1
|
Chen Z, Lu J, Liu G, Liu C, Wu S, Xian L, Zhou X, Zuo L, Su Y. COMPREHENSIVE CHARACTERIZATION OF CYTOKINES IN PATIENTS UNDER EXTRACORPOREAL MEMBRANE OXYGENATION: EVIDENCE FROM INTEGRATED BULK AND SINGLE-CELL RNA SEQUENCING DATA USING MULTIPLE MACHINE LEARNING APPROACHES. Shock 2025; 63:267-281. [PMID: 39503329 PMCID: PMC11776881 DOI: 10.1097/shk.0000000000002425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/22/2024] [Indexed: 11/08/2024]
Abstract
ABSTRACT Background : Extracorporeal membrane oxygenation (ECMO) is an effective technique for providing short-term mechanical support to the heart, lungs, or both. During ECMO treatment, the inflammatory response, particularly involving cytokines, plays a crucial role in pathophysiology. However, the potential effects of cytokines on patients receiving ECMO are not comprehensively understood. Methods : We acquired three ECMO support datasets, namely two bulk and one single-cell RNA sequencing (RNA-seq), from the Gene Expression Omnibus (GEO) combined with hospital cohorts to investigate the expression pattern and potential biological processes of cytokine-related genes (CRGs) in patients under ECMO. Subsequently, machine learning approaches, including support vector machine (SVM), random forest (RF), modified Lasso penalized regression, extreme gradient boosting (XGBoost), and artificial neural network (ANN), were applied to identify hub CRGs, thus developing a prediction model called CRG classifier. The predictive and prognostic performance of the model was comprehensively evaluated in GEO and hospital cohorts. Finally, we mechanistically analyzed the relationship between hub cytokines, immune cells, and pivotal molecular pathways. Results : Analyzing bulk and single-cell RNA-seq data revealed that most CRGs were significantly differentially expressed; the enrichment scores of cytokine and cytokine-cytokine receptor (CCR) interaction were significantly higher during ECMO. Based on multiple machine learning algorithms, nine key CRGs (CCL2, CCL4, IFNG, IL1R2, IL20RB, IL31RA, IL4, IL7, and IL7R) were used to develop the CRG classifier. The CRG classifier exhibited excellent prognostic values (AUC > 0.85), serving as an independent risk factor. It performed better in predicting mortality and yielded a larger net benefit than other clinical features in GEO and hospital cohorts. Additionally, IL1R2, CCL4, and IL7R were predominantly expressed in monocytes, NK cells, and T cells, respectively. Their expression was significantly positively correlated with the relative abundance of corresponding immune cells. Gene set variation analysis (GSVA) revealed that para-inflammation, complement and coagulation cascades, and IL6/JAK/STAT3 signaling were significantly enriched in the subgroup that died after receiving ECMO. Spearman correlation analyses and Mantel tests revealed that the expression of hub cytokines (IL1R2, CCL4, and IL7R) and pivotal molecular pathways scores (complement and coagulation cascades, IL6/JAK/STAT3 signaling, and para-inflammation) were closely related. Conclusion : A predictive model (CRG classifier) comprising nine CRGs based on multiple machine learning algorithms was constructed, potentially assisting clinicians in guiding individualized ECMO treatment. Additionally, elucidating the underlying mechanistic pathways of cytokines during ECMO will provide new insights into its treatment.
