51
|
Bonnesen B, Jensen JUS, Jeschke KN, Mathioudakis AG, Corlateanu A, Hansen EF, Weinreich UM, Hilberg O, Sivapalan P. Management of COVID-19-Associated Acute Respiratory Failure with Alternatives to Invasive Mechanical Ventilation: High-Flow Oxygen, Continuous Positive Airway Pressure, and Noninvasive Ventilation. Diagnostics (Basel) 2021. [DOI: doi.org/10.3390/diagnostics11122259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Patients admitted to hospital with coronavirus disease 2019 (COVID-19) may develop acute respiratory failure (ARF) with compromised gas exchange. These patients require oxygen and possibly ventilatory support, which can be delivered via different devices. Initially, oxygen therapy will often be administered through a conventional binasal oxygen catheter or air-entrainment mask. However, when higher rates of oxygen flow are needed, patients are often stepped up to high-flow nasal cannula oxygen therapy (HFNC), continuous positive airway pressure (CPAP), bilevel positive airway pressure (BiPAP), or invasive mechanical ventilation (IMV). BiPAP, CPAP, and HFNC may be beneficial alternatives to IMV for COVID-19-associated ARF. Current evidence suggests that when nasal catheter oxygen therapy is insufficient for adequate oxygenation of patients with COVID-19-associated ARF, CPAP should be provided for prolonged periods. Subsequent escalation to IMV may be implemented if necessary.
Collapse
|
52
|
Bonnesen B, Jensen JUS, Jeschke KN, Mathioudakis AG, Corlateanu A, Hansen EF, Weinreich UM, Hilberg O, Sivapalan P. Management of COVID-19-Associated Acute Respiratory Failure with Alternatives to Invasive Mechanical Ventilation: High-Flow Oxygen, Continuous Positive Airway Pressure, and Noninvasive Ventilation. Diagnostics (Basel) 2021; 11:2259. [PMID: 34943496 PMCID: PMC8700515 DOI: 10.3390/diagnostics11122259] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/28/2021] [Accepted: 12/01/2021] [Indexed: 02/05/2023] Open
Abstract
Patients admitted to hospital with coronavirus disease 2019 (COVID-19) may develop acute respiratory failure (ARF) with compromised gas exchange. These patients require oxygen and possibly ventilatory support, which can be delivered via different devices. Initially, oxygen therapy will often be administered through a conventional binasal oxygen catheter or air-entrainment mask. However, when higher rates of oxygen flow are needed, patients are often stepped up to high-flow nasal cannula oxygen therapy (HFNC), continuous positive airway pressure (CPAP), bilevel positive airway pressure (BiPAP), or invasive mechanical ventilation (IMV). BiPAP, CPAP, and HFNC may be beneficial alternatives to IMV for COVID-19-associated ARF. Current evidence suggests that when nasal catheter oxygen therapy is insufficient for adequate oxygenation of patients with COVID-19-associated ARF, CPAP should be provided for prolonged periods. Subsequent escalation to IMV may be implemented if necessary.
Collapse
Affiliation(s)
- Barbara Bonnesen
- Department of Medicine, Section of Respiratory Medicine, Herlev and Gentofte Hospital, University of Copenhagen, 2200 Copenhagen, Denmark; (B.B.); (J.-U.S.J.)
| | - Jens-Ulrik Stæhr Jensen
- Department of Medicine, Section of Respiratory Medicine, Herlev and Gentofte Hospital, University of Copenhagen, 2200 Copenhagen, Denmark; (B.B.); (J.-U.S.J.)
- Department of Clinical Medicine, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Klaus Nielsen Jeschke
- Department of Respiratory Medicine, Copenhagen University Hospital-Hvidovre, 2650 Hvidovre, Denmark; (K.N.J.); (E.F.H.)
| | - Alexander G. Mathioudakis
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester Academic Health Science Centre, Manchester M23 9LT, UK;
- North West Lung Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester M23 9LT, UK
| | - Alexandru Corlateanu
- Department of Respiratory Medicine, State University of Medicine and Pharmacy “Nicolae Testemitanu”, 2004 Chisinau, Moldova;
| | - Ejvind Frausing Hansen
- Department of Respiratory Medicine, Copenhagen University Hospital-Hvidovre, 2650 Hvidovre, Denmark; (K.N.J.); (E.F.H.)
| | - Ulla Møller Weinreich
- Department of Respiratory Medicine, Aalborg University Hospital, University of Aalborg, 9100 Aalborg, Denmark;
- The Clinical Institute, Aalborg University, 9220 Aalborg, Denmark
| | - Ole Hilberg
- Department of Medicine, Little Belt Hospital, 7100 Vejle, Denmark;
- Department of Regional Health Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Pradeesh Sivapalan
- Department of Medicine, Section of Respiratory Medicine, Herlev and Gentofte Hospital, University of Copenhagen, 2200 Copenhagen, Denmark; (B.B.); (J.-U.S.J.)
| |
Collapse
|
53
|
Juliana A, Plötz FB, Achten N, Bultman A, Jongman RM, van Meurs M, Wilschut JC, Zonneveld R. Requirement of respiratory support in acute bronchiolitis in infants is linked to endothelial and neutrophil activation. Pediatr Pulmonol 2021; 56:3908-3915. [PMID: 34491635 DOI: 10.1002/ppul.25663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 08/03/2021] [Accepted: 08/19/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND Evidence shows that activation of pulmonary vascular endothelium and neutrophils are involved in the pathophysiology of acute bronchiolitis. We hypothesized that levels of markers of endothelial activation and leukocyte counts are associated with requirement and duration of respiratory support. METHODS Thirty-four infants with bronchiolitis and eight controls were included. Nasopharyngeal swabs and blood samples were taken at admission. Serum levels of Angiopoietin (Ang)-1, Ang-2, sP-selectin, sE-selectin, vascular cell adhesion molecule-1 (sVCAM-1), intercellular adhesion molecule-1 (sICAM-1), and leukocyte counts were measured. For univariate analysis, bronchiolitis cases were grouped into two groups, namely those not requiring and those requiring any form of respiratory support. To control for known risk factors for poor outcome (i.e., age, prematurity, and congenital heart disease), and for days post symptom onset, linear regression analysis was performed with duration of any type of respiratory support in days. RESULTS Ang-2 levels, Ang-2/Ang-1 ratios, sE-selectin levels, immature neutrophil count, and neutrophil/lymphocyte ratio (NLR) were higher in acute bronchiolitis versus controls. Ang-2, and NLR levels were significantly higher, and lymphocyte counts significantly lower, in infants that required respiratory support versus those that did not. Ang-2 levels (β: .32, 95% confidence interval [CI]: 0.19-1.19) and NLR (β: .68, 95% CI: 0.17-1.19) were positive predictors for the duration of respiratory support. CONCLUSIONS Markers of endothelial and neutrophil activation are associated with respiratory support for acute bronchiolitis. Admission Ang-2 levels and NLR may be promising markers to determine requirement of respiratory support and deserve further study.
Collapse
Affiliation(s)
- Amadu Juliana
- Academic Pediatric Center Suriname, Academic Hospital Paramaribo, Paramaribo, Suriname
| | - Frans B Plötz
- Department of Pediatrics, Tergooi Hospitals, Hilversum, The Netherlands.,Emma Children's Hospital, Amsterdam Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Niek Achten
- Academic Pediatric Center Suriname, Academic Hospital Paramaribo, Paramaribo, Suriname
| | - Anita Bultman
- Academic Pediatric Center Suriname, Academic Hospital Paramaribo, Paramaribo, Suriname
| | - Rianne M Jongman
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Anesthesiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Matijs van Meurs
- Department of Anesthesiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Critical Care, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan C Wilschut
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Rens Zonneveld
- Academic Pediatric Center Suriname, Academic Hospital Paramaribo, Paramaribo, Suriname
| |
Collapse
|
54
|
Gotts JE, Maishan M, Chun L, Fang X, Han C, Chiueh V, Khakoo AY, Lee T, Stolina M, Matthay MA. Delayed angiopoietin-2 blockade reduces influenza-induced lung injury and improves survival in mice. Physiol Rep 2021; 9:e15081. [PMID: 34755490 PMCID: PMC8578883 DOI: 10.14814/phy2.15081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/21/2021] [Accepted: 09/25/2021] [Indexed: 11/24/2022] Open
Abstract
Influenza remains a major cause of death and disability with limited treatment options. Studies of acute lung injury have identified angiopoietin-2 (Ang-2) as a key prognostic marker and a potential mediator of Acute respiratory distress syndrome. However, the role of Ang-2 in viral pneumonia remains poorly defined. This study characterized the time course of lung Ang-2 expression in severe influenza pneumonia and tested the therapeutic potential of Ang-2 inhibition. We inoculated adult mice with influenza A (PR8 strain) and measured angiopoietin-1 (Ang-1), Ang-2, and Tie2 expressions during the evolution of inflammatory lung injury over the first 7 days post-infection (dpi). We tested a peptide-antibody inhibitor of Ang-2, L1-7, administered at 2, 4, and 6 dpi and measured arterial oxygen saturation, survival, pulmonary edema, inflammatory cytokines, and viral load. Finally, we infected primary human alveolar type II epithelial (AT2) cells grown in air-liquid interface culture with influenza and measured Ang-2 RNA expression. Influenza caused severe lung injury between 5 and 7 dpi in association with increased Ang-2 lung RNA and a dramatic increase in Ang-2 protein in bronchoalveolar lavage. Inhibition of Ang-2 improved oxygenation and survival and reduced pulmonary edema and alveolar-capillary barrier permeability to protein without major effects on inflammation or viral load. Finally, influenza increased the expression of Ang-2 RNA in human AT2 cells. The increased Ang-2 levels in the airspaces during severe influenza pneumonia and the improvement in clinically relevant outcomes after Ang-2 antagonism suggest that the Ang-1/Ang-2 Tie-2 signaling axis is a promising therapeutic target in influenza and potentially other causes of viral pneumonia.
Collapse
Affiliation(s)
- Jeffrey E. Gotts
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Mazharul Maishan
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Lauren Chun
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Xiaohui Fang
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Chun‐Ya Han
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - Venice Chiueh
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - Aarif Y. Khakoo
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - TaeWeon Lee
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - Marina Stolina
- Department of Cardiometabolic DisordersAmgen ResearchThousand OaksCaliforniaUSA
| | - Michael A. Matthay
- Departments of Medicine and AnesthesiaCardiovascular Research InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
55
|
Felsenstein S, Reiff AO. A hitchhiker's guide through the COVID-19 galaxy. Clin Immunol 2021; 232:108849. [PMID: 34563684 PMCID: PMC8461017 DOI: 10.1016/j.clim.2021.108849] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 09/04/2021] [Indexed: 01/08/2023]
Abstract
Numerous reviews have summarized the epidemiology, pathophysiology and the various therapeutic aspects of Coronavirus disease 2019 (COVID-19), but a practical guide on "how to treat whom with what and when" based on an understanding of the immunological background of the disease stages remains missing. This review attempts to combine the current knowledge about the immunopathology of COVID-19 with published evidence of available and emerging treatment options. We recognize that the information about COVID-19 and its treatment is rapidly changing, but hope that this guide offers those on the frontline of this pandemic an understanding of the host response in COVID-19 patients and supports their ongoing efforts to select the best treatments tailored to their patient's clinical status.
Collapse
Affiliation(s)
- Susanna Felsenstein
- University of Liverpool, Faculty of Health and Life Sciences, Brownlow Hill, Liverpool, L69 3GB, United Kingdom.
| | - Andreas Otto Reiff
- Arthritis & Rheumatic Diseases, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States.
| |
Collapse
|
56
|
Identification of early and intermediate biomarkers for ARDS mortality by multi-omic approaches. Sci Rep 2021; 11:18874. [PMID: 34556700 PMCID: PMC8460799 DOI: 10.1038/s41598-021-98053-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 08/26/2021] [Indexed: 12/29/2022] Open
Abstract
The lack of successful clinical trials in acute respiratory distress syndrome (ARDS) has highlighted the unmet need for biomarkers predicting ARDS mortality and for novel therapeutics to reduce ARDS mortality. We utilized a systems biology multi-“omics” approach to identify predictive biomarkers for ARDS mortality. Integrating analyses were designed to differentiate ARDS non-survivors and survivors (568 subjects, 27% overall 28-day mortality) using datasets derived from multiple ‘omics’ studies in a multi-institution ARDS cohort (54% European descent, 40% African descent). ‘Omics’ data was available for each subject and included genome-wide association studies (GWAS, n = 297), RNA sequencing (n = 93), DNA methylation data (n = 61), and selective proteomic network analysis (n = 240). Integration of available “omic” data identified a 9-gene set (TNPO1, NUP214, HDAC1, HNRNPA1, GATAD2A, FOSB, DDX17, PHF20, CREBBP) that differentiated ARDS survivors/non-survivors, results that were validated utilizing a longitudinal transcription dataset. Pathway analysis identified TP53-, HDAC1-, TGF-β-, and IL-6-signaling pathways to be associated with ARDS mortality. Predictive biomarker discovery identified transcription levels of the 9-gene set (AUC-0.83) and Day 7 angiopoietin 2 protein levels as potential candidate predictors of ARDS mortality (AUC-0.70). These results underscore the value of utilizing integrated “multi-omics” approaches in underpowered datasets from racially diverse ARDS subjects.
