ghent, Belgium
| | - Victor Bosteels
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Center for Inflammation Research, Laboratory for Endoplasmic Reticulum Stress and Inflammation, VIB, Ghent, Belgium
| | - Stijn Vanhee
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Center for Inflammation Research, Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
| | - Sam Dupont
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Center for Inflammation Research, Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
| | - Cedric Bosteels
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Center for Inflammation Research, Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
| | - Robin Browaeys
- Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Niels Vandamme
- Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Kevin Verstaen
- Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Jana Roels
- Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Karel F.A. Van Damme
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Center for Inflammation Research, Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
| | - Bastiaan Maes
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Center for Inflammation Research, Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
| | - Elisabeth De Leeuw
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Center for Inflammation Research, Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
| | - Jozefien Declercq
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Center for Inflammation Research, Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
| | - Helena Aegerter
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Center for Inflammation Research, Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
| | - Leen Seys
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Center for Inflammation Research, Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
| | - Ursula Smole
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Center for Inflammation Research, Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
| | - Sofie De Prijck
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Center for Inflammation Research, Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
| | - Manon Vanheerswynghels
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Center for Inflammation Research, Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
| | - Karlien Claes
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Division of Pediatric Pulmonology, Infectious Diseases and Inborn Errors of Immunity, Ghent University Hospital, Ghent, Belgium
| | - Veronique Debacker
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Division of Pediatric Pulmonology, Infectious Diseases and Inborn Errors of Immunity, Ghent University Hospital, Ghent, Belgium
| | | | - Lynn Backers
- Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University and Ghent University Hospital, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Kathleen B.M. Claes
- Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University and Ghent University Hospital, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent University, Ghent, Belgium
| | - Paul Bastard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Emmanuelle Jouanguy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Shen-Ying Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Gilles Mets
- Department of Internal Medicine and Pediatrics, Division of Pediatric Cardiology, Ghent University Hospital, Ghent, Belgium
| | - Joke Dehoorne
- Department of Internal Medicine and Pediatrics, Division of Pediatric Rheumatology, Ghent University Hospital, Ghent, Belgium
| | - Kristof Vandekerckhove
- Department of Internal Medicine and Pediatrics, Division of Pediatric Cardiology, Ghent University Hospital, Ghent, Belgium
| | - Petra Schelstraete
- Department of Internal Medicine and Pediatrics, Division of Pediatric Pulmonology, Infectious Diseases and Inborn Errors of Immunity, Ghent University Hospital, Ghent, Belgium
| | - Jef Willems
- Department of Critical Care, Division of Pediatric Intensive Care, Ghent University Hospital, Ghent, Belgium
| | | | - Patrick Stordeur
- Belgian National Reference Center for the Complement System, Laboratory of Immunology, LHUB-ULB, Université Libre de Bruxelles, Brussels, Belgium
| | - Sophie Janssens
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Center for Inflammation Research, Laboratory for Endoplasmic Reticulum Stress and Inflammation, VIB, Ghent, Belgium
| | - Rudi Beyaert
- Center for Inflammation Research, Laboratory of Molecular Signal Transduction in Inflammation, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Yvan Saeys
- Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
- Howard Hughes Medical Institute, New York, NY
- Pediatric Hematology and Immunology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Bart N. Lambrecht
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, Ghent, Belgium
- Center for Inflammation Research, Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Pulmonary Medicine, ErasmusMC, Rotterdam, The Netherlands
| | - Filomeen Haerynck
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Division of Pediatric Pulmonology, Infectious Diseases and Inborn Errors of Immunity, Ghent University Hospital, Ghent, Belgium
| | - Simon J. Tavernier
- Primary Immune Deficiency Research Laboratory, Department of Internal Diseases and Pediatrics, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University, Ghent, Belgium
- Center for Inflammation Research, Laboratory of Molecular Signal Transduction in Inflammation, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
23
|
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: 4.7] [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
|
24
|
Ider M, Naseri A, Ok M, Gulersoy E, Bas TM, Uney K, Parlak TM, Abdelaziz A. Serum sRAGE and sE-selectin levels are useful biomarkers of lung injury and prediction of mortality in calves with perinatal asphyxia. Theriogenology 2022; 181:113-118. [PMID: 35078123 DOI: 10.1016/j.theriogenology.2022.01.019] [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: 09/27/2021] [Revised: 01/15/2022] [Accepted: 01/16/2022] [Indexed: 10/19/2022]
Abstract
The objective of the present study was to evaluate the biomarkers specific to lung endothelial and epithelial damage in the determination of lung injury and its severity in calves with perinatal asphyxia and to evaluate their prognostic importance among survivors and non-survivor calves. Ten healthy calves and 20 calves with perinatal asphyxia were enrolled in the study. Clinical examination and laboratory analysis were performed at admission. Serum concentrations of soluble advanced glycation end-product receptor (sRAGE), soluble E-selectin (sE-selectin), clara cell secretory protein (CC16), and soluble intercellular adhesion molecule-1 (sICAM-1) were measured to assess lung injury. Venous pH, sO2, HCO3, and BE of calves with perinatal asphyxia were significantly lower than the healthy calves. sRAGE, sE-selectin, pCO2, and lactate were significantly high in calves with asphyxia. ROC analysis showed that sRAGE, sE-selectin, pCO2, lactate, and respiratory rate were higher while HCO3 and BE were lower in the nonsurvivor calves than survivors. In conclusion, serum sRAGE and sE-selectin concentrations highlight the utility of these biomarkers in determining lung injury in calves with asphyxia. Also, pH, pCO2, lactate, HCO3, BE, and respiratory rate along with serum sRAGE and sE-selectin were useful indicators in the prediction of mortality.
Collapse
Affiliation(s)
- M Ider
- Selcuk University, Faculty of Veterinary Medicine, Department of Internal Medicine, Konya, 42003, Turkey.
| | - A Naseri
- Selcuk University, Faculty of Veterinary Medicine, Department of Internal Medicine, Konya, 42003, Turkey
| | - M Ok
- Selcuk University, Faculty of Veterinary Medicine, Department of Internal Medicine, Konya, 42003, Turkey
| | - E Gulersoy
- Harran University, Faculty of Veterinary Medicine, Department of Internal Medicine, Sanlıurfa, 63200, Turkey
| | - T M Bas
- Selcuk University, Faculty of Veterinary Medicine, Department of Internal Medicine, Konya, 42003, Turkey
| | - K Uney
- Selcuk University, Faculty of Veterinary Medicine, Department of Pharmacology and Toxicology, Konya, 42003, Turkey
| | - T M Parlak
- Selcuk University, Faculty of Veterinary Medicine, Department of Pharmacology and Toxicology, Konya, 42003, Turkey
| | - A Abdelaziz
- Selcuk University, Faculty of Veterinary Medicine, Department of Internal Medicine, Konya, 42003, Turkey
| |
Collapse
|
25
|
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: 1.5] [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
|
26
|
Sinha P, Bos LD. Pathophysiology of the Acute Respiratory Distress Syndrome: Insights from Clinical Studies. Crit Care Clin 2021; 37:795-815. [PMID: 34548134 PMCID: PMC8149201 DOI: 10.1016/j.ccc.2021.05.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Pratik Sinha
- Division of Clinical and Translational Research, Department of Anesthesia, Washington University School of Medicine, 660 S. Euclid Avenue, Campus Box 8054, St Louis, MO 63110, USA.