Collapse
Affiliation(s)
- Zhen Chen
- Department of Intensive Care Unit, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong Province, PR China
| | - Jianhai Lu
- Department of Intensive Care Unit, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong Province, PR China
| | - Genglong Liu
- School of Medicine, Southern Medical University, Foshan, Guangdong Province, PR China
- Editor Office, iMeta, Shenzhen, Guangdong Province, PR China
| | - Changzhi Liu
- Department of Intensive Care Unit, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong Province, PR China
| | - Shumin Wu
- Department of Department of Clinical Pharmacy, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong Province, PR China
| | - Lina Xian
- Department of Intensive Care Unit, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, PR China
| | - Xingliang Zhou
- Department of Intensive Care Unit, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong Province, PR China
| | - Liuer Zuo
- Department of Intensive Care Unit, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong Province, PR China
| | - Yongpeng Su
- Department of Intensive Care Unit, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong Province, PR China
| |
Collapse
|
2
|
Paukner S, Kimber S, Cumper C, Rea-Davies T, Sueiro Ballesteros L, Kirkham C, Hargreaves A, Gelone SP, Richards C, Wicha WW. In Vivo Immune-Modulatory Activity of Lefamulin in an Influenza Virus A (H1N1) Infection Model in Mice. Int J Mol Sci 2024; 25:5401. [PMID: 38791439 PMCID: PMC11121702 DOI: 10.3390/ijms25105401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Lefamulin is a first-in-class systemic pleuromutilin antimicrobial and potent inhibitor of bacterial translation, and the most recent novel antimicrobial approved for the treatment of community-acquired pneumonia (CAP). It exhibits potent antibacterial activity against the most prevalent bacterial pathogens that cause typical and atypical pneumonia and other infectious diseases. Early studies indicate additional anti-inflammatory activity. In this study, we further investigated the immune-modulatory activity of lefamulin in the influenza A/H1N1 acute respiratory distress syndrome (ARDS) model in BALB/c mice. Comparators included azithromycin, an anti-inflammatory antimicrobial, and the antiviral oseltamivir. Lefamulin significantly decreased the total immune cell infiltration, specifically the neutrophils, inflammatory monocytes, CD4+ and CD8+ T-cells, NK cells, and B-cells into the lung by Day 6 at both doses tested compared to the untreated vehicle control group (placebo), whereas azithromycin and oseltamivir did not significantly affect the total immune cell counts at the tested dosing regimens. Bronchioalveolar lavage fluid concentrations of pro-inflammatory cytokines and chemokines including TNF-α, IL-6, IL-12p70, IL-17A, IFN-γ, and GM-CSF were significantly reduced, and MCP-1 concentrations were lowered (not significantly) by lefamulin at the clinically relevant 'low' dose on Day 3 when the viral load peaked. Similar effects were also observed for oseltamivir and azithromycin. Lefamulin also decreased the viral load (TCID50) by half a log10 by Day 6 and showed positive effects on the gross lung pathology and survival. Oseltamivir and lefamulin were efficacious in the suppression of the development of influenza-induced bronchi-interstitial pneumonia, whereas azithromycin did not show reduced pathology at the tested treatment regimen. The observed anti-inflammatory and immune-modulatory activity of lefamulin at the tested treatment regimens highlights a promising secondary pharmacological property of lefamulin. While these results require confirmation in a clinical trial, they indicate that lefamulin may provide an immune-modulatory activity beyond its proven potent antibacterial activity. This additional activity may benefit CAP patients and potentially prevent acute lung injury (ALI) and ARDS.
Collapse
Affiliation(s)
- Susanne Paukner
- Nabriva Therapeutics GmbH, Leberstrasse 20, 1110 Vienna, Austria;
| | - Sandra Kimber
- Charles River Discovery, Portishead BS20 7AW, UK; (S.K.); (C.C.); (T.R.-D.); (L.S.B.); (C.K.); (C.R.)
| | - Charlotte Cumper
- Charles River Discovery, Portishead BS20 7AW, UK; (S.K.); (C.C.); (T.R.-D.); (L.S.B.); (C.K.); (C.R.)
| | - Tina Rea-Davies
- Charles River Discovery, Portishead BS20 7AW, UK; (S.K.); (C.C.); (T.R.-D.); (L.S.B.); (C.K.); (C.R.)
| | - Lorena Sueiro Ballesteros
- Charles River Discovery, Portishead BS20 7AW, UK; (S.K.); (C.C.); (T.R.-D.); (L.S.B.); (C.K.); (C.R.)
| | - Christopher Kirkham
- Charles River Discovery, Portishead BS20 7AW, UK; (S.K.); (C.C.); (T.R.-D.); (L.S.B.); (C.K.); (C.R.)
| | | | | | - Claire Richards
- Charles River Discovery, Portishead BS20 7AW, UK; (S.K.); (C.C.); (T.R.-D.); (L.S.B.); (C.K.); (C.R.)