Collapse
|
57
|
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: 23] [Impact Index Per Article: 5.8] [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
|
58
|
Dutra Silva J, Su Y, Calfee CS, Delucchi KL, Weiss D, McAuley DF, O'Kane C, Krasnodembskaya AD. Mesenchymal stromal cell extracellular vesicles rescue mitochondrial dysfunction and improve barrier integrity in clinically relevant models of ARDS. Eur Respir J 2021; 58:13993003.02978-2020. [PMID: 33334945 PMCID: PMC8318599 DOI: 10.1183/13993003.02978-2020] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 12/02/2020] [Indexed: 12/22/2022]
Abstract
Alveolar epithelial–capillary barrier disruption is a hallmark of acute respiratory distress syndrome (ARDS). Contribution of mitochondrial dysfunction to the compromised alveolar-capillary barrier in ARDS remains unclear. Mesenchymal stromal cells-derived extracellular vesicles (MSC-EVs) are considered as a cell-free therapy for ARDS. Mitochondrial transfer was shown to be important for the therapeutic effects of MSCs and MSC-EVs. Here we investigated the contribution of mitochondrial dysfunction to the injury of alveolar epithelial and endothelial barriers in ARDS and the ability of MSC-EVs to modulate alveolar–capillary barrier integrity through mitochondrial transfer. Primary human small airway epithelial and pulmonary microvascular endothelial cells and human precision cut lung slices (PCLSs) were stimulated with endotoxin or plasma samples from patients with ARDS and treated with MSC-EVs, barrier properties and mitochondrial functions were evaluated. Lipopolysaccharide (LPS)-injured mice were treated with MSC-EVs and degree of lung injury and mitochondrial respiration of the lung tissue were assessed. Inflammatory stimulation resulted in increased permeability coupled with pronounced mitochondrial dysfunction in both types of primary cells and PCLSs. Extracellular vesicles derived from normal MSCs restored barrier integrity and normal levels of oxidative phosphorylation while an extracellular vesicles preparation which did not contain mitochondria was not effective. In vivo, presence of mitochondria was critical for extracellular vesicles ability to reduce lung injury and restore mitochondrial respiration in the lung tissue. In the ARDS environment, MSC-EVs improve alveolar–capillary barrier properties through restoration of mitochondrial functions at least partially via mitochondrial transfer. This study demonstrates that mitochondrial dysfunction is an important mechanism of ARDS pathogenesis. Mitochondrial transfer is crucial for the ability of MSC extracellular vesicles to restore integrity of the alveolar–capillary barrier.https://bit.ly/2JuqoCY
Collapse
Affiliation(s)
- Johnatas Dutra Silva
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Yue Su
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Carolyn S Calfee
- Dept of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco, CA, USA.,Dept of Anesthesia, University of California, San Francisco, San Francisco, CA, USA.,Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Kevin L Delucchi
- Dept of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
| | - Daniel Weiss
- Dept of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Danny F McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Cecilia O'Kane
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Anna D Krasnodembskaya
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| |
Collapse
|
59
|
Wick KD, Leligdowicz A, Zhuo H, Ware LB, Matthay MA. Mesenchymal stromal cells reduce evidence of lung injury in patients with ARDS. JCI Insight 2021; 6:148983. [PMID: 33974564 PMCID: PMC8262503 DOI: 10.1172/jci.insight.148983] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/05/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Whether airspace biomarkers add value to plasma biomarkers in studying acute respiratory distress syndrome (ARDS) is not well understood. Mesenchymal stromal cells (MSCs) are an investigational therapy for ARDS, and airspace biomarkers may provide mechanistic evidence for MSCs’ impact in patients with ARDS. METHODS We carried out a nested cohort study within a phase 2a safety trial of treatment with allogeneic MSCs for moderate-to-severe ARDS. Nonbronchoscopic bronchoalveolar lavage and plasma samples were collected 48 hours after study drug infusion. Airspace and plasma biomarker concentrations were compared between the MSC (n = 17) and placebo (n = 10) treatment arms, and correlation between the two compartments was tested. Airspace biomarkers were also tested for associations with clinical and radiographic outcomes. RESULTS Compared with placebo, MSC treatment significantly reduced airspace total protein, angiopoietin-2 (Ang-2), IL-6, and soluble TNF receptor-1 concentrations. Plasma biomarkers did not differ between groups. Each 10-fold increase in airspace Ang-2 was independently associated with 6.7 fewer days alive and free of mechanical ventilation (95% CI, –12.3 to –1.0, P = 0.023), and each 10-fold increase in airspace receptor for advanced glycation end-products (RAGE) was independently associated with a 6.6-point increase in day 3 radiographic assessment of lung edema score (95% CI, 2.4 to 10.8, P = 0.004). CONCLUSION MSCs reduced biological evidence of lung injury in patients with ARDS. Biomarkers from the airspaces provide additional value for studying pathogenesis, treatment effects, and outcomes in ARDS. TRIAL REGISTRATION ClinicalTrials.gov NCT02097641. FUNDING National Heart, Lung, and Blood Institute.
Collapse
Affiliation(s)
- Katherine D Wick
- Departments of Medicine and Anesthesia and.,Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | - Aleksandra Leligdowicz
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA.,Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Hanjing Zhuo
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| | - Lorraine B Ware
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and.,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michael A Matthay
- Departments of Medicine and Anesthesia and.,Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
60
|
Sui X, Liu W, Liu Z. Exosomes derived from LPS-induced MHs cells prompted an inflammatory response in sepsis-induced acute lung injury. Respir Physiol Neurobiol 2021; 292:103711. [PMID: 34091074 DOI: 10.1016/j.resp.2021.103711] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/30/2021] [Accepted: 06/02/2021] [Indexed: 01/04/2023]
Abstract
Exosome is a novel tool with an essential role in cell communication. However, its role in the pathogenesis of sepsis-induced acute lung injury is currently unknown. Here, we first found that lipopolysaccharide (LPS) could up-regulate the expression of pro-inflammatory cytokines and promote exosomes release in the murine alveolar macrophage cell line (MHs cells). Moreover, we found MHs cells derived exosomes also maintain the pro-inflammatory effect after LPS stimulation. Treating with hydrochloride hydrate (GW4869) could dose-dependently downregulated the release of exosomes and inhibited the upregulation of inflammatory cytokines in MHs cells with LPS treatment. Also, we further identified GW4869 administration induced the remission of histopathologic changes, the reduction of pro-inflammatory cytokines in lung tissue, and inhibit serum exosomes release. These results indicate that the downregulation of exosome release by GW4869 might protect lung tissue from LPS induced injury through the suppression of excessive inflammatory responses, suggesting its potential therapeutic effects on sepsis-induced acute lung injury.
Collapse
Affiliation(s)
- Xintong Sui
- Emergency Department, First Hospital of China Medical University, Shenyang City, 110001, Liaoning Province, China.
| | - Wei Liu
- Emergency Department, First Hospital of China Medical University, Shenyang City, 110001, Liaoning Province, China.
| | - Zhi Liu
- Emergency Department, First Hospital of China Medical University, Shenyang City, 110001, Liaoning Province, China.
| |
Collapse
|
61
|
Zhao C, Mo J, Zheng X, Wu Z, Li Q, Feng J, Luo J, Lu J, Zhang J. Identification of an Alveolar Macrophage-Related Core Gene Set in Acute Respiratory Distress Syndrome. J Inflamm Res 2021; 14:2353-2361. [PMID: 34103966 PMCID: PMC8179830 DOI: 10.2147/jir.s306136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose Acute respiratory distress syndrome (ARDS) is a rapidly progressive diffuse lung injury that is characterized by high mortality and acute onset. The pathological mechanisms of ARDS are still unclear. But alveolar macrophages have been shown to play an important role in inflammatory responses during ARDS. We aimed to find the biomarkers for ARDS for early diagnosis, to give ARDS patients timely treatment. Methods Gene expression profiles were downloaded from Gene Expression Omnibus (GEO) and screened for differentially expressed genes (DEGs). The common upregulated genes in all the datasets were defined as circulating ARDS alveolar macrophage-related genes (cARDSAMGs). We performed a functional enrichment analysis to explore potential biological functions of cARDSAMGs, and we built protein–protein interaction networks. Gene set variation analysis (GSVA) was used to calculate the core gene set variation analysis (CGSVA) score for individual samples. Receiver operating characteristic (ROC) curve analysis was applied on the CGSVA score to evaluate its ability for diagnosis of ARDS. Results A total of 60 genes were upregulated in all ARDS datasets and were therefore denominated as cARDSAMGs. The cARDSAMGs were significantly involved in multiple inflammation-, immunity- and phagocytosis-related biological processes and pathways. In the protein–protein interaction network associated with host responses to ADRS, eight genes were identified as a core gene set: PTCRA, JAG1, C1QB, ADAM17, C1QA, MMP9, VSIG4 and TNFAIP3. ROC curve analysis showed that the CGSVA score may be considered as a biomarker for ARDS: it was significantly higher in patients with ARDS than those in healthy in both alveolar lavage fluid and whole blood. Conclusion The ARDS alveolar macrophage-related CGSVA score may be useful as a biomarker for ARDS.
Collapse
Affiliation(s)
- Chunling Zhao
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| | - Jingjia Mo
- Department of General Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| | - Xiaowen Zheng
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| | - Zimeng Wu
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| | - Qian Li
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| | - Jihua Feng
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| | - Jiefeng Luo
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| | - Junyu Lu
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| | - Jianfeng Zhang
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| |
Collapse
|
62
|
Edwards C. New Horizons: Does Mineralocorticoid Receptor Activation by Cortisol Cause ATP Release and COVID-19 Complications? J Clin Endocrinol Metab 2021; 106:622-635. [PMID: 33249452 PMCID: PMC7798966 DOI: 10.1210/clinem/dgaa874] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Indexed: 12/23/2022]
Abstract
This paper attempts to explain how the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus causes the complications that make coronavirus disease 2019 (COVID-19) a serious disease in specific patient subgroups. It suggests that cortisol-associated activation of the mineralocorticoid receptor (MR) in epithelial and endothelial cells infected with the virus stimulates the release of adenosine 5'-triphosphate (ATP), which then acts back on purinergic receptors. In the lung this could produce the nonproductive cough via purinergic P2X3 receptors on vagal afferent nerves. In endothelial cells it could stimulate exocytosis of Weibel-Palade bodies (WPBs) that contain angiopoietin-2, which is important in the pathogenesis of acute respiratory distress syndrome (ARDS) by increasing capillary permeability and von Willebrand factor (VWF), which mediates platelet adhesion to the endothelium and hence clotting. Angiopoietin-2 and VWF levels both are markedly elevated in COVID-19-associated ARDS. This paper offers an explanation for the sex differences in SARS-CoV-2 complications and also for why these are strongly associated with age, race, diabetes, and body mass index. It also explains why individuals with blood group A have a higher risk of severe infection than those with blood group O. Dexamethasone has been shown to be of benefit in coronavirus ARDS patients and has been thought to act as an anti-inflammatory drug. This paper suggests that a major part of its effect may be due to suppression of cortisol secretion. There is an urgent need to trial the combination of dexamethasone and an MR antagonist such as spironolactone to more effectively block the MR and hence the exocytosis of WPBs.
Collapse
|
63
|
Yu WK, McNeil JB, Wickersham NE, Shaver CM, Bastarache JA, Ware LB. Angiopoietin-2 outperforms other endothelial biomarkers associated with severe acute kidney injury in patients with severe sepsis and respiratory failure. Crit Care 2021; 25:48. [PMID: 33541396 PMCID: PMC7859898 DOI: 10.1186/s13054-021-03474-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/20/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Endothelial dysfunction and injury is a major pathophysiologic feature of sepsis. Sepsis is also the most frequent cause of acute kidney injury (AKI) in critically ill patients. Though most studies of AKI in sepsis have focused on tubular epithelial injury, the role of endothelial dysfunction and injury is less well studied. The goal of this study was first to investigate whether endothelial dysfunction and injury biomarkers were associated with severe AKI in sepsis patients. The second goal was to determine the best performing biomarker for severe AKI and whether this biomarker was associated with severe AKI across different etiologies of sepsis and clinical outcomes. METHODS We studied adults with severe sepsis and acute respiratory failure (ARF) enrolled in the prospective observational Validating Acute Lung Injury markers for Diagnosis (VALID) study. Plasma endothelial dysfunction and injury biomarkers, including angiopoietin-2, soluble vascular endothelial cadherin (sVE-cadherin), endocan and syndecan-1, were measured at study enrollment. Primary analysis focused on the association between endothelial biomarker levels with severe AKI (defined as Kidney Disease: Improving Global Outcomes [KDIGO] AKI stage 2 or 3), other organ dysfunctions (defined by Brussels organ failure scores), and comparison of pulmonary versus non-pulmonary sepsis. RESULTS Among 228 sepsis patients enrolled, 141 developed severe AKI. Plasma levels of angiopoietin-2, endocan, sVE-cadherin, and syndecan-1 were significantly higher in sepsis patients with severe AKI compared to those without severe AKI. Among four endothelial biomarkers, only angiopoietin-2 was independently associated with severe AKI (odds ratio 6.07 per log increase, 95% CI 2.34-15.78, p < 0.001). Plasma angiopoietin-2 levels by quartile were significantly higher in sepsis patients with hepatic, coagulation, and circulatory failure. Plasma angiopoietin-2 levels were also significantly higher in patients with non-pulmonary sepsis compared to subjects with pulmonary sepsis. CONCLUSION Among four biomarkers of endothelial dysfunction and injury, angiopoietin-2 had the most robust independent association with development of severe AKI in patients with severe sepsis and ARF. Plasma angiopoietin-2 levels were also associated with other organ dysfunctions, non-pulmonary sepsis, and death. These findings highlight the importance of early endothelial dysfunction and injury in the pathogenesis of sepsis-induced AKI.