| | - Lieuwe D Bos
- Department of Respiratory Medicine, Infection and Immunity, Amsterdam University Medical Center, AMC, Meibergdreef 9, Amsterdam 1105AZ, The Netherlands
| |
Collapse
|
27
|
First-Days Reduction of Plasma and Skin Advanced Glycation End Products is Related to Outcome in Septic Patients. Shock 2021; 53:400-406. [PMID: 31232862 DOI: 10.1097/shk.0000000000001396] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Advanced glycation end products (AGEs) are a result of nonenzymatic glycation of proteins and lipids, which can attach to either their cell surface receptor (RAGE) or its soluble form (sRAGE). Evidence exists for the implication of AGE-RAGE axis in sepsis, but data are still insufficient and conflicting. We aimed to analyze the kinetics of plasma and skin AGEs and sRAGE during sepsis, and their association with outcome in septic patients. METHODS We performed a prospective observational study. We enrolled 90 consecutive patients with severe sepsis or septic shock, within the first 24 h of Intensive Care Unit admission. During the first 5 days of sepsis, we measured plasma autofluorescence (PAF) and skin autofluorescence (SAF) as surrogates of circulating and skin AGEs, respectively. sRAGE was measured on days 1, 3, and 5. Delta values were defined as the difference between the PAF, SAF, or sRAGE on a specific day and the value on day 1. RESULTS 28-day mortality was 18%. Bivariate analysis found that ΔPAF3-1, ΔPAF4-1, ΔPAF5-1, and ΔSAF5-1 were significantly associated with 28-day mortality. Additionally, sRAGE1 was inversely correlated to ΔPAF4-1 (r = -0.250, P = 0.019) and ΔPAF5-1 (r = -0.246, P = 0.024), and significantly associated with 28-day mortality. In an adjusted multivariate logistic regression analysis, ΔPAF2-1, ΔPAF3-1, ΔPAF4-1, ΔPAF5-1, and ΔSAF5-1 were associated with 28-day mortality. CONCLUSIONS Kinetics of plasma and skin AGEs during the first days of sepsis are independently associated with mortality, where a decrease of plasma and skin AGEs are related to higher mortality.
Collapse
|
28
|
Gutmann C, Takov K, Burnap SA, Singh B, Ali H, Theofilatos K, Reed E, Hasman M, Nabeebaccus A, Fish M, McPhail MJ, O'Gallagher K, Schmidt LE, Cassel C, Rienks M, Yin X, Auzinger G, Napoli S, Mujib SF, Trovato F, Sanderson B, Merrick B, Niazi U, Saqi M, Dimitrakopoulou K, Fernández-Leiro R, Braun S, Kronstein-Wiedemann R, Doores KJ, Edgeworth JD, Shah AM, Bornstein SR, Tonn T, Hayday AC, Giacca M, Shankar-Hari M, Mayr M. SARS-CoV-2 RNAemia and proteomic trajectories inform prognostication in COVID-19 patients admitted to intensive care. Nat Commun 2021; 12:3406. [PMID: 34099652 PMCID: PMC8184784 DOI: 10.1038/s41467-021-23494-1] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 04/28/2021] [Indexed: 02/05/2023] Open
Abstract
Prognostic characteristics inform risk stratification in intensive care unit (ICU) patients with coronavirus disease 2019 (COVID-19). We obtained blood samples (n = 474) from hospitalized COVID-19 patients (n = 123), non-COVID-19 ICU sepsis patients (n = 25) and healthy controls (n = 30). Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA was detected in plasma or serum (RNAemia) of COVID-19 ICU patients when neutralizing antibody response was low. RNAemia is associated with higher 28-day ICU mortality (hazard ratio [HR], 1.84 [95% CI, 1.22-2.77] adjusted for age and sex). RNAemia is comparable in performance to the best protein predictors. Mannose binding lectin 2 and pentraxin-3 (PTX3), two activators of the complement pathway of the innate immune system, are positively associated with mortality. Machine learning identified 'Age, RNAemia' and 'Age, PTX3' as the best binary signatures associated with 28-day ICU mortality. In longitudinal comparisons, COVID-19 ICU patients have a distinct proteomic trajectory associated with mortality, with recovery of many liver-derived proteins indicating survival. Finally, proteins of the complement system and galectin-3-binding protein (LGALS3BP) are identified as interaction partners of SARS-CoV-2 spike glycoprotein. LGALS3BP overexpression inhibits spike-pseudoparticle uptake and spike-induced cell-cell fusion in vitro.
Collapse
Affiliation(s)
- Clemens Gutmann
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Kaloyan Takov
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Sean A Burnap
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Bhawana Singh
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Hashim Ali
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Konstantinos Theofilatos
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Ella Reed
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Maria Hasman
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Adam Nabeebaccus
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
- King's College Hospital NHS Foundation Trust, London, UK
| | - Matthew Fish
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
- Department of Intensive Care Medicine, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Mark Jw McPhail
- King's College Hospital NHS Foundation Trust, London, UK
- Department of Inflammation Biology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
- Institute of Liver Studies, King's College Hospital, London, UK
| | - Kevin O'Gallagher
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
- King's College Hospital NHS Foundation Trust, London, UK
| | - Lukas E Schmidt
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Christian Cassel
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Marieke Rienks
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Xiaoke Yin
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Georg Auzinger
- King's College Hospital NHS Foundation Trust, London, UK
| | - Salvatore Napoli
- Department of Inflammation Biology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Salma F Mujib
- Institute of Liver Studies, King's College Hospital, London, UK
| | - Francesca Trovato
- King's College Hospital NHS Foundation Trust, London, UK
- Department of Inflammation Biology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
- Institute of Liver Studies, King's College Hospital, London, UK
| | - Barnaby Sanderson
- Department of Intensive Care Medicine, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Blair Merrick
- Clinical Infection and Diagnostics Research group, Department of Infection, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Umar Niazi
- NIHR Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Mansoor Saqi
- NIHR Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Konstantina Dimitrakopoulou
- NIHR Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Rafael Fernández-Leiro
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Silke Braun
- Medical Clinic I, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Romy Kronstein-Wiedemann
- Experimental Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Katie J Doores
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Jonathan D Edgeworth
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
- Clinical Infection and Diagnostics Research group, Department of Infection, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Ajay M Shah
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
- King's College Hospital NHS Foundation Trust, London, UK
| | - Stefan R Bornstein
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Department of Diabetes, School of Life Course Science and Medicine, King's College London, London, UK
| | - Torsten Tonn
- Experimental Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Institute for Transfusion Medicine, German Red Cross Blood Donation Service North East, Dresden, Germany
| | - Adrian C Hayday
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
- The Francis Crick Institute, London, UK
| | - Mauro Giacca
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK
| | - Manu Shankar-Hari
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK.