| | | |
Collapse
|
3
|
Saki N, Javan M, Moghimian-Boroujeni B, Kast RE. Interesting effects of interleukins and immune cells on acute respiratory distress syndrome. Clin Exp Med 2023; 23:2979-2996. [PMID: 37330918 DOI: 10.1007/s10238-023-01118-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 06/10/2023] [Indexed: 06/20/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is a medical condition characterized by widespread inflammation in the lungs with consequent proportional loss of gas exchange function. ARDS is linked with severe pulmonary or systemic infection. Several factors, including secretory cytokines, immune cells, and lung epithelial and endothelial cells, play a role in the development and progression of this disease. The present study is based on Pubmed database information (1987-2022) using the words "Acute respiratory distress syndrome", "Interleukin", "Cytokines" and "Immune cells". Cytokines and immune cells play an important role in this disease, with particular emphasis on the balance between pro-inflammatory and anti-inflammatory factors. Neutrophils are one of several important mediators of Inflammation, lung tissue destruction, and malfunction during ARDS. Some immune cells, such as macrophages and eosinophils, play a dual role in releasing inflammatory mediators, recruitment inflammatory cells and the progression of ARDS, or releasing anti-inflammatory mediators, clearing the lung of inflammatory cells, and helping to improve the disease. Different interleukins play a role in the development or inhibition of ARDS by helping to activate various signaling pathways, helping to secrete other inflammatory or anti-inflammatory interleukins, and playing a role in the production and balance between immune cells involved in ARDS. As a result, immune cells and, inflammatory cytokines, especially interleukins play an important role in the pathogenesis of this disease Therefore, understanding the relevant mechanisms will help in the proper diagnosis and treatment of this disease.
Collapse
Affiliation(s)
- Najmaldin Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammadreza Javan
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran
| | - Bahareh Moghimian-Boroujeni
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, 61357-15794, Iran.
| | | |
Collapse
|
4
|
Grins E, Leacche M, Shrestha NM, Bjursten H, Ederoth P, Jovinge S. Interleukin-10: A Potential Pre-Cannulation Marker for Development of Acute Kidney Injury in Patients Receiving Veno-Arterial Extracorporeal Membrane Oxygenation. Blood Purif 2023; 52:631-641. [PMID: 37586332 DOI: 10.1159/000531328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 05/18/2023] [Indexed: 08/18/2023]
Abstract
INTRODUCTION Acute kidney injury (AKI) in patients treated with veno-arterial extracorporeal membrane oxygenation (VA-ECMO) is associated with high mortality. The objective of this study was to investigate whether cytokine levels before the initiation of ECMO treatment could predict AKI. We also aimed to investigate the impact of AKI on 30-day and 1-year mortality. METHODS Serum cytokine levels were analyzed in 100 consecutive VA-ECMO-treated patients at pre-cannulation, at 48 h post-cannulation, and at 8 days. Clinical data to establish the incidence and outcome of AKI after the start of ECMO was retrieved from the local ECMO registry. SETTING The study was conducted at tertiary care, university hospital. Participants included 100 patients treated with VA-ECMO. INTERVENTIONS The blood samples for cytokine analysis were collected before VA-ECMO treatment, at 48 h after VA-ECMO treatment was started, and at 8 days. RESULTS Pre-cannulation serum IL-10 levels were significantly higher in patients who developed AKI (212 [38.9, 620.7]) versus those who did not (49.0 [11.9, 102.2]; p = 0.007), and the development of AKI can be predicted by pre-cannulation IL-10 levels (p = 0.025, OR = 1.2 [1.02-1.32]). The development of AKI during ECMO treatment is associated with increased 30-day mortality (p = 0.049) compared to patients who did not develop AKI and had a pre-cannulation estimated glomerular filtration rate ≥ 45 mL/min. The 1-year survival rate for patients with AKI who survived the first 30 days of ECMO treatment is comparable to that of patients without AKI. CONCLUSION Increased pre-cannulation IL-10 levels are associated with the development of AKI during VA-ECMO support. AKI is associated with increased 30-day mortality compared to patients with no AKI and better renal function. However, patients with AKI who survive the first 30 days have a 1-year survival rate similar to those without AKI.