Collapse
Affiliation(s)
- Wen-Kuang Yu
- grid.278247.c0000 0004 0604 5314Division of Respiratory Therapy, Department of Chest Medicine, Taipei Veterans General Hospital, Number 201, Section 2, Shipai Road, Beitou District, Taipei City, 11217 Taiwan, ROC ,grid.260770.40000 0001 0425 5914Institute of Physiology, National Yang-Ming University, Taipei, Taiwan ,grid.412807.80000 0004 1936 9916Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, T1218 MCN, 1161 21st, Avenue S, Nashville, TN 37232 USA
| | - J. Brennan McNeil
- grid.412807.80000 0004 1936 9916Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, T1218 MCN, 1161 21st, Avenue S, Nashville, TN 37232 USA
| | - Nancy E. Wickersham
- grid.412807.80000 0004 1936 9916Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, T1218 MCN, 1161 21st, Avenue S, Nashville, TN 37232 USA
| | - Ciara M. Shaver
- grid.412807.80000 0004 1936 9916Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, T1218 MCN, 1161 21st, Avenue S, Nashville, TN 37232 USA
| | - Julie A. Bastarache
- grid.412807.80000 0004 1936 9916Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, T1218 MCN, 1161 21st, Avenue S, Nashville, TN 37232 USA ,grid.152326.10000 0001 2264 7217Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN USA ,grid.152326.10000 0001 2264 7217Department of Medicine and Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, T1218 MCN, 1161 21st, Avenue S, Nashville, TN 37232 USA
| | - Lorraine B. Ware
- grid.412807.80000 0004 1936 9916Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, T1218 MCN, 1161 21st, Avenue S, Nashville, TN 37232 USA ,grid.152326.10000 0001 2264 7217Department of Medicine and Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, T1218 MCN, 1161 21st, Avenue S, Nashville, TN 37232 USA
| |
Collapse
|
64
|
Bos LDJ, Artigas A, Constantin JM, Hagens LA, Heijnen N, Laffey JG, Meyer N, Papazian L, Pisani L, Schultz MJ, Shankar-Hari M, Smit MR, Summers C, Ware LB, Scala R, Calfee CS. Precision medicine in acute respiratory distress syndrome: workshop report and recommendations for future research. Eur Respir Rev 2021; 30:30/159/200317. [PMID: 33536264 DOI: 10.1183/16000617.0317-2020] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 11/11/2020] [Indexed: 12/18/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a devastating critical illness that can be triggered by a wide range of insults and remains associated with a high mortality of around 40%. The search for targeted treatment for ARDS has been disappointing, possibly due to the enormous heterogeneity within the syndrome. In this perspective from the European Respiratory Society research seminar on "Precision medicine in ARDS", we will summarise the current evidence for heterogeneity, explore the evidence in favour of precision medicine and provide a roadmap for further research in ARDS. There is evident variation in the presentation of ARDS on three distinct levels: 1) aetiological; 2) physiological and 3) biological, which leads us to the conclusion that there is no typical ARDS. The lack of a common presentation implies that intervention studies in patients with ARDS need to be phenotype aware and apply a precision medicine approach in order to avoid the lack of success in therapeutic trials that we faced in recent decades. Deeper phenotyping and integrative analysis of the sources of variation might result in identification of additional treatable traits that represent specific pathobiological mechanisms, or so-called endotypes.
Collapse
Affiliation(s)
- Lieuwe D J Bos
- Intensive Care, Amsterdam UMC - location AMC, University of Amsterdam, Amsterdam, The Netherlands .,Laboratory of Intensive Care and Anesthesiology Amsterdam UMC - location AMC, University of Amsterdam, Amsterdam, The Netherlands.,Dept of Respiratory Medicine, Amsterdam UMC - location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Antonio Artigas
- Critical Care Center, Corporació Sanitaria Universitaria Parc Tauli, CIBER Enfermedades Respiratorias, Autonomouus University of Barcelona, Sabadell, Spain
| | - Jean-Michel Constantin
- Dept of Anaesthesiology and Critical Care, Sorbonne University, GRC 29, AP-HP, DMU DREAM, Pitié-Salpêtrière Hospital, Paris, France
| | - Laura A Hagens
- Intensive Care, Amsterdam UMC - location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nanon Heijnen
- Intensive care, Maastricht UMC, University of Maastricht, Maastricht, The Netherlands
| | - John G Laffey
- Anaesthesia and Intensive Care Medicine, School of Medicine, and Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.,Dept of Anaesthesia, University Hospital Galway, Saolta Hospital Group, Galway, Ireland
| | - Nuala Meyer
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Laurent Papazian
- Intensive Care Medicine and regional ECMO center, North hospital - Aix-Marseille University, Marseille, France
| | - Lara Pisani
- Dipartimento Cardio-Toraco-Vascolare, Policlinico S.Orsola-Malpighi, Bologna, Italy
| | - Marcus J Schultz
- Intensive Care, Amsterdam UMC - location AMC, University of Amsterdam, Amsterdam, The Netherlands.,Laboratory of Intensive Care and Anesthesiology Amsterdam UMC - location AMC, University of Amsterdam, Amsterdam, The Netherlands.,Dept of Respiratory Medicine, Amsterdam UMC - location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Manu Shankar-Hari
- School of Immunology & Microbial Sciences, Kings College London, London, UK
| | - Marry R Smit
- Intensive Care, Amsterdam UMC - location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | | | | | - Raffaele Scala
- Respiratory Division with Pulmonary Intensive Care Unit, S. Donato Hospital, Usl Toscana Sudest, Arezzo, Italy
| | - Carolyn S Calfee
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Dept of Medicine, University of California, San Francisco, CA, USA.,Dept of Anesthesia, University of California, San Francisco, CA, USA
| |
Collapse
|
65
|
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
|
66
|
Understanding Restrictive Versus Liberal Fluid Therapy for Major Abdominal Surgery Trial Results: Did Liberal Fluids Associate With Increased Endothelial Injury Markers? Crit Care Explor 2021; 3:e0316. [PMID: 33521643 PMCID: PMC7838007 DOI: 10.1097/cce.0000000000000316] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Supplemental Digital Content is available in the text. Liberal fluid strategies in critically ill patients are associated with harm, thought to be due to endothelial and glycocalyx injury. As the restrictive versus liberal fluid therapy for major abdominal surgery trial not only failed to report survival benefit with restrictive fluids but was associated with a higher rate of acute kidney injury, we hypothesized that factors other than endothelial and glycocalyx injury were likely to account for these findings. Consequently, we measured injury biomarkers in a cohort of the restrictive versus liberal fluid therapy for major abdominal surgery trial.
Collapse
|
67
|
Zhou B, Kojima S, Kawamoto A, Fukushima M. COVID-19 pathogenesis, prognostic factors, and treatment strategy: Urgent recommendations. J Med Virol 2021; 93:2694-2704. [PMID: 33368358 DOI: 10.1002/jmv.26754] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/16/2020] [Accepted: 12/21/2020] [Indexed: 01/08/2023]
Abstract
The pandemic of novel coronavirus disease (COVID-19) is not yet close to being over, more than 8 months after the first cases, but researchers are making great progress in fighting the disease. We have conducted a brief review of the geographic differences in the prevalence of COVID-19, the updated pathological findings, prognostic factors, and treatments for disease prevention and improvement of prognosis. Although hydroxychloroquine and tocilizumab have been recommended by some researchers, many clinical trials have failed to confirm any beneficial effect of these and other drugs on COVID-19, in terms of improved clinical status or reduced patient mortality. Currently, glucocorticoid is the only drug that reduces the mortality of COVID-19 in a randomized controlled trial; however, it is still necessary to establish the optimal timing of administration. It is also urgent to set up an international or national cohort to address the risk factors associated with infection, the natural history of COVID-19, including the disease type, surrogate markers for critically ill, long-term sequelae, and reinfection after exposure, identify responders to glucocorticoid, and establish optimal treatment strategies for disease control.
Collapse
Affiliation(s)
- Bin Zhou
- Department of Medscience, Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation, Kobe, Japan
| | - Shinsuke Kojima
- Department of Medscience, Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation, Kobe, Japan
| | - Atsuhiko Kawamoto
- Department of Medscience, Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation, Kobe, Japan
| | - Masanori Fukushima
- Department of Medscience, Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation, Kobe, Japan
| |
Collapse
|
68
|
Alge J, Dolan K, Angelo J, Thadani S, Virk M, Akcan Arikan A. Two to Tango: Kidney-Lung Interaction in Acute Kidney Injury and Acute Respiratory Distress Syndrome. Front Pediatr 2021; 9:744110. [PMID: 34733809 PMCID: PMC8559585 DOI: 10.3389/fped.2021.744110] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/03/2021] [Indexed: 12/17/2022] Open
Abstract
Acute Kidney Injury (AKI) is an independent risk factor for mortality in hospitalized patients. AKI syndrome leads to fluid overload, electrolyte and acid-base disturbances, immunoparalysis, and propagates multiple organ dysfunction through organ "crosstalk". Preclinical models suggest AKI causes acute lung injury (ALI), and conversely, mechanical ventilation and ALI cause AKI. In the clinical setting, respiratory complications are a key driver of increased mortality in patients with AKI, highlighting the bidirectional relationship. This article highlights the challenging and complex interactions between the lung and kidney in critically ill patients with AKI and acute respiratory distress syndrome (ARDS) and global implications of AKI. We discuss disease-specific molecular mediators and inflammatory pathways involved in organ crosstalk in the AKI-ARDS construct, and highlight the reciprocal hemodynamic effects of elevated pulmonary vascular resistance and central venous pressure (CVP) leading to renal hypoperfusion and pulmonary edema associated with fluid overload and increased right ventricular afterload. Finally, we discuss the notion of different ARDS "phenotypes" and the response to fluid overload, suggesting differential organ crosstalk in specific pathological states. While the directionality of effect remains challenging to distinguish at the bedside due to lag in diagnosis with conventional renal function markers and lack of tangible damage markers, this review provides a paradigm for understanding kidney-lung interactions in the critically ill patient.
Collapse
Affiliation(s)
- Joseph Alge
- Division of Nephrology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Kristin Dolan
- Division of Nephrology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States.,Division of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Joseph Angelo
- Division of Nephrology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Sameer Thadani
- Division of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Manpreet Virk
- Division of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Ayse Akcan Arikan
- Division of Nephrology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States.,Division of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| |
Collapse
|
69
|
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: 10] [Impact Index Per Article: 2.0] [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
|
70
|
Kangelaris KN, Clemens R, Fang X, Jauregui A, Liu T, Vessel K, Deiss T, Sinha P, Leligdowicz A, Liu KD, Zhuo H, Alder MN, Wong HR, Calfee CS, Lowell C, Matthay MA. A neutrophil subset defined by intracellular olfactomedin 4 is associated with mortality in sepsis. Am J Physiol Lung Cell Mol Physiol 2020; 320:L892-L902. [PMID: 33355521 DOI: 10.1152/ajplung.00090.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Sepsis is a heterogeneous syndrome clinically and biologically, but biomarkers of distinct host response pathways for early prognostic information and testing targeted treatments are lacking. Olfactomedin 4 (OLFM4), a matrix glycoprotein of neutrophil-specific granules, defines a distinct neutrophil subset that may be an independent risk factor for poor outcomes in sepsis. We hypothesized that increased percentage of OLFM4+ neutrophils on sepsis presentation would be associated with mortality. In a single-center, prospective cohort study, we enrolled adults admitted to an academic medical center from the emergency department (ED) with suspected sepsis [identified by 2 or greater systemic inflammatory response syndrome (SIRS) criteria and antibiotic receipt] from March 2016 through December 2017, followed by sepsis adjudication according to Sepsis-3. We collected 200 µL of whole blood within 24 h of admission and stained for the neutrophil surface marker CD66b followed by intracellular staining for OLFM4 quantitated by flow cytometry. The predictors for 60-day mortality were 1) percentage of OLFM4+ neutrophils and 2) OLFM4+ neutrophils at a cut point of ≥37.6% determined by the Youden Index. Of 120 enrolled patients with suspected sepsis, 97 had sepsis and 23 had nonsepsis SIRS. The mean percentage of OLFM4+ neutrophils was significantly increased in both sepsis and nonsepsis SIRS patients who died (P ≤ 0.01). Among sepsis patients with elevated OLFM4+ (≥37.6%), 56% died, compared with 18% with OLFM4+ <37.6% (P = 0.001). The association between OLFM4+ and mortality withstood adjustment for age, sex, absolute neutrophil count, comorbidities, and standard measures of severity of illness (SOFA score, APACHE III) (P < 0.03). In summary, OLFM4+ neutrophil percentage is independently associated with 60-day mortality in sepsis and may represent a novel measure of the heterogeneity of host response to sepsis.