- Department of Intensive Care Medicine, Guy's and St Thomas' NHS Foundation Trust, London, UK.
| | - Manuel Mayr
- King's College London British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, UK.
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany.
| |
Collapse
|
29
|
Radiological pattern in ARDS patients: partitioned respiratory mechanics, gas exchange and lung recruitability. Ann Intensive Care 2021; 11:78. [PMID: 33999274 PMCID: PMC8128955 DOI: 10.1186/s13613-021-00870-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 05/05/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The ARDS is characterized by different degrees of impairment in oxygenation and distribution of the lung disease. Two radiological patterns have been described: a focal and a diffuse one. These two patterns could present significant differences both in gas exchange and in the response to a recruitment maneuver. At the present time, it is not known if the focal and the diffuse pattern could be characterized by a difference in the lung and chest wall mechanical characteristics. Our aims were to investigate, at two levels of PEEP, if focal vs. diffuse ARDS patterns could be characterized by different lung CT characteristics, partitioned respiratory mechanics and lung recruitability. METHODS CT patterns were analyzed by two radiologists and were classified as focal or diffuse. The changes from 5 to 15 cmH2O in blood gas analysis and partitioned respiratory mechanics were analyzed. Lung CT scan was performed at 5 and 45 cmH2O of PEEP to evaluate lung recruitability. RESULTS One-hundred and ten patients showed a diffuse pattern, while 58 showed a focal pattern. At 5 cmH2O of PEEP, the driving pressure and the elastance, both the respiratory system and of the lung, were significantly higher in the diffuse pattern compared to the focal (14 [11-16] vs 11 [9-15 cmH2O; 28 [23-34] vs 21 [17-27] cmH2O/L; 22 [17-28] vs 14 [12-19] cmH2O/L). By increasing PEEP, the driving pressure and the respiratory system elastance significantly decreased in diffuse pattern, while they increased or did not change in the focal pattern (Δ15-5: - 1 [- 2 to 1] vs 0 [- 1 to 2]; - 1 [- 4 to 2] vs 1 [- 2 to 5]). At 5 cmH2O of PEEP, the diffuse pattern had a lower lung gas (743 [537-984] vs 1222 [918-1974] mL) and higher lung weight (1618 [1388-2001] vs 1222 [1059-1394] g) compared to focal pattern. The lung recruitability was significantly higher in diffuse compared to focal pattern 21% [13-29] vs 11% [6-16]. Considering the median of lung recruitability of the whole population (16.1%), the recruiters were 65% and 22% in the diffuse and focal pattern, respectively. CONCLUSIONS An early identification of lung morphology can be useful to choose the ventilatory setting. A diffuse pattern has a better response to the increase of PEEP and to the recruitment maneuver.
Collapse
|
30
|
Changes in Plasma Soluble Receptor for Advanced Glycation End-Products Are Associated with Survival in Patients with Acute Respiratory Distress Syndrome. J Clin Med 2021; 10:jcm10102076. [PMID: 34066048 PMCID: PMC8150905 DOI: 10.3390/jcm10102076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/08/2021] [Accepted: 05/10/2021] [Indexed: 01/11/2023] Open
Abstract
The plasma soluble receptor for advanced glycation end-products (sRAGE) is a marker of lung epithelial injury with prognostic value when measured at baseline in acute respiratory distress syndrome (ARDS). However, whether changes in plasma sRAGE could inform prognosis in ARDS remains unknown. In this secondary analysis of the Lung Imaging for Ventilator Setting in ARDS (LIVE) multicenter randomized controlled trial, which evaluated a personalized ventilation strategy tailored to lung morphology, plasma sRAGE was measured upon study entry (baseline) and on days one, two, three, four and six. The association between changes in plasma sRAGE over time and 90-day survival was evaluated. Higher baseline plasma sRAGE (HR per-one log increment, 1.53; 95% CI, 1.16–2.03; p = 0.003) and an increase in sRAGE over time (HR for each one-log increment in plasma sRAGE per time unit, 1.01; 95% CI, 1.01–1.02; p < 10−3) were both associated with increased 90-day mortality. Each 100-unit increase in the plasma sRAGE level per unit of time increased the risk of death at day 90 by 1% in joint modeling. Plasma sRAGE increased over time when a strategy of maximal alveolar recruitment was applied in patients with focal ARDS. Current findings suggest that the rate of change in plasma sRAGE over time is associated with 90-day survival and could be helpful as a surrogate outcome in ARDS.
Collapse
|
31
|
Wendel Garcia PD, Caccioppola A, Coppola S, Pozzi T, Ciabattoni A, Cenci S, Chiumello D. Latent class analysis to predict intensive care outcomes in Acute Respiratory Distress Syndrome: a proposal of two pulmonary phenotypes. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2021; 25:154. [PMID: 33888134 PMCID: PMC8060783 DOI: 10.1186/s13054-021-03578-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 04/13/2021] [Indexed: 02/07/2023]
Abstract
Background Acute respiratory distress syndrome remains a heterogeneous syndrome for clinicians and researchers difficulting successful tailoring of interventions and trials. To this moment, phenotyping of this syndrome has been approached by means of inflammatory laboratory panels. Nevertheless, the systemic and inflammatory expression of acute respiratory distress syndrome might not reflect its respiratory mechanics and gas exchange. Methods Retrospective analysis of a prospective cohort of two hundred thirty-eight patients consecutively admitted patients under mechanical ventilation presenting with acute respiratory distress syndrome. All patients received standardized monitoring of clinical variables, respiratory mechanics and computed tomography scans at predefined PEEP levels. Employing latent class analysis, an unsupervised structural equation modelling method, on respiratory mechanics, gas-exchange and computed tomography-derived gas- and tissue-volumes at a PEEP level of 5cmH2O, distinct pulmonary phenotypes of acute respiratory distress syndrome were identified. Results Latent class analysis was applied to 54 respiratory mechanics, gas-exchange and CT-derived gas- and tissue-volume variables, and a two-class model identified as best fitting. Phenotype 1 (non-recruitable) presented lower respiratory system elastance, alveolar dead space and amount of potentially recruitable lung volume than phenotype 2 (recruitable). Phenotype 2 (recruitable) responded with an increase in ventilated lung tissue, compliance and PaO2/FiO2 ratio (p < 0.001), in addition to a decrease in alveolar dead space (p < 0.001), to a standardized recruitment manoeuvre. Patients belonging to phenotype 2 (recruitable) presented a higher intensive care mortality (hazard ratio 2.9, 95% confidence interval 1.7–2.7, p = 0.001). Conclusions The present study identifies two ARDS phenotypes based on respiratory mechanics, gas-exchange and computed tomography-derived gas- and tissue-volumes. These phenotypes are characterized by distinctly diverse responses to a standardized recruitment manoeuvre and by a diverging mortality. Given multicentre validation, the simple and rapid identification of these pulmonary phenotypes could facilitate enrichment of future prospective clinical trials addressing mechanical ventilation strategies in ARDS. Supplementary Information The online version contains supplementary material available at 10.1186/s13054-021-03578-6.