Collapse
Affiliation(s)
- Edgars Grins
- Department of Anesthesiology and Intensive Care, Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Cardiothoracic and Vascular Surgery, Anesthesia, and Intensive Care, Skåne University Hospital, Lund, Sweden
- DeVos Cardiovascular Research Program, Spectrum Health and Van Andel Institute, Grand Rapids, Michigan, USA
| | - Marzia Leacche
- DeVos Cardiovascular Research Program, Spectrum Health and Van Andel Institute, Grand Rapids, Michigan, USA
- Fredrik Meijer Heart and Vascular Institute, Spectrum Health Grand Rapids, Grand Rapids, Michigan, USA
| | - Nabin Manandhar Shrestha
- DeVos Cardiovascular Research Program, Spectrum Health and Van Andel Institute, Grand Rapids, Michigan, USA
- Fredrik Meijer Heart and Vascular Institute, Spectrum Health Grand Rapids, Grand Rapids, Michigan, USA
| | - Henrik Bjursten
- Department of Cardiothoracic and Vascular Surgery, Anesthesia, and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Per Ederoth
- Department of Anesthesiology and Intensive Care, Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Cardiothoracic and Vascular Surgery, Anesthesia, and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Stefan Jovinge
- DeVos Cardiovascular Research Program, Spectrum Health and Van Andel Institute, Grand Rapids, Michigan, USA
- Fredrik Meijer Heart and Vascular Institute, Spectrum Health Grand Rapids, Grand Rapids, Michigan, USA
- Cardiovascular Institute Stanford University, Palo Alto, California, USA
| |
Collapse
|
5
|
See KC. Personalizing Care for Critically Ill Adults Using Omics: A Concise Review of Potential Clinical Applications. Cells 2023; 12:541. [PMID: 36831207 PMCID: PMC9954497 DOI: 10.3390/cells12040541] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/30/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Current guidelines for critically ill patients use broad recommendations to promote uniform protocols for the management of conditions such as acute kidney injury, acute respiratory distress syndrome, and sepsis. Although these guidelines have enabled the substantial improvement of care, mortality for critical illness remains high. Further outcome improvement may require personalizing care for critically ill patients, which involves tailoring management strategies for different patients. However, the current understanding of disease heterogeneity is limited. For critically ill patients, genomics, transcriptomics, proteomics, and metabolomics have illuminated such heterogeneity and unveiled novel biomarkers, giving clinicians new means of diagnosis, prognosis, and monitoring. With further engineering and economic development, omics would then be more accessible and affordable for frontline clinicians. As the knowledge of pathophysiological pathways mature, targeted treatments can then be developed, validated, replicated, and translated into clinical practice.
Collapse
Affiliation(s)
- Kay Choong See
- Division of Respiratory and Critical Care Medicine, Department of Medicine, National University Hospital, Singapore 119228, Singapore
| |
Collapse
|
6
|
Chu CM, Chung CJ, Huang CY, Yu CC, Wang CH, Li LF, Wu HP. Serial Increases in Human Leukocyte Antigen-DR Expression and Decreases in Interleukin-10 Expression in Alveolar Monocytes of Survivors of Pneumonia-Related Acute Respiratory Distress Syndrome. BIOLOGY 2022; 11:biology11121793. [PMID: 36552302 PMCID: PMC9775347 DOI: 10.3390/biology11121793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/24/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
ARDS is a potentially lethal syndrome. HLA-DR expression in monocytes reflects their activation and antigen-presenting capacity. However, the correlation between clinical outcomes and HLA-DR expression in alveolar monocytes/macrophages in patients with pneumonia-related ARDS remains unclear. Thus, we determined the trends of HLA-DR and cytokine expressions in alveolar monocytes using repeated measurements to answer this question. Thirty-one pneumonia patients with respiratory failure and ARDS without coronavirus disease 2019 between November 2019 and November 2021 were enrolled in our intensive care unit and three without complete data were excluded. Interleukin (IL)-10, IL-12, and HLA-DR expression in bronchoalveolar lavage (BAL) monocytes were determined on days one and eight. Monocyte HLA-DR expression (mHLA-DR) and CD4 T lymphocytes percentages in BAL cells of survivors increased remarkably after seven days. Monocyte IL-10 expression and monocytes percentages in BAL cells of survivors decreased substantially after seven days. The mHLA-DR was negatively correlated with disease severity scores on day one and eight. In conclusion, serial increases in HLA-DR expression and decreases in IL-10 expression were observed in BAL monocytes of survivors of pneumonia-related ARDS. More studies are needed to confirm this point of view, and then development of a therapeutic agent restoring mHLA-DR and preventing IL-10 production can be considered.