Collapse
Affiliation(s)
- Kirsten N Kangelaris
- Division of Hospital Medicine, Department of Medicine, University of California, San Francisco, California
| | - Regina Clemens
- Division of Hospital Medicine, Department of Medicine, University of California, San Francisco, California
| | - Xiaohui Fang
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California.,Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Alejandra Jauregui
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California.,Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Tom Liu
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California.,Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Kathryn Vessel
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California.,Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Thomas Deiss
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California.,Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Pratik Sinha
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California.,Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Aleksandra Leligdowicz
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California.,Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California.,Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Kathleen D Liu
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California.,Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Hanjing Zhuo
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California.,Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Matthew N Alder
- Division of Critical Care Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Hector R Wong
- Division of Critical Care Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Carolyn S Calfee
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California.,Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California
| | - Clifford Lowell
- Department of Laboratory Medicine, University of California, San Francisco, California
| | - Michael A Matthay
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California.,Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California
| |
Collapse
|
71
|
Bime C, Camp SM, Casanova N, Oita RC, Ndukum J, Lynn H, Garcia JGN. The acute respiratory distress syndrome biomarker pipeline: crippling gaps between discovery and clinical utility. Transl Res 2020; 226:105-115. [PMID: 32599095 PMCID: PMC7319618 DOI: 10.1016/j.trsl.2020.06.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/17/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022]
Abstract
Recent innovations in translational research have ushered an exponential increase in the discovery of novel biomarkers, thereby elevating the hope for deeper insights into "personalized" medicine approaches to disease phenotyping and care. However, a critical gap exists between the fast pace of biomarker discovery and the successful translation to clinical use. This gap underscores the fundamental biomarker conundrum across various acute and chronic disorders: how does a biomarker address a specific unmet need? Additionally, the gap highlights the need to shift the paradigm from a focus on biomarker discovery to greater translational impact and the need for a more streamlined drug approval process. The unmet need for biomarkers in acute respiratory distress syndrome (ARDS) is for reliable and validated biomarkers that minimize heterogeneity and allow for stratification of subject selection for enrollment in clinical trials of tailored therapies. This unmet need is particularly highlighted by the ongoing SARS-CoV-2/COVID-19 pandemic. The unprecedented numbers of COVID-19-induced ARDS cases has strained health care systems across the world and exposed the need for biomarkers that would accelerate drug development and the successful phenotyping of COVID-19-infected patients at risk for development of ARDS and ARDS mortality. Accordingly, this review discusses the current state of ARDS biomarkers in the context of the drug development pipeline and highlight gaps between biomarker discovery and clinical implementation while proposing potential paths forward. We discuss potential ARDS biomarkers by category and by context of use, highlighting progress in the development continuum. We conclude by discussing challenges to successful translation of biomarker candidates to clinical impact and proposing possible novel strategies.
Collapse
Affiliation(s)
- Christian Bime
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona.
| | - Sara M Camp
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Nancy Casanova
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Radu C Oita
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Juliet Ndukum
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Heather Lynn
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Joe G N Garcia
- College of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| |
Collapse
|
72
|
Kwong YD, Mehta KM, Miaskowski C, Zhuo H, Yee K, Jauregui A, Ke S, Deiss T, Abbott J, Kangelaris KN, Sinha P, Hendrickson C, Gomez A, Leligdowicz A, Matthay MA, Calfee CS, Liu KD. Using best subset regression to identify clinical characteristics and biomarkers associated with sepsis-associated acute kidney injury. Am J Physiol Renal Physiol 2020; 319:F979-F987. [PMID: 33044866 PMCID: PMC7792692 DOI: 10.1152/ajprenal.00281.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/24/2020] [Accepted: 10/07/2020] [Indexed: 12/23/2022] Open
Abstract
Sepsis-associated acute kidney injury (AKI) is a complex clinical disorder associated with inflammation, endothelial dysfunction, and dysregulated coagulation. With standard regression methods, collinearity among biomarkers may lead to the exclusion of important biological pathways in a single final model. Best subset regression is an analytic technique that identifies statistically equivalent models, allowing for more robust evaluation of correlated variables. Our objective was to identify common clinical characteristics and biomarkers associated with sepsis-associated AKI. We enrolled 453 septic adults within 24 h of intensive care unit admission. Using best subset regression, we evaluated for associations using a range of models consisting of 1-38 predictors (composed of clinical risk factors and plasma and urine biomarkers) with AKI as the outcome [defined as a serum creatinine (SCr) increase of ≥0.3 mg/dL within 48 h or ≥1.5× baseline SCr within 7 days]. Two hundred ninety-seven patients had AKI. Five-variable models were found to be of optimal complexity, as the best subset of five- and six-variable models were statistically equivalent. Within the subset of five-variable models, 46 permutations of predictors were noted to be statistically equivalent. The most common predictors in this subset included diabetes, baseline SCr, angiopoetin-2, IL-8, soluble tumor necrosis factor receptor-1, and urine neutrophil gelatinase-associated lipocalin. The models had a c-statistic of ∼0.70 (95% confidence interval: 0.65-0.75). In conclusion, using best subset regression, we identified common clinical characteristics and biomarkers associated with sepsis-associated AKI. These variables may be especially relevant in the pathogenesis of sepsis-associated AKI.
Collapse
Affiliation(s)
- Y Diana Kwong
- Division of Nephrology, Department of Medicine, University of California, San Francisco, California
| | - Kala M Mehta
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California
| | - Christine Miaskowski
- Department of Physiological Nursing, University of California, San Francisco, California
| | - Hanjing Zhuo
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Kimberly Yee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Alejandra Jauregui
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Serena Ke
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Thomas Deiss
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Jason Abbott
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Kirsten N Kangelaris
- Division of Hospital Medicine, Department of Medicine, University of California, San Francisco, California
| | - Pratik Sinha
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Carolyn Hendrickson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Antonio Gomez
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Aleksandra Leligdowicz
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Michael A Matthay
- Cardiovascular Research Institute, Department of Medicine and Department of Anesthesia, University of California, San Francisco, California
| | - Carolyn S Calfee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, California
| | - Kathleen D Liu
- Division of Nephrology, Department of Medicine, University of California, San Francisco, California
- Division of Critical Care Medicine, Department of Anesthesia, University of California, San Francisco, California
| |
Collapse
|
73
|
Alternative Tobacco Product Use in Critically Ill Patients. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17238707. [PMID: 33255164 PMCID: PMC7727672 DOI: 10.3390/ijerph17238707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/20/2020] [Accepted: 11/20/2020] [Indexed: 01/15/2023]
Abstract
Background: Alternative tobacco product (ATP) use has bee linked to critical illness, however, few studies have examined the use of these substances in critically ill populations. We sought to examine ATP use within critically ill patients and to define barriers in accurately assessing use within this population. Methods: We prospectively studied 533 consecutive patients from the Early Assessment of Renal and Lung Injury study, enrolled between 2013 and 2016 at a tertiary referral center and a safety-net hospital. ATP use information (electronic cigarettes, cigars, pipes, hookahs/waterpipes, and snus/chewing tobacco) was obtained from the patient or surrogate using a detailed survey. Reasons for non-completion of the survey were recorded, and differences between survey responders vs. non-responders, self- vs. surrogate responders, and ATP users vs. non-users were explored. Results: Overall, 80% (n = 425) of subjects (56% male) completed a tobacco product use survey. Of these, 12.2% (n = 52) reported current ATP use, while 5.6% reported using multiple ATP products. When restricted to subjects who were self-responders, 17% reported ATP use, while 10% reported current cigarette smoking alone. The mean age of ATP users was 57 ± 17 years. Those who did not complete a survey were sicker and more likely to have died during admission. Subjects who completed the survey as self-responders reported higher levels of ATP use than ones with surrogate responders (p < 0.0001). Conclusion: ATP use is common among critically ill patients despite them being generally older than traditional users. Survey self-responders were more likely than surrogate responders to report use. These findings highlight the importance of improving our current methods of surveillance of ATP use in older adults in the outpatient setting.
Collapse
|
74
|
Vassiliou AG, Kotanidou A, Dimopoulou I, Orfanos SE. Endothelial Damage in Acute Respiratory Distress Syndrome. Int J Mol Sci 2020; 21:ijms21228793. [PMID: 33233715 PMCID: PMC7699909 DOI: 10.3390/ijms21228793] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/14/2020] [Accepted: 11/18/2020] [Indexed: 01/01/2023] Open
Abstract
The pulmonary endothelium is a metabolically active continuous monolayer of squamous endothelial cells that internally lines blood vessels and mediates key processes involved in lung homoeostasis. Many of these processes are disrupted in acute respiratory distress syndrome (ARDS), which is marked among others by diffuse endothelial injury, intense activation of the coagulation system and increased capillary permeability. Most commonly occurring in the setting of sepsis, ARDS is a devastating illness, associated with increased morbidity and mortality and no effective pharmacological treatment. Endothelial cell damage has an important role in the pathogenesis of ARDS and several biomarkers of endothelial damage have been tested in determining prognosis. By further understanding the endothelial pathobiology, development of endothelial-specific therapeutics might arise. In this review, we will discuss the underlying pathology of endothelial dysfunction leading to ARDS and emerging therapies. Furthermore, we will present a brief overview demonstrating that endotheliopathy is an important feature of hospitalised patients with coronavirus disease-19 (COVID-19).
Collapse
Affiliation(s)
- Alice G. Vassiliou
- 1st Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (A.G.V.); (A.K.); (I.D.)
| | - Anastasia Kotanidou
- 1st Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (A.G.V.); (A.K.); (I.D.)
| | - Ioanna Dimopoulou
- 1st Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (A.G.V.); (A.K.); (I.D.)
| | - Stylianos E. Orfanos
- 1st Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, Evangelismos Hospital, 106 76 Athens, Greece; (A.G.V.); (A.K.); (I.D.)
- 2nd Department of Critical Care, School of Medicine, National and Kapodistrian University of Athens, Attikon Hospital, 124 62 Athens, Greece
- Correspondence: or ; Tel.: +30-2107-235-521
| |
Collapse
|
75
|
Angiopoietin-2 is released during anaphylactic hypotension in anesthetized and unanesthetized rats. PLoS One 2020; 15:e0242026. [PMID: 33201925 PMCID: PMC7671552 DOI: 10.1371/journal.pone.0242026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 10/26/2020] [Indexed: 11/19/2022] Open
Abstract
Angiopoietin (Angpt)-2, a permeability-increasing growth factor, is involved in vascular leakage of sepsis and acute lung injury, and could be released from endothelium in response to anaphylaxis-related secretagogues such as histamine and leukotrienes, or cytokines. However, roles of Angpt-2 in the hyperpermeability during systemic anaphylaxis are not known. Thus, we determined plasma levels of Angpt-2 and cytokines and vascular permeability during anaphylactic hypotension in unanesthetized rats. Anaphylaxis was induced by an intravenous injection of ovalbumin antigen. Mean arterial blood pressure (MBP) was measured, and hematocrit (Hct) and plasma levels of Angpt-2 and cytokines were assessed for 24 h after antigen injection. Separately, vascular permeability was measured in various organs using the Evans blue dye method, and Angpt-2 mRNA expression in liver was measured. After antigen injection, MBP decreased to the nadir at 6 min, and returned to baseline at 45 min, and Hct peaked at 20 min and thereafter progressively declined, suggesting that vascular leak and hypotension occurred within 20 min. Plasma Angpt-2 levels began to increase significantly at 1 h after antigen, reaching the peak 2.7-fold baseline at 6 h with a return to baseline at 24 h. Detected cytokines of IL-1α, IL-1β, IL-6, IL-10, and TNF-α peaked 1 or 2 h after antigen. Angpt-2 mRNA increased at 2 h and showed an increasing tendency at 6 h. Vascular permeability in bronchus, trachea, intestines, mesentery and skeletal muscle was increased at 10 min but not at 6 h after antigen. In addition, we confirmed using anesthetized rat anaphylaxis models that plasma Angpt-2 levels increased at 1 h after antigen. In conclusion, plasma Angpt-2 is elevated presumably due to increased cytokines and enhanced gene transcription during anaphylaxis in anesthetized and unanesthetized rats.