Collapse
Affiliation(s)
- Pedro D Wendel Garcia
- Institute of Intensive Care Medicine, University Hospital of Zurich, Zurich, Switzerland
| | - Alessio Caccioppola
- Department of Anesthesia and Intensive Care, ASST Santi Paolo E Carlo, San Paolo University Hospital, Via Di Rudinì, Milan, Italy.,Department of Health Sciences, University of Milan, Milan, Italy
| | - Silvia Coppola
- Department of Anesthesia and Intensive Care, ASST Santi Paolo E Carlo, San Paolo University Hospital, Via Di Rudinì, Milan, Italy
| | - Tommaso Pozzi
- Department of Anesthesia and Intensive Care, ASST Santi Paolo E Carlo, San Paolo University Hospital, Via Di Rudinì, Milan, Italy.,Department of Health Sciences, University of Milan, Milan, Italy
| | - Arianna Ciabattoni
- Department of Anesthesia and Intensive Care, ASST Santi Paolo E Carlo, San Paolo University Hospital, Via Di Rudinì, Milan, Italy.,Department of Health Sciences, University of Milan, Milan, Italy
| | - Stefano Cenci
- Department of Anesthesia and Intensive Care, ASST Santi Paolo E Carlo, San Paolo University Hospital, Via Di Rudinì, Milan, Italy.,Department of Health Sciences, University of Milan, Milan, Italy
| | - Davide Chiumello
- Department of Anesthesia and Intensive Care, ASST Santi Paolo E Carlo, San Paolo University Hospital, Via Di Rudinì, Milan, Italy. .,Department of Health Sciences, University of Milan, Milan, Italy. .,Coordinated Research Center on Respiratory Failure, University of Milan, Milan, Italy.
| |
Collapse
|
32
|
Lim A, Radujkovic A, Weigand MA, Merle U. Soluble receptor for advanced glycation end products (sRAGE) as a biomarker of COVID-19 disease severity and indicator of the need for mechanical ventilation, ARDS and mortality. Ann Intensive Care 2021; 11:50. [PMID: 33751264 PMCID: PMC7983090 DOI: 10.1186/s13613-021-00836-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/08/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND COVID-19 pneumonia and subsequent respiratory failure is causing an immense strain on intensive care units globally. Early prediction of severe disease enables clinicians to avoid acute respiratory distress syndrome (ARDS) development and improve management of critically ill patients. The soluble receptor of advanced glycation endproducts (sRAGE) is a biomarker shown to predict ARDS. Although sRAGE level varies depending on the type of disease, there is limited information available on changes in sRAGE levels in COVID-19. Therefore, sRAGE was measured in COVID-19 patients to determine sRAGE level variation in COVID-19 severity and to examine its ability to predict the need for mechanical ventilation (MV) and mortality in COVID-19. METHODS In this single-centre observational cohort study in Germany, serum sRAGE during acute COVID-19, 20 weeks after the start of COVID-19 symptoms, as well as in control groups of non-COVID-19 pneumonia patients and healthy controls were measured using ELISA. The primary endpoint was severe disease (high-flow nasal oxygen therapy (HFNO)/MV and need of organ support). The secondary endpoints were respiratory failure with need of MV and 30-day mortality. The area under the curve (AUC), cut-off based on Youden's index and odds ratio with 95% CI for sRAGE were calculated with regard to prediction of MV need and mortality. RESULTS Serum sRAGE in 164 COVID-19 patients, 101 matched COVID-19 convalescent patients, 23 non-COVID-19 pneumonia patients and 15 healthy volunteers were measured. sRAGE level increased with COVID-19 severity, need for oxygen therapy, HFNO/MV, ARDS severity, need of dialysis and catecholamine support, 30-day mortality, sequential organ failure assessment (SOFA) and quick SOFA (qSOFA) score. sRAGE was found to be a good predictor of MV need in COVID-19 inpatients and mortality with an AUC of 0.871 (0.770-0.973) and 0.903 (0.817-0.990), respectively. When adjusted for male gender, age, comorbidity and SOFA score ≥ 3, sRAGE was independently associated with risk of need for HFNO/MV. When adjusted for SOFA score ≥ 3, sRAGE was independently associated with risk of need for MV. CONCLUSIONS Serum sRAGE concentrations are elevated in COVID-19 patients as disease severity increases. sRAGE should be considered as a biomarker for predicting the need for MV and mortality in COVID-19.
Collapse
Affiliation(s)
- Adeline Lim
- Department of Internal Medicine IV, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
| | - Aleksandar Radujkovic
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Markus A Weigand
- Department of Anesthesiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Uta Merle
- Department of Internal Medicine IV, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| |
Collapse
|
33
|
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
|
34
|
Sivapalan P, Bonnesen B, Jensen JU. Novel Perspectives Regarding the Pathology, Inflammation, and Biomarkers of Acute Respiratory Distress Syndrome. Int J Mol Sci 2020; 22:E205. [PMID: 33379178 PMCID: PMC7796016 DOI: 10.3390/ijms22010205] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 12/29/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is an acute inflammation of the lung resulting from damage to the alveolar-capillary membrane, and it is diagnosed using a combination of clinical and physiological variables. ARDS develops in approximately 10% of hospitalised patients with pneumonia and has a mortality rate of approximately 40%. Recent research has identified several biomarkers associated with ARDS pathophysiology, and these may be useful for diagnosing and monitoring ARDS. They may also highlight potential therapeutic targets. This review summarises our current understanding of those clinical biomarkers: (1) biomarkers of alveolar and bronchiolar injury, (2) biomarkers of endothelial damage and coagulation, and (3) biomarkers for treatment responses.