Collapse
Affiliation(s)
- Chien-Ming Chu
- Division of Pulmonary, Critical Care and Sleep Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Chia-Jung Chung
- Division of Pulmonary, Critical Care and Sleep Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Chih-Yu Huang
- Division of Pulmonary, Critical Care and Sleep Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chung-Chieh Yu
- Division of Pulmonary, Critical Care and Sleep Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chao-Hung Wang
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Heart Failure Research Center, Division of Cardiology, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Li-Fu Li
- Division of Pulmonary, Critical Care and Sleep Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Huang-Pin Wu
- Division of Pulmonary, Critical Care and Sleep Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Correspondence: ; Tel.: +886-2-24313131 (ext. 6204); Fax: +886-2-24335342
| |
Collapse
|
7
|
Battaglini D, Al-Husinat L, Normando AG, Leme AP, Franchini K, Morales M, Pelosi P, Rocco PR. Personalized medicine using omics approaches in acute respiratory distress syndrome to identify biological phenotypes. Respir Res 2022; 23:318. [PMID: 36403043 PMCID: PMC9675217 DOI: 10.1186/s12931-022-02233-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/01/2022] [Indexed: 11/21/2022] Open
Abstract
In the last decade, research on acute respiratory distress syndrome (ARDS) has made considerable progress. However, ARDS remains a leading cause of mortality in the intensive care unit. ARDS presents distinct subphenotypes with different clinical and biological features. The pathophysiologic mechanisms of ARDS may contribute to the biological variability and partially explain why some pharmacologic therapies for ARDS have failed to improve patient outcomes. Therefore, identifying ARDS variability and heterogeneity might be a key strategy for finding effective treatments. Research involving studies on biomarkers and genomic, metabolomic, and proteomic technologies is increasing. These new approaches, which are dedicated to the identification and quantitative analysis of components from biological matrixes, may help differentiate between different types of damage and predict clinical outcome and risk. Omics technologies offer a new opportunity for the development of diagnostic tools and personalized therapy in ARDS. This narrative review assesses recent evidence regarding genomics, proteomics, and metabolomics in ARDS research.
Collapse
Affiliation(s)
- Denise Battaglini
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, Instituto di Ricovero e Cura a Carattere Scientifico (IRCCS) for Oncology and Neuroscience, Genoa, Italy
- Department of Surgical Science and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Lou'i Al-Husinat
- Department of Clinical Medical Sciences, Faculty of Medicine, Yarmouk University, P.O. Box 566, Irbid, 21163, Jordan
| | - Ana Gabriela Normando
- Brazilian Biosciences National Laboratory, LNBio, Brazilian Center for Research in Energy and Materials, CNPEM, Campinas, Brazil
| | - Adriana Paes Leme
- Brazilian Biosciences National Laboratory, LNBio, Brazilian Center for Research in Energy and Materials, CNPEM, Campinas, Brazil
| | - Kleber Franchini
- Brazilian Biosciences National Laboratory, LNBio, Brazilian Center for Research in Energy and Materials, CNPEM, Campinas, Brazil
| | - Marcelo Morales
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paolo Pelosi
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, Instituto di Ricovero e Cura a Carattere Scientifico (IRCCS) for Oncology and Neuroscience, Genoa, Italy
- Department of Surgical Science and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Patricia Rm Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
8
|
Martinez-Orengo N, Tahmazian S, Lai J, Wang Z, Sinharay S, Schreiber-Stainthorp W, Basuli F, Maric D, Reid W, Shah S, Hammoud DA. Assessing organ-level immunoreactivity in a rat model of sepsis using TSPO PET imaging. Front Immunol 2022; 13:1010263. [PMID: 36439175 PMCID: PMC9685400 DOI: 10.3389/fimmu.2022.1010263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
Abstract
There is current need for new approaches to assess/measure organ-level immunoreactivity and ensuing dysfunction in systemic inflammatory response syndrome (SIRS) and sepsis, in order to protect or recover organ function. Using a rat model of systemic sterile inflammatory shock (intravenous LPS administration), we performed PET imaging with a translocator protein (TSPO) tracer, [18F]DPA-714, as a biomarker for reactive immunoreactive changes in the brain and peripheral organs. In vivo dynamic PET/CT scans showed increased [18F]DPA-714 binding in the brain, lungs, liver and bone marrow, 4 hours after LPS injection. Post-LPS mean standard uptake values (SUVmean) at equilibrium were significantly higher in those organs compared to baseline. Changes in spleen [18F]DPA-714 binding were variable but generally decreased after LPS. SUVmean values in all organs, except the spleen, positively correlated with several serum cytokines/chemokines. In vitro measures of TSPO expression and immunofluorescent staining validated the imaging results. Noninvasive molecular imaging with [18F]DPA-714 PET in a rat model of systemic sterile inflammatory shock, along with in vitro measures of TSPO expression, showed brain, liver and lung inflammation, spleen monocytic efflux/lymphocytic activation and suggested increased bone marrow hematopoiesis. TSPO PET imaging can potentially be used to quantify SIRS and sepsis-associated organ-level immunoreactivity and assess the effectiveness of therapeutic and preventative approaches for associated organ failures, in vivo.