Collapse
|
76
|
Potential Value of Biomarker Signatures in Sepsis and Acute Respiratory Distress Syndrome in Children and Adults. Crit Care Med 2020; 48:428-430. [PMID: 32058380 DOI: 10.1097/ccm.0000000000004169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
77
|
Gustafson D, Raju S, Wu R, Ching C, Veitch S, Rathnakumar K, Boudreau E, Howe KL, Fish JE. Overcoming Barriers: The Endothelium As a Linchpin of Coronavirus Disease 2019 Pathogenesis? Arterioscler Thromb Vasc Biol 2020; 40:1818-1829. [PMID: 32510978 PMCID: PMC7370857 DOI: 10.1161/atvbaha.120.314558] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 05/26/2020] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Coronavirus disease 2019 (COVID-19) is a global pandemic involving >5 500 000 cases worldwide as of May 26, 2020. The culprit is the severe acute respiratory syndrome coronavirus-2, which invades cells by binding to ACE2 (angiotensin-converting enzyme 2). While the majority of patients mount an appropriate antiviral response and recover at home, others progress to respiratory distress requiring hospital admission for supplemental oxygen. In severe cases, deterioration to acute respiratory distress syndrome necessitating mechanical ventilation, development of severe thrombotic events, or cardiac injury and dysfunction occurs. In this review, we highlight what is known to date about COVID-19 and cardiovascular risk, focusing in on the putative role of the endothelium in disease susceptibility and pathogenesis. Approach and Results: Cytokine-driven vascular leak in the lung alveolar-endothelial interface facilitates acute lung injury in the setting of viral infection. Given that the virus affects multiple organs, including the heart, it likely gains access into systemic circulation by infecting or passing from the respiratory epithelium to the endothelium for viral dissemination. Indeed, cardiovascular complications of COVID-19 are highly prevalent and include acute cardiac injury, myocarditis, and a hypercoagulable state, all of which may be influenced by altered endothelial function. Notably, the disease course is worse in individuals with preexisting comorbidities that involve endothelial dysfunction and may be linked to elevated ACE2 expression, such as diabetes mellitus, hypertension, and cardiovascular disease. CONCLUSIONS Rapidly emerging data on COVID-19, together with results from studies on severe acute respiratory syndrome coronavirus-1, are providing insight into how endothelial dysfunction may contribute to the pandemic that is paralyzing the globe. This may, in turn, inform the design of biomarkers predictive of disease course, as well as therapeutics targeting pathogenic endothelial responses.
Collapse
Affiliation(s)
- Dakota Gustafson
- From the Toronto General Hospital Research Institute, University Health Network, Canada (D.G., S.R., R.W., C.C., S.V., K.R., E.B., K.L.H., J.E.F.)
- Department of Laboratory Medicine and Pathobiology (D.G., R.W., S.V., J.E.F.), University of Toronto, Canada
| | - Sneha Raju
- From the Toronto General Hospital Research Institute, University Health Network, Canada (D.G., S.R., R.W., C.C., S.V., K.R., E.B., K.L.H., J.E.F.)
- Institute of Medical Science (S.R., C.C., K.L.H., J.E.F.), University of Toronto, Canada
- Division of Vascular Surgery (S.R., K.L.H.), Toronto General Hospital, Canada
| | - Ruilin Wu
- From the Toronto General Hospital Research Institute, University Health Network, Canada (D.G., S.R., R.W., C.C., S.V., K.R., E.B., K.L.H., J.E.F.)
- Department of Laboratory Medicine and Pathobiology (D.G., R.W., S.V., J.E.F.), University of Toronto, Canada
| | - Crizza Ching
- From the Toronto General Hospital Research Institute, University Health Network, Canada (D.G., S.R., R.W., C.C., S.V., K.R., E.B., K.L.H., J.E.F.)
- Institute of Medical Science (S.R., C.C., K.L.H., J.E.F.), University of Toronto, Canada
| | - Shawn Veitch
- From the Toronto General Hospital Research Institute, University Health Network, Canada (D.G., S.R., R.W., C.C., S.V., K.R., E.B., K.L.H., J.E.F.)
- Department of Laboratory Medicine and Pathobiology (D.G., R.W., S.V., J.E.F.), University of Toronto, Canada
| | - Kumaragurubaran Rathnakumar
- From the Toronto General Hospital Research Institute, University Health Network, Canada (D.G., S.R., R.W., C.C., S.V., K.R., E.B., K.L.H., J.E.F.)
| | - Emilie Boudreau
- From the Toronto General Hospital Research Institute, University Health Network, Canada (D.G., S.R., R.W., C.C., S.V., K.R., E.B., K.L.H., J.E.F.)
| | - Kathryn L. Howe
- From the Toronto General Hospital Research Institute, University Health Network, Canada (D.G., S.R., R.W., C.C., S.V., K.R., E.B., K.L.H., J.E.F.)
- Institute of Medical Science (S.R., C.C., K.L.H., J.E.F.), University of Toronto, Canada
- Division of Vascular Surgery (S.R., K.L.H.), Toronto General Hospital, Canada
- Peter Munk Cardiac Centre (K.L.H., J.E.F.), Toronto General Hospital, Canada
| | - Jason E. Fish
- From the Toronto General Hospital Research Institute, University Health Network, Canada (D.G., S.R., R.W., C.C., S.V., K.R., E.B., K.L.H., J.E.F.)
- Department of Laboratory Medicine and Pathobiology (D.G., R.W., S.V., J.E.F.), University of Toronto, Canada
- Institute of Medical Science (S.R., C.C., K.L.H., J.E.F.), University of Toronto, Canada
- Peter Munk Cardiac Centre (K.L.H., J.E.F.), Toronto General Hospital, Canada
| |
Collapse
|
78
|
Fernandez-Bustamante A, Sprung J, Parker RA, Bartels K, Weingarten TN, Kosour C, Thompson BT, Vidal Melo MF. Individualized PEEP to optimise respiratory mechanics during abdominal surgery: a pilot randomised controlled trial. Br J Anaesth 2020; 125:383-392. [PMID: 32682559 DOI: 10.1016/j.bja.2020.06.030] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 05/24/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Higher intraoperative driving pressures (ΔP) are associated with increased postoperative pulmonary complications (PPC). We hypothesised that dynamic adjustment of PEEP throughout abdominal surgery reduces ΔP, maintains positive end-expiratory transpulmonary pressures (Ptp_ee) and increases respiratory system static compliance (Crs) with PEEP levels that are variable between and within patients. METHODS In a prospective multicentre pilot study, adults at moderate/high risk for PPC undergoing elective abdominal surgery were randomised to one of three ventilation protocols: (1) PEEP≤2 cm H2O, compared with periodic recruitment manoeuvres followed by individualised PEEP to either optimise respiratory system compliance (PEEPmaxCrs) or maintain positive end-expiratory transpulmonary pressure (PEEPPtp_ee). The composite primary outcome included intraoperative ΔP, Ptp_ee, Crs, and PEEP values (median (interquartile range) and coefficients of variation [CVPEEP]). RESULTS Thirty-seven patients (48.6% female; age range: 47-73 yr) were assigned to control (PEEP≤2 cm H2O; n=13), PEEPmaxCrs (n=16), or PEEPPtp_ee (n=8) groups. The PEEPPtp_ee intervention could not be delivered in two patients. Subjects assigned to PEEPmaxCrs had lower ΔP (median8 cm H2O [7-10]), compared with the control group (12 cm H2O [10-15]; P=0.006). PEEPmaxCrs was also associated with higher Ptp_ee (2.0 cm H2O [-0.7 to 4.5] vs controls: -8.3 cm H2O [-13.0 to -4.0]; P≤0.001) and higher Crs (47.7 ml cm H2O [43.2-68.8] vs controls: 39.0 ml cm H2O [32.9-43.4]; P=0.009). Individualised PEEP (PEEPmaxCrs and PEEPPtp_ee combined) varied widely (median: 10 cm H2O [8-15]; CVPEEP=0.24 [0.14-0.35]), both between, and within, subjects throughout surgery. CONCLUSIONS This pilot study suggests that individualised PEEP management strategies applied during abdominal surgery reduce driving pressure, maintain positive Ptp_ee and increase static compliance. The wide range of PEEP observed suggests that an individualised approach is required to optimise respiratory mechanics during abdominal surgery. CLINICAL TRIAL REGISTRATION NCT02671721.
Collapse
Affiliation(s)
- Ana Fernandez-Bustamante
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA; Webb-Waring Center, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Juraj Sprung
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Robert A Parker
- Department of Medicine, Biostatistics Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Karsten Bartels
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Toby N Weingarten
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Carolina Kosour
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - B Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marcos F Vidal Melo
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
79
|
Auriemma CL, Zhuo H, Delucchi K, Deiss T, Liu T, Jauregui A, Ke S, Vessel K, Lippi M, Seeley E, Kangelaris KN, Gomez A, Hendrickson C, Liu KD, Matthay MA, Ware LB, Calfee CS. Acute respiratory distress syndrome-attributable mortality in critically ill patients with sepsis. Intensive Care Med 2020; 46:1222-1231. [PMID: 32206845 PMCID: PMC7224051 DOI: 10.1007/s00134-020-06010-9] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 03/11/2020] [Indexed: 11/18/2022]
Abstract
PURPOSE Previous studies assessing impact of acute respiratory distress syndrome (ARDS) on mortality have shown conflicting results. We sought to assess the independent association of ARDS with in-hospital mortality among intensive care unit (ICU) patients with sepsis. METHODS We studied two prospective sepsis cohorts drawn from the Early Assessment of Renal and Lung Injury (EARLI; n = 474) and Validating Acute Lung Injury markers for Diagnosis (VALID; n = 337) cohorts. ARDS was defined by Berlin criteria. We used logistic regression to compare in-hospital mortality in patients with and without ARDS, controlling for baseline severity of illness. We also estimated attributable mortality, adjusted for illness severity by stratification. RESULTS ARDS occurred in 195 EARLI patients (41%) and 99 VALID patients (29%). ARDS was independently associated with risk of hospital death in multivariate analysis, even after controlling for severity of illness, as measured by APACHE II (odds ratio [OR] 1.65 (95% confidence interval [CI] 1.02, 2.67), p = 0.04 in EARLI; OR 2.12 (CI 1.16, 3.92), p = 0.02 in VALID). Patients with severe ARDS (P/F < 100) primarily drove this relationship. The attributable mortality of ARDS was 27% (CI 14%, 37%) in EARLI and 37% (CI 10%, 51%) in VALID. ARDS was independently associated with ICU mortality, hospital length of stay (LOS), ICU LOS, and ventilator-free days. CONCLUSIONS Development of ARDS among ICU patients with sepsis confers increased risk of ICU and in-hospital mortality in addition to other important outcomes. Clinical trials targeting patients with severe ARDS will be best poised to detect measurable differences in these outcomes.
Collapse
Affiliation(s)
- Catherine L Auriemma
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Hospital of the University of Pennsylvania, 839 W. Gates, 3600 Spruce Street, Philadelphia, PA, 19103-4283, USA.
- Palliative and Advanced Illness Research (PAIR) Center, University of Pennsylvania, Philadelphia, PA, USA.