Collapse
Affiliation(s)
- Pradeesh Sivapalan
- Respiratory Medicine Section, Department of Internal Medicine, Herlev and Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark; (B.B.); (J.-U.J.)
| | | | | |
Collapse
|
35
|
Pierrakos C, Velissaris D, Bisdorff M, Marshall JC, Vincent JL. Biomarkers of sepsis: time for a reappraisal. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:287. [PMID: 32503670 PMCID: PMC7273821 DOI: 10.1186/s13054-020-02993-5] [Citation(s) in RCA: 300] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/14/2020] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Sepsis biomarkers can have important diagnostic, therapeutic, and prognostic functions. In a previous review, we identified 3370 references reporting on 178 different biomarkers related to sepsis. In the present review, we evaluate the progress in the research of sepsis biomarkers. METHODS Using the same methodology as in our previous review, we searched the PubMed database from 2009 until September 2019 using the terms "Biomarker" AND "Sepsis." There were no restrictions by age or language, and all studies, clinical and experimental, were included. RESULTS We retrieved a total of 5367 new references since our previous review. We identified 258 biomarkers, 80 of which were new compared to our previous list. The majority of biomarkers have been evaluated in fewer than 5 studies, with 81 (31%) being assessed in just a single study. Apart from studies of C-reactive protein (CRP) or procalcitonin (PCT), only 26 biomarkers have been assessed in clinical studies with more than 300 participants. Forty biomarkers have been compared to PCT and/or CRP for their diagnostic value; 9 were shown to have a better diagnostic value for sepsis than either or both of these biomarkers. Forty-four biomarkers have been evaluated for a role in answering a specific clinical question rather than for their general diagnostic or prognostic properties in sepsis. CONCLUSIONS The number of biomarkers being identified is still increasing although at a slower rate than in the past. Most of the biomarkers have not been well-studied; in particular, the clinical role of these biomarkers needs to be better evaluated.
Collapse
Affiliation(s)
- Charalampos Pierrakos
- Intensive Care Department, Brugmann University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Max Bisdorff
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium
| | - John C Marshall
- Surgery/Critical Care Medicine, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Jean-Louis Vincent
- Department of Intensive Care, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium.
| |
Collapse
|
36
|
Zhai R, Blondonnet R, Ebrahimi E, Belville C, Audard J, Gross C, Choltus H, Henrioux F, Constantin JM, Pereira B, Blanchon L, Sapin V, Jabaudon M. The receptor for advanced glycation end-products enhances lung epithelial wound repair: An in vitro study. Exp Cell Res 2020; 391:112030. [PMID: 32330509 DOI: 10.1016/j.yexcr.2020.112030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/17/2020] [Accepted: 04/19/2020] [Indexed: 12/22/2022]
Abstract
Re-epithelialization of the alveolar surface is a key process of lung alveolar epithelial barrier repair after acute lung injury. The receptor for advanced glycation end-products (RAGE) pathway plays key roles in lung homeostasis, and its involvement in wound repair has been already reported in human bronchial epithelial cells. However, its effects on lung alveolar epithelial repair after injury remain unknown. We investigated whether RAGE stimulation with its ligands high-mobility group box 1 protein (HMGB1) or advanced glycation end-products (AGEs), alone or associated with RAGE inhibition using RAGE antagonist peptide, affects in vitro wound healing in human alveolar epithelial A549 cells. We further asked whether these effects could be associated with changes in cell proliferation and migration. We found that treatment of A549 cells with HMGB1 or AGEs promotes RAGE-dependent wound healing after a scratch assay. In addition, both RAGE ligands increased cell proliferation in a RAGE-dependent manner. Treatment with HMGB1 increased migration of alveolar epithelial cells at 12 h, independently of RAGE, whereas AGEs stimulated migration as measured 48 h after injury in a RAGE-dependent manner. Taken together, these results suggest that RAGE pathway is involved in lung alveolar epithelial wound repair, possibly through enhanced cell migration and proliferation.
Collapse
Affiliation(s)
- Ruoyang Zhai
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France
| | - Raiko Blondonnet
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France; Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Ebrahim Ebrahimi
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France; Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Corinne Belville
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France
| | - Jules Audard
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France; Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Christelle Gross
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France
| | - Helena Choltus
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France
| | - Fanny Henrioux
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France
| | - Jean-Michel Constantin
- Sorbonne University, GRC 29, AP-HP, DMU DREAM, Department of Anesthesiology and Critical Care, Pitié-Salpêtrière Hospital, Paris, France
| | - Bruno Pereira
- Biostatistics Unit, Department of Clinical Research and Innovation (DRCI), CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Loic Blanchon
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France
| | - Vincent Sapin
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France; Department of Medical Biochemistry and Molecular Genetics, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Matthieu Jabaudon
- Université Clermont Auvergne, CNRS, INSERM, GReD, Clermont-Ferrand, France; Department of Perioperative Medicine, CHU Clermont-Ferrand, Clermont-Ferrand, France; Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
37
|
Decreased Soluble Receptor of Advanced Glycation End Product Levels Correlated with Inflammation in Silicosis. Mediators Inflamm 2020; 2020:2683753. [PMID: 32351319 PMCID: PMC7178542 DOI: 10.1155/2020/2683753] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/07/2020] [Accepted: 03/30/2020] [Indexed: 12/12/2022] Open
Abstract
Silicosis is a devastating disease caused by inhalation of silica dust that leads to inflammatory cascade and then scarring of the lung tissue. Increasing evidences indicate that soluble receptor for advanced glycation end products (sRAGE) is involved in inflammatory diseases. However, no data on the possible relationship between sRAGE and inflammation of silicosis are available. In this study, serum from subjects with silicosis (n = 59) or from healthy controls (HC, n = 14) was analyzed for the secretion of sRAGE, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), transforming growth factor-β1 (TGF-β1), and oxidized low-density lipoprotein (ox-LDL). The associations between sRAGE and cytokines and ox-LDL and lung function were assessed by Pearson's correlation analyses. Mean levels of serum sRAGE were lower in silicosis than those in controls (p < 0.05). The subjects who had a longer term of occupational exposure had higher levels of sRAGE (p < 0.05). The secretion of TNF-α, IL-1β, IL-6, TGF-β1, and ox-LDL was significantly higher in the silicosis group than that in the HC group (p < 0.05). Furthermore, the levels of sRAGE were negatively correlated with TNF-α, IL-6, IL-1β, and ox-LDL. There is no correlation between sRAGE and TGF-β1 and lung function. The optimal point of sRAGE for differentiating silicosis from healthy controls was 14250.02 pg/ml by ROC curve analysis. A decrease in serum sRAGE and its association with inflammatory response might suggest a role for sRAGE in the pathogenesis of silicosis.