Collapse
Affiliation(s)
- Neysha Martinez-Orengo
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Sarine Tahmazian
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Jianhao Lai
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Zeping Wang
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Sanhita Sinharay
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - William Schreiber-Stainthorp
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Rockville, MD, United States
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - William Reid
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Swati Shah
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Dima A. Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Dima A. Hammoud,
| |
Collapse
|
9
|
Predicting the severity of viral bronchiolitis in children. ACTA BIOMEDICA SCIENTIFICA 2022. [DOI: 10.29413/abs.2022-7.3.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Acute viral bronchiolitis is one of the common causes of hospitalization and mortality, especially among children in the first year of life who have risk factors (prematurity, congenital heart defects, bronchopulmonary dysplasia, immunosuppression). As factors associated with the severe course of bronchiolitis, along with the traditional ones, single nucleotide polymorphisms of the genes of the immune response molecules can be considered.The aim. Based on the analysis of clinical, laboratory and molecular genetic parameters, to identify prognostic criteria for the severe course of acute viral bronchiolitis in children.Materials and methods. The study included 106 children with acute viral bronchiolitis (severe course – 34, mild course – 72), the etiology of which in 67.9 % was respiratory syncytial virus. Forty-seven anamnestic, clinical, traditional laboratory and molecular genetic parameters were assessed as prognostic criteria. Determination of SNP genes of cytokines IL-4 (C-589T), IL-10 (G-1082A), IL-10 (C-592A), IL-10 (C-819T), TNF-α (G-308A), IL-17A (G197A), IL-17F (His161Arg), TLR2-753ArgGln, TLR6-Ser249Pro in venous blood was carried out by the polymerase chain reaction method.Results. An additional criterion for the risk of developing a severe course of bronchiolitis can be the mutant genotype (AA) SNP of the IL-10 gene (C-592A), which was detected exclusively in the group of patients with severe bronchiolitis, increasing the risk of developing a severe disease by 16.11 times (OR = 16.11; 95 % CI: 0.81–121.22, p = 0.02) in conjunction with already established modifying factors: the presence of congenital heart disease, bronchopulmonary dysplasia, prematurity, birth weight < 1500 g. Based on a comprehensive assessment of the established risk factors, a method has been developed that allows calculate the likelihood of developing a severe course of acute viral bronchiolitis. Conclusion. The use of the developed prediction method will not only increase the likelihood of developing severe acute viral bronchiolitis in children, but also determine the priority group among children with predictors of severe viral bronchiolitis for priority immunoprophylaxis against RS-virus infection.
Collapse
|
10
|
Mukherjee D, Chora ÂF, Lone JC, Ramiro RS, Blankenhaus B, Serre K, Ramirez M, Gordo I, Veldhoen M, Varga-Weisz P, Mota MM. Host lung microbiota promotes malaria-associated acute respiratory distress syndrome. Nat Commun 2022; 13:3747. [PMID: 35768411 PMCID: PMC9243033 DOI: 10.1038/s41467-022-31301-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 06/13/2022] [Indexed: 11/14/2022] Open
Abstract
Severe malaria can manifest itself with a variety of well-recognized clinical phenotypes that are highly predictive of death - severe anaemia, coma (cerebral malaria), multiple organ failure, and respiratory distress. The reasons why an infected individual develops one pathology rather than another remain poorly understood. Here we use distinct rodent models of infection to show that the host microbiota is a contributing factor for the development of respiratory distress syndrome and host mortality in the context of malaria infections (malaria-associated acute respiratory distress syndrome, MA-ARDS). We show that parasite sequestration in the lung results in sustained immune activation. Subsequent production of the anti-inflammatory cytokine IL-10 by T cells compromises microbial control, leading to severe lung disease. Notably, bacterial clearance with linezolid, an antibiotic commonly used in the clinical setting to control lung-associated bacterial infections, prevents MA-ARDS-associated lethality. Thus, we propose that the host's anti-inflammatory response to limit tissue damage can result in loss of microbial control, which promotes MA-ARDS. This must be considered when intervening against life-threatening respiratory complications.