| | - Hanjing Zhuo
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Kevin Delucchi
- Department of Psychiatry, University of California, San Francisco, USA
| | - Thomas Deiss
- Department of Biochemistry and Biophysics, University of California, San Francisco, USA
| | - Tom Liu
- Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| | | | - Serena Ke
- Department of Pulmonary and Critical Care Medicine, Zuckerberg San Francisco General Hospital and Trauma Center, University of California, San Francisco, USA
| | - Kathryn Vessel
- Department of Medicine, University of California, San Francisco, USA
| | - Matthew Lippi
- University of Colorado School of Medicine, Aurora, CO, USA
| | - Eric Seeley
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, USA
| | | | - Antonio Gomez
- Department of Pulmonary and Critical Care Medicine, Zuckerberg San Francisco General Hospital and Trauma Center, University of California, San Francisco, USA
| | - Carolyn Hendrickson
- Department of Pulmonary and Critical Care Medicine, Zuckerberg San Francisco General Hospital and Trauma Center, University of California, San Francisco, USA
| | - Kathleen D Liu
- Division of Nephrology, Department of Medicine, University of California, San Francisco, USA
- Department of Anesthesia, University of California San Francisco, San Francisco, CA, USA
| | - Michael A Matthay
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, USA
- Department of Anesthesia, University of California San Francisco, San Francisco, CA, USA
| | - Lorraine B Ware
- Departments of Medicine and Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, USA
| | - Carolyn S Calfee
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, USA
- Department of Anesthesia, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
80
|
van der Zee P, Rietdijk W, Somhorst P, Endeman H, Gommers D. A systematic review of biomarkers multivariately associated with acute respiratory distress syndrome development and mortality. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:243. [PMID: 32448370 PMCID: PMC7245629 DOI: 10.1186/s13054-020-02913-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022]
Abstract
Background Heterogeneity of acute respiratory distress syndrome (ARDS) could be reduced by identification of biomarker-based phenotypes. The set of ARDS biomarkers to prospectively define these phenotypes remains to be established. Objective To provide an overview of the biomarkers that were multivariately associated with ARDS development or mortality. Data sources We performed a systematic search in Embase, MEDLINE, Web of Science, Cochrane CENTRAL, and Google Scholar from inception until 6 March 2020. Study selection Studies assessing biomarkers for ARDS development in critically ill patients at risk for ARDS and mortality due to ARDS adjusted in multivariate analyses were included. Data extraction and synthesis We included 35 studies for ARDS development (10,667 patients at risk for ARDS) and 53 for ARDS mortality (15,344 patients with ARDS). These studies were too heterogeneous to be used in a meta-analysis, as time until outcome and the variables used in the multivariate analyses varied widely between studies. After qualitative inspection, high plasma levels of angiopoeitin-2 and receptor for advanced glycation end products (RAGE) were associated with an increased risk of ARDS development. None of the biomarkers (plasma angiopoeitin-2, C-reactive protein, interleukin-8, RAGE, surfactant protein D, and Von Willebrand factor) was clearly associated with mortality. Conclusions Biomarker data reporting and variables used in multivariate analyses differed greatly between studies. Angiopoeitin-2 and RAGE in plasma were positively associated with increased risk of ARDS development. None of the biomarkers independently predicted mortality. Therefore, we suggested to structurally investigate a combination of biomarkers and clinical parameters in order to find more homogeneous ARDS phenotypes. PROSPERO identifier PROSPERO, CRD42017078957
Collapse
Affiliation(s)
- Philip van der Zee
- Department of Adult Intensive Care, Erasmus Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| | - Wim Rietdijk
- Department of Adult Intensive Care, Erasmus Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Peter Somhorst
- Department of Adult Intensive Care, Erasmus Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Henrik Endeman
- Department of Adult Intensive Care, Erasmus Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Diederik Gommers
- Department of Adult Intensive Care, Erasmus Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| |
Collapse
|
81
|
Metwaly SM, Winston BW. Systems Biology ARDS Research with a Focus on Metabolomics. Metabolites 2020; 10:metabo10050207. [PMID: 32438561 PMCID: PMC7281154 DOI: 10.3390/metabo10050207] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/09/2020] [Accepted: 05/15/2020] [Indexed: 12/19/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a clinical syndrome that inflicts a considerably heavy toll in terms of morbidity and mortality. While there are multitudes of conditions that can lead to ARDS, the vast majority of ARDS cases are caused by a relatively small number of diseases, especially sepsis and pneumonia. Currently, there is no clinically agreed upon reliable diagnostic test for ARDS, and the detection or diagnosis of ARDS is based on a constellation of laboratory and radiological tests in the absence of evidence of left ventricular dysfunction, as specified by the Berlin definition of ARDS. Virtually all the ARDS biomarkers to date have been proven to be of very limited clinical utility. Given the heterogeneity of ARDS due to the wide variation in etiology, clinical and molecular manifestations, there is a current scientific consensus agreement that ARDS is not just a single entity but rather a spectrum of conditions that need further study for proper classification, the identification of reliable biomarkers and the adequate institution of therapeutic targets. This scoping review aims to elucidate ARDS omics research, focusing on metabolomics and how metabolomics can boost the study of ARDS biomarkers and help to facilitate the identification of ARDS subpopulations.
Collapse
Affiliation(s)
- Sayed M. Metwaly
- Department of Critical Care Medicine, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada;
| | - Brent W. Winston
- Department of Critical Care Medicine, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada;
- Departments of Medicine and Biochemistry and Molecular Biology, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Correspondence: ; Tel.: +1-(403)-220-4331; Fax: +1-(403)-283-1267
| |
Collapse
|
82
|
Rogers CJ, Harman RJ, Bunnell BA, Schreiber MA, Xiang C, Wang FS, Santidrian AF, Minev BR. Rationale for the clinical use of adipose-derived mesenchymal stem cells for COVID-19 patients. J Transl Med 2020; 18:203. [PMID: 32423449 PMCID: PMC7232924 DOI: 10.1186/s12967-020-02380-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 05/14/2020] [Indexed: 02/08/2023] Open
Abstract
In late 2019, a novel coronavirus (SARS-CoV-2) emerged in Wuhan, capital city of Hubei province in China. Cases of SARS-CoV-2 infection quickly grew by several thousand per day. Less than 100 days later, the World Health Organization declared that the rapidly spreading viral outbreak had become a global pandemic. Coronavirus disease 2019 (COVID-19) is typically associated with fever and respiratory symptoms. It often progresses to severe respiratory distress and multi-organ failure which carry a high mortality rate. Older patients or those with medical comorbidities are at greater risk for severe disease. Inflammation, pulmonary edema and an over-reactive immune response can lead to hypoxia, respiratory distress and lung damage. Mesenchymal stromal/stem cells (MSCs) possess potent and broad-ranging immunomodulatory activities. Multiple in vivo studies in animal models and ex vivo human lung models have demonstrated the MSC's impressive capacity to inhibit lung damage, reduce inflammation, dampen immune responses and aid with alveolar fluid clearance. Additionally, MSCs produce molecules that are antimicrobial and reduce pain. Upon administration by the intravenous route, the cells travel directly to the lungs where the majority are sequestered, a great benefit for the treatment of pulmonary disease. The in vivo safety of local and intravenous administration of MSCs has been demonstrated in multiple human clinical trials, including studies of acute respiratory distress syndrome (ARDS). Recently, the application of MSCs in the context of ongoing COVID-19 disease and other viral respiratory illnesses has demonstrated reduced patient mortality and, in some cases, improved long-term pulmonary function. Adipose-derived stem cells (ASC), an abundant type of MSC, are proposed as a therapeutic option for the treatment of COVID-19 in order to reduce morbidity and mortality. Additionally, when proven to be safe and effective, ASC treatments may reduce the demand on critical hospital resources. The ongoing COVID-19 outbreak has resulted in significant healthcare and socioeconomic burdens across the globe. There is a desperate need for safe and effective treatments. Cellular based therapies hold great promise for the treatment of COVID-19. This literature summary reviews the scientific rationale and need for clinical studies of adipose-derived stem cells and other types of mesenchymal stem cells in the treatment of patients who suffer with COVID-19.
Collapse
Affiliation(s)
| | | | - Bruce A. Bunnell
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA USA
| | - Martin A. Schreiber
- Department of Surgery, Oregon Health and Science University, Portland, OR USA
| | - Charlie Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003 China
| | - Fu-Sheng Wang
- Treatment and Research Center for Infectious Diseases, The Fifth Medical Center, Beijing, 100039 China
| | | | - Boris R. Minev
- Calidi Biotherapeutics, Inc., San Diego, CA USA
- Department of Radiation Medicine and Applied Sciences, Moores UCSD Cancer Center, San Diego, CA USA
| |
Collapse
|
83
|
Abstract
Biomarker panels have the potential to advance the field of critical care medicine by stratifying patients according to prognosis and/or underlying pathophysiology. This article discusses the discovery and validation of biomarker panels, along with their translation to the clinical setting. The current literature on the use of biomarker panels in sepsis, acute respiratory distress syndrome, and acute kidney injury is reviewed.
Collapse
Affiliation(s)
- Susan R Conway
- Division of Critical Care Medicine, Children's National Medical Center, 111 Michigan Avenue Northwest, Washington, DC 20010, USA; Department of Pediatrics, George Washington University School of Medicine, Washington, DC, USA.
| | - Hector R Wong
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati Children's Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
84
|
Shah J, Rana SS. Acute respiratory distress syndrome in acute pancreatitis. Indian J Gastroenterol 2020; 39:123-132. [PMID: 32285399 DOI: 10.1007/s12664-020-01016-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 01/14/2020] [Indexed: 02/07/2023]
Abstract
Development of organ failure is one of the major determinants of mortality in patients with acute pancreatitis (AP). Acute respiratory distress syndrome (ARDS) is an important cause of respiratory failure in AP and is associated with high mortality. Pathogenesis of ARDS in AP is incompletely understood. Release of various cytokines plays an important role in development of ARDS in AP. Increased gut permeability due to various toxins, inflammatory mediators, and pancreatic enzymes potentiates lung injury by gut-lymph-lung axis leading on to increased translocation of bacterial endotoxins. Various scoring systems, serum levels of various cytokines and lung ultrasound have been evaluated for prediction of development of ARDS in AP with varying results. Various drugs have shown encouraging results in prevention of ARDS in animal models but these encouraging results in animal models are yet to be confirmed in clinical studies. There is no specific effective treatment for ARDS. Treatment of sepsis and local complications of AP should be done according to the standard management strategies. Lung protective ventilatory strategies are of paramount importance to improve outcome of patients of AP with ARDS and therefore effective coordination between gastroenterologists and intensivists is needed for effective management of these patients.
Collapse
Affiliation(s)
- Jimil Shah
- Department of Gastroenterology, Post Graduate Institute of Medical Education and Research, Sector 12, Chandigarh, 160 012, India
| | - Surinder S Rana
- Department of Gastroenterology, Post Graduate Institute of Medical Education and Research, Sector 12, Chandigarh, 160 012, India.
| |
Collapse
|
85
|
Abstract
Ventilation-induced lung injury results from mechanical stress and strain that occur during tidal ventilation in the susceptible lung. Classical descriptions of ventilation-induced lung injury have focused on harm from positive pressure ventilation. However, injurious forces also can be generated by patient effort and patient–ventilator interactions. While the role of global mechanics has long been recognized, regional mechanical heterogeneity within the lungs also appears to be an important factor propagating clinically significant lung injury. The resulting clinical phenotype includes worsening lung injury and a systemic inflammatory response that drives extrapulmonary organ failures. Bedside recognition of ventilation-induced lung injury requires a high degree of clinical acuity given its indistinct presentation and lack of definitive diagnostics. Yet the clinical importance of ventilation-induced lung injury is clear. Preventing such biophysical injury remains the most effective management strategy to decrease morbidity and mortality in patients with acute respiratory distress syndrome and likely benefits others at risk.
Collapse
Affiliation(s)
- Purnema Madahar
- Center for Acute Respiratory Failure, Columbia University College of Physicians and Surgeons, New York City, NY, USA.,Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University College of Physicians and Surgeons, New York City, NY, USA.,Department of Medicine, New York-Presbyterian Hospital, New York City, NY, USA
| | - Jeremy R Beitler
- Center for Acute Respiratory Failure, Columbia University College of Physicians and Surgeons, New York City, NY, USA.,Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University College of Physicians and Surgeons, New York City, NY, USA.,Department of Medicine, New York-Presbyterian Hospital, New York City, NY, USA
| |
Collapse
|
86
|
Abstract
PURPOSE OF REVIEW To provide an overview of the current research in identifying homogeneous subgroups and phenotypes in ARDS. RECENT FINDINGS In recent years, investigations have used either physiology, clinical data, biomarkers or a combination of these to stratify patients with ARDS into distinct subgroups with divergent clinical outcomes. In some studies, there has also been evidence of differential treatment response within subgroups. Physiologic approaches include stratification based on P/F ratio and ventilatory parameters; stratification based on P/F ratio is already being employed in clinical trials. Clinical approaches include stratification based on ARDS risk factor or direct vs. indirect ARDS. Combined clinical and biological data has been used to identify two phenotypes across five cohorts of ARDS, termed hyperinflammatory and hypoinflammatory. These phenotypes have widely divergent clinical outcomes and differential response to mechanical ventilation, fluid therapy, and simvastatin in secondary analysis of completed trials. Next steps in the field include prospective validation of inflammatory phenotypes and integration of high-dimensional 'omics' data into our understanding of ARDS heterogeneity. SUMMARY Identification of distinct subgroups or phenotypes in ARDS may impact future conduct of clinical trials and can enhance our understanding of the disorder, with potential future clinical implications.
Collapse
|
87
|
Qian Q, Cao X, Wang B, Dong X, Pei J, Xue L, Feng F. Endoplasmic reticulum stress potentiates the autophagy of alveolar macrophage to attenuate acute lung injury and airway inflammation. Cell Cycle 2020; 19:567-576. [PMID: 32057287 PMCID: PMC7100984 DOI: 10.1080/15384101.2020.1718851] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Endoplasmic reticulum (ER) stress has been reported to play a role in acute lung injury (ALI), yet the in-depth mechanism remains elusive. This study aims to investigate the effect of ER stress-induced autophagy of alveolar macrophage (AM) on acute lung injury (ALI) and airway inflammation using mouse models. ALI models were induced by intranasal instillation of lipopolysaccharide (LPS). The lung weight/body weight (LW/BW) ratio and excised lung gas volume (ELGV) in each group were measured. Mouse bronchoalveolar lavage fluid (BALF) was collected for cell sorting and protein concentration determination. Expression of tumor necrosis factor α (TNF-α) and interleukin-6 (IL-6) in lung tissues and BALF was also detected. Mouse AMs were isolated to observe the autophagy. Expression of GRP78, PERK, LC3I, LC3II and Beclin1 was further determined. The results indicated that tunicamycin (TM) elevated GRP78 and PERK expression of AMs in ALI mice in a dose-dependent manner. Low dosage of TM abated LC3I expression, increased LC3II and Beclin1 expression, triggered ER stress and AM autophagy, and alleviated pathological changes of AMs in ALI mice. Also, in ALI mice, low dosage of TM attenuated goblet cell proliferation of tracheal wall, and declined LW/BW ratio, ELGV, total cells and neutrophils, protein concentrations in BALF, and IL-6 and TNF-α expression in lung tissues and BALF. Collectively, this study suggests that a low dosage of TM-induced ER stress can enhance the autophagy of AM in ALI mice models, thus attenuating the progression of ALI and airway inflammation.