Collapse
|
38
|
Yang P, Esper AM, Martin GS. The Future of ARDS Biomarkers: Where Are the Gaps in Implementation of Precision Medicine? ANNUAL UPDATE IN INTENSIVE CARE AND EMERGENCY MEDICINE 2020. [DOI: 10.1007/978-3-030-37323-8_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
39
|
Lynn H, Sun X, Casanova N, Gonzales-Garay M, Bime C, Garcia JGN. Genomic and Genetic Approaches to Deciphering Acute Respiratory Distress Syndrome Risk and Mortality. Antioxid Redox Signal 2019; 31:1027-1052. [PMID: 31016989 PMCID: PMC6939590 DOI: 10.1089/ars.2018.7701] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significance: Acute respiratory distress syndrome (ARDS) is a severe, highly heterogeneous critical illness with staggering mortality that is influenced by environmental factors, such as mechanical ventilation, and genetic factors. Significant unmet needs in ARDS are addressing the paucity of validated predictive biomarkers for ARDS risk and susceptibility that hamper the conduct of successful clinical trials in ARDS and the complete absence of novel disease-modifying therapeutic strategies. Recent Advances: The current ARDS definition relies on clinical characteristics that fail to capture the diversity of disease pathology, severity, and mortality risk. We undertook a comprehensive survey of the available ARDS literature to identify genes and genetic variants (candidate gene and limited genome-wide association study approaches) implicated in susceptibility to developing ARDS in hopes of uncovering novel biomarkers for ARDS risk and mortality and potentially novel therapeutic targets in ARDS. We further attempted to address the well-known health disparities that exist in susceptibility to and mortality from ARDS. Critical Issues: Bioinformatic analyses identified 201 ARDS candidate genes with pathway analysis indicating a strong predominance in key evolutionarily conserved inflammatory pathways, including reactive oxygen species, innate immunity-related inflammation, and endothelial vascular signaling pathways. Future Directions: Future studies employing a system biology approach that combines clinical characteristics, genomics, transcriptomics, and proteomics may allow for a better definition of biologically relevant pathways and genotype-phenotype connections and result in improved strategies for the sub-phenotyping of diverse ARDS patients via molecular signatures. These efforts should facilitate the potential for successful clinical trials in ARDS and yield a better fundamental understanding of ARDS pathobiology.
Collapse
Affiliation(s)
- Heather Lynn
- Department of Physiological Sciences and University of Arizona, Tucson, Arizona.,Department of Health Sciences, University of Arizona, Tucson, Arizona
| | - Xiaoguang Sun
- Department of Health Sciences, University of Arizona, Tucson, Arizona
| | - Nancy Casanova
- Department of Health Sciences, University of Arizona, Tucson, Arizona
| | | | - Christian Bime
- Department of Health Sciences, University of Arizona, Tucson, Arizona
| | - Joe G N Garcia
- Department of Health Sciences, University of Arizona, Tucson, Arizona
| |
Collapse
|
40
|
Encou-RAGE-ing Lung Protection in Patients With Acute Respiratory Distress Syndrome Under Extracorporeal Membrane Oxygenation*. Crit Care Med 2019; 47:1654-1655. [DOI: 10.1097/ccm.0000000000003984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
41
|
Ultra-Protective Ventilation Reduces Biotrauma in Patients on Venovenous Extracorporeal Membrane Oxygenation for Severe Acute Respiratory Distress Syndrome*. Crit Care Med 2019; 47:1505-1512. [DOI: 10.1097/ccm.0000000000003894] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
42
|
Constantin JM, Jabaudon M, Lefrant JY, Jaber S, Quenot JP, Langeron O, Ferrandière M, Grelon F, Seguin P, Ichai C, Veber B, Souweine B, Uberti T, Lasocki S, Legay F, Leone M, Eisenmann N, Dahyot-Fizelier C, Dupont H, Asehnoune K, Sossou A, Chanques G, Muller L, Bazin JE, Monsel A, Borao L, Garcier JM, Rouby JJ, Pereira B, Futier E, Sophie C, Thomas G, Renaud G, Camille V, Russel C, Bernard C, Raiko B, Alexandre L, Nathanael E, Laurent M, Pablo M, Caroline B, Saber B, Claire R, Fouad B, Moussa C, Marion M, Matthieu C, Julie C, Audrey DJ, Auguste D, Pascal A, Thomas L, Yoann L, Antoine R, Raphael C, Caroline B, Anne-Charlotte T, Mathilde B, Benjamin C, Edouard L, Pierre-Marie B, Charlotte A, Laurent Z, Emmanuelle H, Garry D, Calypso M, Herve D, Benoit V, Jean-Christophe O, Hervé Q, Thomas R, Julien CC, Marinne LC, Fabien G, Mona A, Frank P, Jerome M, Serge M, Nanadougmar H. Personalised mechanical ventilation tailored to lung morphology versus low positive end-expiratory pressure for patients with acute respiratory distress syndrome in France (the LIVE study): a multicentre, single-blind, randomised controlled trial. THE LANCET RESPIRATORY MEDICINE 2019; 7:870-880. [DOI: 10.1016/s2213-2600(19)30138-9] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/08/2019] [Accepted: 03/11/2019] [Indexed: 12/29/2022]
|
43
|
Yehya N. Lessons learned in acute respiratory distress syndrome from the animal laboratory. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:503. [PMID: 31728356 DOI: 10.21037/atm.2019.09.33] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Since the description of the acute respiratory distress syndrome (ARDS) in 1967, investigators have struggled to reproduce the syndrome in the animal laboratory. While several different models of experimental acute lung injury (ALI) have been developed, none completely capture the inciting etiologies, initial inflammation, heterogeneity, and resolution of human ARDS. This potentially has contributed to the poor translation of potential therapeutics between animal ALI models and human ARDS. It was only recently that standardized criteria were suggested for what makes an ALI model comparable to human ARDS. Nevertheless, despite model heterogeneity, these models have contributed substantially to our understanding of the syndrome. From the initial studies identifying the risks of mechanical ventilation to the identification of potentially targetable inflammatory mediators, to modern studies focusing on regional heterogeneity and novel molecular pathways, animal models continue to inform our understanding of ARDS. This review will cover several major lessons learned from animal models of ALI, and provide some direction for future studies in this field.
Collapse
Affiliation(s)
- Nadir Yehya
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
44
|
Inhibition of the Receptor for Advanced Glycation End-Products in Acute Respiratory Distress Syndrome: A Randomised Laboratory Trial in Piglets. Sci Rep 2019; 9:9227. [PMID: 31239497 PMCID: PMC6592897 DOI: 10.1038/s41598-019-45798-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 06/12/2019] [Indexed: 02/08/2023] Open
Abstract
The receptor for advanced glycation end-products (RAGE) modulates the pathogenesis of acute respiratory distress syndrome (ARDS). RAGE inhibition attenuated lung injury and restored alveolar fluid clearance (AFC) in a mouse model of ARDS. However, clinical translation will require assessment of this strategy in larger animals. Forty-eight anaesthetised Landrace piglets were randomised into a control group and three treatment groups. Animals allocated to treatment groups underwent orotracheal instillation of hydrochloric acid (i) alone; (ii) in combination with intravenous administration of a RAGE antagonist peptide (RAP), or (iii) recombinant soluble (s)RAGE. The primary outcome was net AFC at 4 h. Arterial oxygenation was assessed hourly and alveolar-capillary permeability, alveolar inflammation and lung histology were assessed at 4 h. Treatment with either RAP or sRAGE improved net AFC (median [interquartile range], 21.2 [18.8–21.7] and 19.5 [17.1–21.5] %/h, respectively, versus 12.6 [3.2–18.8] %/h in injured, untreated controls), oxygenation and decreased alveolar inflammation and histological evidence of tissue injury after ARDS. These findings suggest that RAGE inhibition restored AFC and attenuated lung injury in a piglet model of acid-induced ARDS.