Collapse
Affiliation(s)
- Debanjan Mukherjee
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Ângelo Ferreira Chora
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Jean-Christophe Lone
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
- School of Life Sciences, University of Essex, Colchester, UK
| | | | - Birte Blankenhaus
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Karine Serre
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Mário Ramirez
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Isabel Gordo
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Marc Veldhoen
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Patrick Varga-Weisz
- School of Life Sciences, University of Essex, Colchester, UK
- São Paulo Excellence Chair, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Maria M Mota
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal.
| |
Collapse
|
11
|
Abstract
This article is one of ten reviews selected from the Annual Update in Intensive Care and Emergency Medicine 2021. Other selected articles can be found online at https://www.biomedcentral.com/collections/annualupdate2021 . Further information about the Annual Update in Intensive Care and Emergency Medicine is available from https://link.springer.com/bookseries/8901 .
Collapse
Affiliation(s)
- Marlies Ostermann
- Department of Critical Care, King's College London, Guy's and St Thomas' NHS Foundation Trust, London, UK.
| | - Nuttha Lumlertgul
- Department of Critical Care, King's College London, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Division of Nephrology and Excellence Centre for Critical Care Nephrology, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
- Critical Care Nephrology Research Unit, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
12
|
Drohan CM, Nouraie SM, Bain W, Shah FA, Evankovich J, Zhang Y, Morris A, McVerry BJ, Kitsios GD. Biomarker-Based Classification of Patients With Acute Respiratory Failure Into Inflammatory Subphenotypes: A Single-Center Exploratory Study. Crit Care Explor 2021; 3:e0518. [PMID: 34476405 PMCID: PMC8378789 DOI: 10.1097/cce.0000000000000518] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVES Hyper- and hypoinflammatory subphenotypes discovered in patients with acute respiratory distress syndrome predict clinical outcomes and therapeutic responses. These subphenotypes may be important in broader critically ill patient populations with acute respiratory failure regardless of clinical diagnosis. We investigated subphenotyping with latent class analysis in an inclusive population of acute respiratory failure, derived a parsimonious model for subphenotypic predictions based on a small set of variables, and examined associations with clinical outcomes. DESIGN Prospective, observational cohort study. SETTING Single-center, academic medical ICU. PATIENTS Mechanically ventilated patients with acute respiratory failure. MEASUREMENTS AND MAIN RESULTS We included 498 patients with acute respiratory failure (acute respiratory distress syndrome: 143, at-risk for acute respiratory distress syndrome: 198, congestive heart failure: 37, acute on chronic respiratory failure: 23, airway protection: 61, and multifactorial: 35) in our derivation cohort and measured 10 baseline plasma biomarkers. Latent class analysis considering clinical variables and biomarkers determined that a two-class model offered optimal fit (23% hyperinflammatory subphenotype). Distribution of hyperinflammatory subphenotype varied among acute respiratory failure etiologies (acute respiratory distress syndrome: 31%, at-risk for acute respiratory distress syndrome: 27%, congestive heart failure: 22%, acute on chronic respiratory failure 0%, airway protection: 5%, and multifactorial: 14%). Hyperinflammatory patients had higher Sequential Organ Failure Assessment scores, fewer ventilator-free days, and higher 30- and 90-day mortality (all p < 0.001). We derived a parsimonious model consisting of angiopoietin-2, soluble tumor necrosis factor receptor-1, procalcitonin, and bicarbonate and classified subphenotypes in a validation cohort (n = 139). Hyperinflammatory patients (19%) demonstrated higher levels of inflammatory biomarkers not included in the model (p < 0.01) and worse outcomes. CONCLUSIONS Host-response subphenotypes are observable in a heterogeneous population with acute respiratory failure and predict clinical outcomes. Simple, biomarker-based models can offer prognostic enrichment in patients with acute respiratory failure. The differential distribution of subphenotypes by specific etiologies of acute respiratory failure indicates that subphenotyping may be more relevant in patients with hypoxemic causes of acute respiratory failure and not in patients intubated for airway protection or acute on chronic decompensation.