Collapse
Affiliation(s)
- Qingzeng Qian
- College of Public Health, North China University of Science and Technology, Tangshan, P. R. China
| | - Xiangke Cao
- College of Life Sciences, North China University of Science and Technology, Tangshan, P. R. China
| | - Bin Wang
- Department of Pediatrics, Affiliated Hospital of North China University of Science and Technology, Tangshan, P. R. China
| | - Xiaoliu Dong
- Department of Neurology, Tangshan People's Hospital, Tangshan, P. R. China
| | - Jian Pei
- Department of Neurosurgery, Tangshan Worker's Hospital, Tangshan, P. R. China
| | - Ling Xue
- College of Public Health, North China University of Science and Technology, Tangshan, P. R. China
| | - Fumin Feng
- College of Public Health, North China University of Science and Technology, Tangshan, P. R. China
| |
Collapse
|
88
|
Liu S, Wang C, Green G, Zhuo H, Liu KD, Kangelaris KN, Gomez A, Jauregui A, Vessel K, Ke S, Hendrickson C, Matthay MA, Calfee CS, Ware LB, Wolters PJ. Peripheral blood leukocyte telomere length is associated with survival of sepsis patients. Eur Respir J 2020; 55:13993003.01044-2019. [PMID: 31619475 DOI: 10.1183/13993003.01044-2019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 09/21/2019] [Indexed: 12/12/2022]
Abstract
Shorter peripheral blood leukocyte (PBL) telomere length (TL) has been associated with poor outcomes in various chronic lung diseases. Whether PBL-TL is associated with survival from critical illness was tested in this study.We analysed data from a prospective observational cohort study of 937 critically ill patients at Vanderbilt University Medical Center (VUMC). PBL-TL was measured using quantitative PCR of DNA isolated from PBLs. Findings were validated in an independent cohort of 394 critically ill patients with sepsis admitted to the University of California San Francisco (UCSF).In the VUMC cohort, shorter PBL-TL was associated with worse 90-day survival (adjusted hazard ratio (aHR) 1.3, 95% CI 1.1-1.6 per 1 kb TL decrease; p=0.004); in subgroup analyses, shorter PBL-TL was associated with worse 90-day survival for patients with sepsis (aHR 1.5, 95% CI 1.2-2.0 per 1 kb TL decrease; p=0.001), but not trauma. Although not associated with development of acute respiratory distress syndrome (ARDS), among ARDS subjects, shorter PBL-TL was associated with more severe ARDS (OR 1.7, 95% CI 1.2-2.5 per 1 kb TL decrease; p=0.006). The associations of PBL-TL with survival (adjusted HR 1.6, 95% CI 1.2-2.1 per 1 kb TL decrease; p=0.003) and risk for developing severe ARDS (OR 2.5, 95% CI 1.1-6.3 per 1 kb TL decrease; p=0.044) were validated in the UCSF cohort.Short PBL-TL is strongly associated with worse survival and more severe ARDS in critically ill patients, especially patients with sepsis. These findings suggest that telomere dysfunction may contribute to outcomes from critical illness.
Collapse
Affiliation(s)
- Shuo Liu
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Dept of Medicine, University of California, San Francisco, San Francisco, CA, USA.,Dept of Respiratory Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.,Dept of Respiratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Chunxue Wang
- Dept of Medicine and Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Gary Green
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Dept of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Hanjing Zhuo
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Dept of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Kathleen D Liu
- Division of Nephrology, Dept of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Kirsten N Kangelaris
- Division of Hospital Medicine, Dept of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Antonio Gomez
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Dept of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Alejandra Jauregui
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Dept of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Kathryn Vessel
- The Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Serena Ke
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Dept of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Carolyn Hendrickson
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Dept of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Michael A Matthay
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Dept of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Carolyn S Calfee
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Dept of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Lorraine B Ware
- Dept of Medicine and Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Paul J Wolters
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Dept of Medicine, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
89
|
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]
|
90
|
Juliana A, Zonneveld R, Plötz FB, van Meurs M, Wilschut J. Neutrophil-endothelial interactions in respiratory syncytial virus bronchiolitis: An understudied aspect with a potential for prediction of severity of disease. J Clin Virol 2019; 123:104258. [PMID: 31931445 DOI: 10.1016/j.jcv.2019.104258] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/25/2019] [Accepted: 12/30/2019] [Indexed: 01/25/2023]
Abstract
Respiratory syncytial virus (RSV) lower respiratory tract infection (LRTI) causes significant morbidity and mortality among young infants worldwide. It is currently widely accepted that neutrophil influx into the airways is a hallmark of the pathophysiology. However, the exact mechanism of neutrophil migration from the vasculature into the alveolar space in RSV LRTI has received little attention. Data shows that endothelial cells become activated upon RSV infection, driving a 'pro-adhesive state' for circulating neutrophils with upregulation of endothelial intercellular adhesion molecule-1 (ICAM-1). During RSV LRTI different subsets of immature and mature neutrophils are present in the bloodstream, that upregulate integrins lymphocyte-function associated antigen (LFA)-1 and macrophage (Mac)-1, serving as ICAM-1 ligands. An alveolar gradient of interleukin-8 may serve as a potent chemoattractant for circulating neutrophils. Neutrophils from lung aspirates of RSV-infected infants show further signs of inflammatory and migratory activation, while soluble endothelial cell adhesion molecules (sCAMs), such as sICAM-1, have become measurable in the systemic circulation. Whether these mechanisms are solely responsible for neutrophil migration into the alveolar space remains under debate. However, data indicate that the currently postulated neutrophil influx into the lungs should rather be regarded as a neutrophil efflux from the vasculature, involving substantial neutrophil-endothelial interactions. Molecular patterns of these interactions may be clinically useful to predict outcomes of RSV LRTI and deserve further study.
Collapse
Affiliation(s)
- Amadu Juliana
- Academic Pediatric Center Suriname, Academic Hospital Paramaribo, Paramaribo, Suriname.
| | - Rens Zonneveld
- Department of Microbiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Frans B Plötz
- Department of Pediatrics, Tergooi Hospitals, Blaricum, The Netherlands
| | - Matijs van Meurs
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan Wilschut
- Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
91
|
Anderson BJ, Calfee CS, Liu KD, Reilly JP, Kangelaris KN, Shashaty MGS, Lazaar AL, Bayliffe AI, Gallop RJ, Miano TA, Dunn TG, Johansson E, Abbott J, Jauregui A, Deiss T, Vessel K, Belzer A, Zhuo H, Matthay MA, Meyer NJ, Christie JD. Plasma sTNFR1 and IL8 for prognostic enrichment in sepsis trials: a prospective cohort study. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2019; 23:400. [PMID: 31818332 PMCID: PMC6902425 DOI: 10.1186/s13054-019-2684-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/22/2019] [Indexed: 01/07/2023]
Abstract
Background Enrichment strategies improve therapeutic targeting and trial efficiency, but enrichment factors for sepsis trials are lacking. We determined whether concentrations of soluble tumor necrosis factor receptor-1 (sTNFR1), interleukin-8 (IL8), and angiopoietin-2 (Ang2) could identify sepsis patients at higher mortality risk and serve as prognostic enrichment factors. Methods In a multicenter prospective cohort study of 400 critically ill septic patients, we derived and validated thresholds for each marker and expressed prognostic enrichment using risk differences (RD) of 30-day mortality as predictive values. We then used decision curve analysis to simulate the prognostic enrichment of each marker and compare different prognostic enrichment strategies. Measurements and main results An admission sTNFR1 concentration > 8861 pg/ml identified patients with increased mortality in both the derivation (RD 21.6%) and validation (RD 17.8%) populations. Among immunocompetent patients, an IL8 concentration > 94 pg/ml identified patients with increased mortality in both the derivation (RD 17.7%) and validation (RD 27.0%) populations. An Ang2 level > 9761 pg/ml identified patients at 21.3% and 12.3% increased risk of mortality in the derivation and validation populations, respectively. Using sTNFR1 or IL8 to select high-risk patients improved clinical trial power and efficiency compared to selecting patients with septic shock. Ang2 did not outperform septic shock as an enrichment factor. Conclusions Thresholds for sTNFR1 and IL8 consistently identified sepsis patients with higher mortality risk and may have utility for prognostic enrichment in sepsis trials.
Collapse
Affiliation(s)
- Brian J Anderson
- Division of Pulmonary, Allergy and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, 5036 Gates Building, Philadelphia, PA, 19104, USA.
| | - Carolyn S Calfee
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco, USA
| | - Kathleen D Liu
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco, USA
| | - John P Reilly
- Division of Pulmonary, Allergy and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, 5036 Gates Building, Philadelphia, PA, 19104, USA
| | - Kirsten N Kangelaris
- Division of Hospital Medicine, Department of Medicine, University of California San Francisco, San Francisco, USA
| | - Michael G S Shashaty
- Division of Pulmonary, Allergy and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, 5036 Gates Building, Philadelphia, PA, 19104, USA.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Aili L Lazaar
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.,GlaxoSmithKline R&D, Brentford, UK
| | | | - Robert J Gallop
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA.,Department of Mathematics, West Chester University, West Chester, USA
| | - Todd A Miano
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Thomas G Dunn
- Division of Pulmonary, Allergy and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, 5036 Gates Building, Philadelphia, PA, 19104, USA
| | - Erik Johansson
- Division of Pulmonary, Allergy and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, 5036 Gates Building, Philadelphia, PA, 19104, USA
| | - Jason Abbott
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco, USA
| | - Alejandra Jauregui
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco, USA
| | - Thomas Deiss
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco, USA
| | - Kathryn Vessel
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco, USA
| | - Annika Belzer
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco, USA
| | - Hanjing Zhuo
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco, USA
| | - Michael A Matthay
- Division of Pulmonary and Critical Care Medicine, University of California San Francisco, San Francisco, USA
| | - Nuala J Meyer
- Division of Pulmonary, Allergy and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, 5036 Gates Building, Philadelphia, PA, 19104, USA
| | - Jason D Christie
- Division of Pulmonary, Allergy and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, 5036 Gates Building, Philadelphia, PA, 19104, USA.,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
92
|
Shirali AS, Lluri G, Guihard PJ, Conrad MB, Kim H, Pawlikowska L, Boström KI, Iruela-Arispe ML, Aboulhosn JA. Angiopoietin-2 predicts morbidity in adults with Fontan physiology. Sci Rep 2019; 9:18328. [PMID: 31797976 PMCID: PMC6892891 DOI: 10.1038/s41598-019-54776-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 11/14/2019] [Indexed: 12/14/2022] Open
Abstract
Morbidity in patients with single-ventricle Fontan circulation is common and includes arrhythmias, edema, and pulmonary arteriovenous malformations (PAVM) among others. We sought to identify biomarkers that may predict such complications. Twenty-five patients with Fontan physiology and 12 control patients with atrial septal defects (ASD) that underwent cardiac catheterization were included. Plasma was collected from the hepatic vein and superior vena cava and underwent protein profiling for a panel of 20 analytes involved in angiogenesis and endothelial dysfunction. Ten (40%) of Fontan patients had evidence of PAVM, eighteen (72%) had a history of arrhythmia, and five (20%) were actively in arrhythmia or had a recent arrhythmia. Angiopoietin-2 (Ang-2) was higher in Fontan patients (8,875.4 ± 3,336.9 pg/mL) versus the ASD group (1,663.6 ± 587.3 pg/mL, p < 0.0001). Ang-2 was higher in Fontan patients with active or recent arrhythmia (11,396.0 ± 3,457.7 vs 8,118.2 ± 2,795.1 pg/mL, p < 0.05). A threshold of 8,500 pg/mL gives Ang-2 a negative predictive value of 100% and positive predictive value of 42% in diagnosing recent arrhythmia. Ang-2 is elevated among adults with Fontan physiology. Ang-2 level is associated with active or recent arrhythmia, but was not found to be associated with PAVM.