Collapse
|
45
|
Ramin S, Charbit J, Jaber S, Capdevila X. Acute respiratory distress syndrome after chest trauma: Epidemiology, specific physiopathology and ventilation strategies. Anaesth Crit Care Pain Med 2019; 38:265-276. [DOI: 10.1016/j.accpm.2018.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/05/2018] [Accepted: 09/06/2018] [Indexed: 01/07/2023]
|
46
|
Sklar MC, Patel BK, Beitler JR, Piraino T, Goligher EC. Optimal Ventilator Strategies in Acute Respiratory Distress Syndrome. Semin Respir Crit Care Med 2019; 40:81-93. [PMID: 31060090 PMCID: PMC7117088 DOI: 10.1055/s-0039-1683896] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mechanical ventilation practices in patients with acute respiratory distress syndrome (ARDS) have progressed with a growing understanding of the disease pathophysiology. Paramount to the care of affected patients is the delivery of lung-protective mechanical ventilation which prioritizes tidal volume and plateau pressure limitation. Lung protection can probably be further enhanced by scaling target tidal volumes to the specific respiratory mechanics of individual patients. The best procedure for selecting optimal positive end-expiratory pressure (PEEP) in ARDS remains uncertain; several relevant issues must be considered when selecting PEEP, particularly lung recruitability. Noninvasive ventilation must be used with caution in ARDS as excessively high respiratory drive can further exacerbate lung injury; newer modes of delivery offer promising approaches in hypoxemic respiratory failure. Airway pressure release ventilation offers an alternative approach to maximize lung recruitment and oxygenation, but clinical trials have not demonstrated a survival benefit of this mode over conventional ventilation strategies. Rescue therapy with high-frequency oscillatory ventilation is an important option in refractory hypoxemia. Despite a disappointing lack of benefit (and possible harm) in patients with moderate or severe ARDS, possibly due to lung hyperdistention and right ventricular dysfunction, high-frequency oscillation may improve outcome in patients with very severe hypoxemia.
Collapse
Affiliation(s)
- Michael C Sklar
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Bhakti K Patel
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Jeremy R Beitler
- Center for Acute Respiratory Failure and Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University, New York, New York
| | - Thomas Piraino
- Keenan Centre for Biomedical Research, St. Michael's Hospital, Toronto, Ontario, Canada.,Division of Critical Care, Department of Anesthesia, McMaster University, Hamilton, Ontario, Canada.,Department of Respiratory Therapy, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Ewan C Goligher
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada.,Toronto General Hospital Research Institute, Toronto, Ontario, Canada.,Department of Medicine, Division of Respirology, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
47
|
Egron C, Roszyk L, Rochette E, Jabaudon M, Sapin V, Mulliez A, Labbé A, Coste K. Serum soluble receptor for advanced glycation end-products during acute bronchiolitis in infant: Prospective study in 93 cases. Pediatr Pulmonol 2018; 53:1429-1435. [PMID: 30113140 PMCID: PMC7167909 DOI: 10.1002/ppul.24141] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 07/25/2018] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Acute bronchiolitis is a major cause of acute respiratory distress in infants. The soluble receptor for advanced glycation end-products (sRAGE) is a biomarker of pulmonary damage processes, with a diagnostic and a prognostic value in acute respiratory distress syndrome (ARDS). The RAGE pathway is also implicated in the pathogenesis of other respiratory diseases like asthma, but the value of sRAGE levels in acute bronchiolitis remains under-investigated. MATERIAL AND METHODS A prospective, observational, and analytical study was conducted at Clermont-Ferrand University Hospital. The main objective was to evaluate the correlation between serum sRAGE and clinical severity of bronchiolitis in hospitalized infants aged <1 year. We analyzed correlations between serum sRAGE and Wainwright score, short-term morbidity attributable to bronchiolitis, causal viruses and risk for recurrent wheezing at 1 year. RESULTS The study included 93 infants. sRAGE levels were significantly lower in acute bronchiolitis patients (mean 1101 pg/mL) than in controls (2203 pg/mL, P < 0.001) but did not correlate with clinical severity. No correlation was found between serum sRAGE and severity score, respiratory viruses, and recurrent wheezing at 1 year. Serum sRAGE levels were negatively correlated with age (r = -0.45, P < 0.001). CONCLUSION Serum sRAGE levels are decreased in acute bronchiolitis but not correlated with disease severity. sRAGE levels should be age-adjusted in infants. Serum sRAGE levels measured in the setting of acute bronchiolitis were not predictive of recurrent wheezing.
Collapse
Affiliation(s)
- Carole Egron
- Department of Pediatrics, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Laurence Roszyk
- Department of Medical Biochemistry and Molecular Biology, CHU Clermont-Ferrand and GReD, Université Clermont Auvergne, CNRS UMR 6293, INSERM U1103, Clermont-Ferrand, France
| | - Emmanuelle Rochette
- Department of Clinical Research for Children (CRECHE) at CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Matthieu Jabaudon
- Department of Perioperative Medicine, CHU Clermont-Ferrand and GReD, Université Clermont Auvergne, CNRS UMR 6293, INSERM U1103, Clermont-Ferrand, France
| | - Vincent Sapin
- Department of Medical Biochemistry and Molecular Biology, CHU Clermont-Ferrand and GReD, Université Clermont Auvergne, CNRS UMR 6293, INSERM U1103, Clermont-Ferrand, France
| | - Aurélien Mulliez
- Department of Clinical Research and Innovation at CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - André Labbé
- Department of Pediatrics, CHU Clermont-Ferrand, Clermont-Ferrand and Université Clermont Auvergne, Clermont-Ferrand, France
| | - Karen Coste
- Department of Pediatrics, CHU Clermont-Ferrand and GReD, Université Clermont Auvergne, CNRS UMR 6293, INSERM U1103, Clermont-Ferrand, France
| |
Collapse
|
48
|
Jabaudon M, Blondonnet R, Pereira B, Cartin-Ceba R, Lichtenstern C, Mauri T, Determann RM, Drabek T, Hubmayr RD, Gajic O, Uhle F, Coppadoro A, Pesenti A, Schultz MJ, Ranieri MV, Brodska H, Mrozek S, Sapin V, Matthay MA, Constantin JM, Calfee CS. Plasma sRAGE is independently associated with increased mortality in ARDS: a meta-analysis of individual patient data. Intensive Care Med 2018; 44:1388-1399. [PMID: 30051136 PMCID: PMC6132684 DOI: 10.1007/s00134-018-5327-1] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 07/17/2018] [Indexed: 12/20/2022]
Abstract
PURPOSE The soluble receptor for advanced glycation end-products (sRAGE) is a marker of lung epithelial injury and alveolar fluid clearance (AFC), with promising values for assessing prognosis and lung injury severity in acute respiratory distress syndrome (ARDS). Because AFC is impaired in most patients with ARDS and is associated with higher mortality, we hypothesized that baseline plasma sRAGE would predict mortality, independently of two key mediators of ventilator-induced lung injury. METHODS We conducted a meta-analysis of individual data from 746 patients enrolled in eight prospective randomized and observational studies in which plasma sRAGE was measured in ARDS articles published through March 2016. The primary outcome was 90-day mortality. Using multivariate and mediation analyses, we tested the association between baseline plasma sRAGE and mortality, independently of driving pressure and tidal volume. RESULTS Higher baseline plasma sRAGE [odds ratio (OR) for each one-log increment, 1.18; 95% confidence interval (CI) 1.01-1.38; P = 0.04], driving pressure (OR for each one-point increment, 1.04; 95% CI 1.02-1.07; P = 0.002), and tidal volume (OR for each one-log increment, 1.98; 95% CI 1.07-3.64; P = 0.03) were independently associated with higher 90-day mortality in multivariate analysis. Baseline plasma sRAGE mediated a small fraction of the effect of higher ΔP on mortality but not that of higher VT. CONCLUSIONS Higher baseline plasma sRAGE was associated with higher 90-day mortality in patients with ARDS, independently of driving pressure and tidal volume, thus reinforcing the likely contribution of alveolar epithelial injury as an important prognostic factor in ARDS. Registration: PROSPERO (ID: CRD42018100241).
Collapse
Affiliation(s)
- Matthieu Jabaudon
- Department of Perioperative Medicine, CHU Clermont-Ferrand, 1 Place Lucie Aubrac, 63003, Clermont-Ferrand Cedex 1, France.
- CNRS, UMR 6293, INSERM U1103, GReD, Université Clermont Auvergne, Clermont-Ferrand, France.
| | - Raiko Blondonnet
- Department of Perioperative Medicine, CHU Clermont-Ferrand, 1 Place Lucie Aubrac, 63003, Clermont-Ferrand Cedex 1, France
- CNRS, UMR 6293, INSERM U1103, GReD, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Bruno Pereira
- Biostatistics Unit, Department of Clinical Research and Innovation (DRCI), CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Rodrigo Cartin-Ceba
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Tommaso Mauri
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | | | - Tomas Drabek
- Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rolf D Hubmayr
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ognjen Gajic
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | - Florian Uhle
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Andrea Coppadoro
- Department of Emergency Medicine, San Gerardo Hospital, Monza, Italy
| | - Antonio Pesenti
- Department of Anesthesia, Critical Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Marcus J Schultz
- Department of Intensive Care Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Mahidol-Oxford Tropical Medicine Research Unit (MORU), Bangkok, Thailand
| | - Marco V Ranieri
- Department of Surgical Sciences, Molinette Hospital, City of Health and Science, University of Turin, Turin, Italy
| | - Helena Brodska
- Institute of Clinical Biochemistry and Laboratory Diagnostics, Faculty of Medicine, General University Hospital, Charles University, Prague, Czech Republic
| | - Ségolène Mrozek
- Department of Anesthesia and Intensive Care, University Hospital of Toulouse, University Toulouse 3 Paul Sabatier, Toulouse, France
| | - Vincent Sapin
- CNRS, UMR 6293, INSERM U1103, GReD, Université Clermont Auvergne, Clermont-Ferrand, France
- Department of Medical Biochemistry and Molecular Biology, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Michael A Matthay
- Division of Pulmonary and Critical Care Medicine, Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Jean-Michel Constantin
- Department of Perioperative Medicine, CHU Clermont-Ferrand, 1 Place Lucie Aubrac, 63003, Clermont-Ferrand Cedex 1, France
- CNRS, UMR 6293, INSERM U1103, GReD, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Carolyn S Calfee
- Division of Pulmonary and Critical Care Medicine, Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
49
|
Clinical and Biological Predictors of Plasma Levels of Soluble RAGE in Critically Ill Patients: Secondary Analysis of a Prospective Multicenter Observational Study. DISEASE MARKERS 2018; 2018:7849675. [PMID: 29861796 PMCID: PMC5971347 DOI: 10.1155/2018/7849675] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 04/11/2018] [Indexed: 02/07/2023]
Abstract
Rationale Although soluble forms of the receptor for advanced glycation end products (RAGE) have been recently proposed as biomarkers in multiple acute or chronic diseases, few studies evaluated the influence of usual clinical and biological parameters, or of patient characteristics and comorbidities, on circulating levels of soluble RAGE in the intensive care unit (ICU) setting. Objectives To determine, among clinical and biological parameters that are usually recorded upon ICU admission, which variables, if any, could be associated with plasma levels of soluble RAGE. Methods Data for this ancillary study were prospectively obtained from adult patients with at least one ARDS risk factor upon ICU admission enrolled in a large multicenter observational study. At ICU admission, plasma levels of total soluble RAGE (sRAGE) and endogenous secretory (es)RAGE were measured by duplicate ELISA and baseline patient characteristics, comorbidities, and usual clinical and biological indices were recorded. After univariate analyses, significant variables were used in multivariate, multidimensional analyses. Measurements and Main Results 294 patients were included in this ancillary study, among whom 62% were admitted for medical reasons, including septic shock (11%), coma (11%), and pneumonia (6%). Although some variables were associated with plasma levels of RAGE soluble forms in univariate analysis, multidimensional analyses showed no significant association between admission parameters and baseline plasma sRAGE or esRAGE. Conclusions We found no obvious association between circulating levels of soluble RAGE and clinical and biological indices that are usually recorded upon ICU admission. This trial is registered with NCT02070536.
Collapse
|
50
|
Riché A, Adam N, Monsel A, Xia J, Langeron O, Rouby JJ. Sevoflurane in Acute Respiratory Distress Syndrome: Are Lung Protection and Anesthesia Depth Influenced by Pulmonary Morphology? Am J Respir Crit Care Med 2018; 197:830-832. [DOI: 10.1164/rccm.201709-1864le] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
| | | | | | - Jing Xia
- First Affiliated Hospital of Kunming Medical UniversityKunming, China
| | | | | |
Collapse
|