Collapse
Affiliation(s)
- Aditya S Shirali
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, USA
| | - Gentian Lluri
- Ahmanson/UCLA Adult Congenital Heart Disease Center, Division of Cardiology, University of California Los Angeles, Los Angeles, CA, USA.
| | - Pierre J Guihard
- Division of Cardiology, University of California Los Angeles, Los Angeles, CA, USA
| | - Miles B Conrad
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Helen Kim
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA.,Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Ludmila Pawlikowska
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA, USA.,Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Kristina I Boström
- Division of Cardiology, University of California Los Angeles, Los Angeles, CA, USA
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell & Developmental Biology, Molecular Biology Institute and Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
| | - Jamil A Aboulhosn
- Ahmanson/UCLA Adult Congenital Heart Disease Center, Division of Cardiology, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
93
|
Pham TT, Punsawad C, Glaharn S, De Meyer SF, Viriyavejakul P, Van den Steen PE. Release of endothelial activation markers in lungs of patients with malaria-associated acute respiratory distress syndrome. Malar J 2019; 18:395. [PMID: 31796023 PMCID: PMC6891978 DOI: 10.1186/s12936-019-3040-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/26/2019] [Indexed: 11/17/2022] Open
Abstract
Background Malaria-associated acute respiratory distress syndrome (MA-ARDS) is an understudied complication of malaria and is characterized by pulmonary inflammation and disruption of the alveolar-capillary membrane. Its pathogenesis remains poorly understood. Since endothelial activation plays an important role in other malarial complications, the expression of two endothelial activation markers, von Willebrand factor (VWF) and angiopoietin-2 (ANG-2), was investigated in the lungs of patients with MA-ARDS. Methods Post-mortem lung sections of Plasmodium falciparum-infected patients without alveolar oedema (NA), P. falciparum-infected patients with alveolar oedema (MA-ARDS), and uninfected people who died accidentally with no pathological changes to the lungs (CON) were immunohistochemically stained for VWF and ANG-2, and were evaluated with semi-quantitative analysis. Results Alveolar oedematous VWF and ANG-2 and intravascular VWF staining were significantly increased in patients with MA-ARDS versus infected and uninfected control groups. The levels of VWF in the alveolar septa and endothelial lining of large blood vessels of patients with MA-ARDS was significantly decreased compared to controls. ANG-2 expression was increased in the alveolar septa of malaria patients without alveolar oedema versus control patients, while ANG-2+ leukocytes were increased in the alveoli in both infected patient groups. Conclusions This study documents a high level of VWF and ANG-2, two endothelial activation markers in the oedematous alveoli of post-mortem lung sections of Thai patients with MA-ARDS. Decreased detection of VWF in the endothelial lining of blood vessels, in parallel with an increased presence of intravascular VWF staining suggests marked endothelial activation and Weibel–Palade body release in the lungs of patients with MA-ARDS.
Collapse
Affiliation(s)
- Thao-Thy Pham
- Laboratory of Immunoparasitology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Leuven, Belgium
| | - Chuchard Punsawad
- School of Medicine, Walailak University, Nakhon Si Thammarat, Thailand
| | - Supattra Glaharn
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Simon F De Meyer
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Parnpen Viriyavejakul
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Philippe E Van den Steen
- Laboratory of Immunoparasitology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Leuven, Belgium.
| |
Collapse
|
94
|
Erice C, Kain KC. New insights into microvascular injury to inform enhanced diagnostics and therapeutics for severe malaria. Virulence 2019; 10:1034-1046. [PMID: 31775570 PMCID: PMC6930010 DOI: 10.1080/21505594.2019.1696621] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/05/2019] [Accepted: 11/13/2019] [Indexed: 12/25/2022] Open
Abstract
Severe malaria (SM) has high mortality and morbidity rates despite treatment with potent antimalarials. Disease onset and outcome is dependent upon both parasite and host factors. Infected erythrocytes bind to host endothelium contributing to microvascular occlusion and dysregulated inflammatory and immune host responses, resulting in endothelial activation and microvascular damage. This review focuses on the mechanisms of host endothelial and microvascular injury. Only a small percentage of malaria infections (≤1%) progress to SM. Early recognition and treatment of SM can improve outcome, but we lack triage tools to identify SM early in the course of infection. Current point-of-care pathogen-based rapid diagnostic tests do not address this critical barrier. Immune and endothelial activation have been implicated in the pathobiology of SM. We hypothesize that measuring circulating mediators of these pathways at first clinical presentation will enable early triage and treatment of SM. Moreover, that host-based interventions that modulate these pathways will stabilize the microvasculature and improve clinical outcome over that of antimalarial therapy alone.
Collapse
Affiliation(s)
- Clara Erice
- Sandra-Rotman Centre for Global Health, Toronto General Research Institute, University Health Network-Toronto General Hospital, Toronto, Ontario, Canada
| | - Kevin C Kain
- Sandra-Rotman Centre for Global Health, Toronto General Research Institute, University Health Network-Toronto General Hospital, Toronto, Ontario, Canada
- Tropical Disease Unit, Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
95
|
Jagrosse ML, Dean DA, Rahman A, Nilsson BL. RNAi therapeutic strategies for acute respiratory distress syndrome. Transl Res 2019; 214:30-49. [PMID: 31401266 PMCID: PMC7316156 DOI: 10.1016/j.trsl.2019.07.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 12/11/2022]
Abstract
Acute respiratory distress syndrome (ARDS), replacing the clinical term acute lung injury, involves serious pathophysiological lung changes that arise from a variety of pulmonary and nonpulmonary injuries and currently has no pharmacological therapeutics. RNA interference (RNAi) has the potential to generate therapeutic effects that would increase patient survival rates from this condition. It is the purpose of this review to discuss potential targets in treating ARDS with RNAi strategies, as well as to outline the challenges of oligonucleotide delivery to the lung and tactics to circumvent these delivery barriers.
Collapse
Affiliation(s)
| | - David A Dean
- Department of Pediatrics and Neonatology, University of Rochester Medical Center, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Arshad Rahman
- Department of Pediatrics and Neonatology, University of Rochester Medical Center, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Bradley L Nilsson
- Department of Chemistry, University of Rochester, Rochester, New York.
| |
Collapse
|
96
|
Kida Y, Ohshimo S, Kyo M, Hosokawa K, Amatya VJ, Takeshima Y, Shime N. Retrospective immunohistological study of autopsied lungs in patients with acute exacerbation of interstitial pneumonia managed with extracorporeal membrane oxygenation. J Thorac Dis 2019; 11:4436-4443. [PMID: 31903231 DOI: 10.21037/jtd.2019.11.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Acute exacerbation of interstitial pneumonia (AE-IP) is a life-threatening pulmonary condition that involves various pathogeneses. In patients with AE-IP who need mechanical ventilation with high driving pressure and oxygen concentration, veno-venous extracorporeal membrane oxygenation (V-V ECMO) may diminish alveolar epithelial damage by decreasing ventilator settings. The pathophysiological benefit of this therapeutic option is not well investigated. Methods We retrospectively collected 15 autopsied patients with AE-IP who were treated with mechanical ventilation in the intensive care unit (ICU) at Hiroshima University Hospital (Hiroshima, Japan) between 2010 and 2016. The patients were grouped by whether they were managed with mechanical ventilation only (the ventilator group, n=6) or with mechanical ventilation and V-V ECMO (the ECMO group, n=9). Results The median age of the ventilator and ECMO group patients were similar (65 and 64 years, respectively). The severity score APACHE II in the ECMO group (35.0) is significantly higher than that of ventilator group (14.5) (P=0.006). Ventilator days were significantly shorter in the ventilator group (17.5 days) than in the ECMO group (30.0 days) (P=0.04). Compared with the ECMO group, the ventilator group had a stronger Masson-trichrome stain grade (4 vs. 6, P=0.04) and higher immunoreactivity grades for Krebs von den Lungen-6 (4 vs. 6, P=0.04) and IL-8 (3 vs. 6, P=0.02). Between the ventilator and ECMO groups, the immunoreactivity grades of angiopoietin 2 (4 vs. 1, P=0.08) and receptor for advanced glycation end products (2 vs. 1, P=0.52) did not differ. Conclusions The lungs of mechanically ventilated AE-IP patients treated with V-V ECMO had decreased fibrosis, endothelial injury, and inflammation. This finding suggests the lung-protective efficacy of adjunctive V-V ECMO therapy.
Collapse
Affiliation(s)
- Yoshiko Kida
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shinichiro Ohshimo
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Michihito Kyo
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Koji Hosokawa
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Vishwa Jeet Amatya
- Department of Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yukio Takeshima
- Department of Pathology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Nobuaki Shime
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
97
|
Carlton EF, Flori HR. Biomarkers in pediatric acute respiratory distress syndrome. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:505. [PMID: 31728358 DOI: 10.21037/atm.2019.09.29] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Pediatric acute respiratory distress syndrome (PARDS) is a heterogenous process resulting in a severe acute lung injury. A single indicator does not exist for PARDS diagnosis. Rather, current diagnosis requires a combination of clinical and physiologic variables. Similarly, there is little ability to predict the path of disease, identify those at high risk of poor outcomes or target therapies specific to the underlying pathophysiology. Biomarkers, a measured indicator of a pathologic state or response to intervention, have been studied in PARDS due to their potential in diagnosis, prognostication and measurement of therapeutic response. Additionally, PARDS biomarkers show great promise in furthering our understanding of specific subgroups or endotypes in this highly variable disease, and thereby predict which patients may benefit and which may be harmed by PARDS specific therapies. In this chapter, we review the what, when, why and how of biomarkers in PARDS and discuss future directions in this quickly changing landscape.
Collapse
Affiliation(s)
- Erin F Carlton
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| | - Heidi R Flori
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
98
|
Gutbier B, Neuhauß AK, Reppe K, Ehrler C, Santel A, Kaufmann J, Scholz M, Weissmann N, Morawietz L, Mitchell TJ, Aliberti S, Hippenstiel S, Suttorp N, Witzenrath M. Prognostic and Pathogenic Role of Angiopoietin-1 and -2 in Pneumonia. Am J Respir Crit Care Med 2019; 198:220-231. [PMID: 29447449 DOI: 10.1164/rccm.201708-1733oc] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
RATIONALE During pneumonia, pathogen-host interaction evokes inflammation and lung barrier dysfunction. Tie2 activation by angiopoietin-1 reduces, whereas Tie2 blockade by angiopoietin-2 increases, inflammation and permeability during sepsis. The role of angiopoietin-1/-2 in pneumonia remains unidentified. OBJECTIVES To investigate the prognostic and pathogenic impact of angiopoietins in regulating pulmonary vascular barrier function and inflammation in bacterial pneumonia. METHODS Serum angiopoietin levels were quantified in pneumonia patients of two independent cohorts (n = 148, n = 395). Human postmortem lung tissue, pneumolysin- or angiopoietin-2-stimulated endothelial cells, isolated perfused and ventilated mouse lungs, and mice with pneumococcal pneumonia were investigated. MEASUREMENTS AND MAIN RESULTS In patients with pneumonia, decreased serum angiopoietin-1 and increased angiopoietin-2 levels were observed as compared with healthy subjects. Higher angiopoietin-2 serum levels were found in patients with community-acquired pneumonia who died within 28 days of diagnosis compared with survivors. Receiver operating characteristic analysis revealed improved prognostic accuracy of CURB-65 for 28-day survival, intensive care treatment, and length of hospital stay if combined with angiopoietin-2 serum levels. In vitro, pneumolysin enhanced endothelial angiopoietin-2 release, angiopoietin-2 increased endothelial permeability, and angiopoietin-1 reduced pneumolysin-evoked endothelial permeability. Ventilated and perfused lungs of mice with angiopoietin-2 knockdown showed reduced permeability on pneumolysin stimulation. Increased pulmonary angiopoietin-2 and reduced angiopoietin-1 mRNA expression were observed in Streptococcus pneumoniae-infected mice. Finally, angiopoietin-1 therapy reduced inflammation and permeability in murine pneumonia. CONCLUSIONS These data suggest a central role of angiopoietin-1/-2 in pneumonia-evoked inflammation and permeability. Increased angiopoietin-2 serum levels predicted mortality and length of hospital stay, and angiopoietin-1 may provide a therapeutic target for severe pneumonia.
Collapse
Affiliation(s)
- Birgitt Gutbier
- 1 Division of Pulmonary Inflammation and.,2 Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Anne-Kathrin Neuhauß
- 2 Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Katrin Reppe
- 1 Division of Pulmonary Inflammation and.,2 Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Carolin Ehrler
- 2 Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | | | | | - Markus Scholz
- 4 Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Norbert Weissmann
- 5 Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Justus-Liebig-University, Giessen, Germany
| | - Lars Morawietz
- 6 Pathology, Healthcare Center Fuerstenberg-Karree, Berlin, Germany
| | - Timothy J Mitchell
- 7 Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Stefano Aliberti
- 8 Department of Pathophysiology and Transplantation, University of Milan, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Cà Granda Ospedale Maggiore Policlinico, Milan, Italy; and
| | - Stefan Hippenstiel
- 2 Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Norbert Suttorp
- 2 Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,9 CAPNETZ STIFTUNG, Hannover, Germany
| | - Martin Witzenrath
- 1 Division of Pulmonary Inflammation and.,2 Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,9 CAPNETZ STIFTUNG, Hannover, Germany
| | | |
Collapse
|
99
|
Plasma Angiopoietin-2/-1 Ratio is Elevated and Angiopoietin-2 Levels Correlate With Plasma Syndecan-1 Following Pediatric Trauma. Shock 2019; 52:340-346. [DOI: 10.1097/shk.0000000000001267] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
100
|
Torres A. New Insights into the Regulation of Endothelial Lung Permeability in Pneumonia. The Interplay between Angiopoietins 1 and 2. Am J Respir Crit Care Med 2019. [PMID: 29529378 DOI: 10.1164/rccm.201802-0377ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Antoni Torres
- 1 Hospital Clinic Universitat de Barcelona Barcelona, Spain and.,2 Icrea Academia Institut d'Investigacions Biomèdiques August Pi i Sunyer Barcelona, Spain
| |
Collapse
|