Collapse
Affiliation(s)
- Callie M Drohan
- Division of General Internal Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - S Mehdi Nouraie
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - William Bain
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Staff Physician, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA
| | - Faraaz A Shah
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Staff Physician, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA
| | - John Evankovich
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Yingze Zhang
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Alison Morris
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Center for Medicine and the Microbiome, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Bryan J McVerry
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Center for Medicine and the Microbiome, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Georgios D Kitsios
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Center for Medicine and the Microbiome, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
13
|
Stenlo M, Silva IAN, Hyllén S, Bölükbas DA, Niroomand A, Grins E, Ederoth P, Hallgren O, Pierre L, Wagner DE, Lindstedt S. Monitoring lung injury with particle flow rate in LPS- and COVID-19-induced ARDS. Physiol Rep 2021; 9:e14802. [PMID: 34250766 PMCID: PMC8273428 DOI: 10.14814/phy2.14802] [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: 02/07/2021] [Revised: 02/18/2021] [Accepted: 02/21/2021] [Indexed: 11/24/2022] Open
Abstract
In severe acute respiratory distress syndrome (ARDS), extracorporeal membrane oxygenation (ECMO) is a life-prolonging treatment, especially among COVID-19 patients. Evaluation of lung injury progression is challenging with current techniques. Diagnostic imaging or invasive diagnostics are risky given the difficulties of intra-hospital transportation, contraindication of biopsies, and the potential for the spread of infections, such as in COVID-19 patients. We have recently shown that particle flow rate (PFR) from exhaled breath could be a noninvasive, early detection method for ARDS during mechanical ventilation. We hypothesized that PFR could also measure the progress of lung injury during ECMO treatment. Lipopolysaccharide (LPS) was thus used to induce ARDS in pigs under mechanical ventilation. Eight were connected to ECMO, whereas seven animals were not. In addition, six animals received sham treatment with saline. Four human patients with ECMO and ARDS were also monitored. In the pigs, as lung injury ensued, the PFR dramatically increased and a particular spike followed the establishment of ECMO in the LPS-treated animals. PFR remained elevated in all animals with no signs of lung recovery. In the human patients, in the two that recovered, PFR decreased. In the two whose lung function deteriorated while on ECMO, there was increased PFR with no sign of recovery in lung function. The present results indicate that real-time monitoring of PFR may be a new, complementary approach in the clinic for measurement of the extent of lung injury and recovery over time in ECMO patients with ARDS.
Collapse
Affiliation(s)
- Martin Stenlo
- Department of Cardiothoracic Anaesthesia and Intensive Care and Cardiothoracic Surgery and TransplantationSkåne University HospitalLund UniversitySweden
- Wallenberg Center for Molecular MedicineLund UniversitySweden
- Department of Clinical SciencesLund UniversitySweden
| | - Iran A. N. Silva
- Department of Experimental Medical SciencesLung Bioengineering and RegenerationLund UniversitySweden
- Wallenberg Center for Molecular MedicineLund UniversitySweden
- Lund Stem Cell CenterLund UniversitySweden
| | - Snejana Hyllén
- Department of Cardiothoracic Anaesthesia and Intensive Care and Cardiothoracic Surgery and TransplantationSkåne University HospitalLund UniversitySweden
- Department of Clinical SciencesLund UniversitySweden
| | - Deniz A. Bölükbas
- Department of Experimental Medical SciencesLung Bioengineering and RegenerationLund UniversitySweden
- Wallenberg Center for Molecular MedicineLund UniversitySweden
- Lund Stem Cell CenterLund UniversitySweden
| | - Anna Niroomand
- Department of Clinical SciencesLund UniversitySweden
- Rutgers Robert UniversityNew BrunswickNew JerseyUSA
| | - Edgars Grins
- Department of Cardiothoracic Anaesthesia and Intensive Care and Cardiothoracic Surgery and TransplantationSkåne University HospitalLund UniversitySweden
- Department of Clinical SciencesLund UniversitySweden
| | - Per Ederoth
- Department of Cardiothoracic Anaesthesia and Intensive Care and Cardiothoracic Surgery and TransplantationSkåne University HospitalLund UniversitySweden
- Department of Clinical SciencesLund UniversitySweden
| | - Oskar Hallgren
- Wallenberg Center for Molecular MedicineLund UniversitySweden
- Department of Clinical SciencesLund UniversitySweden
| | - Leif Pierre
- Department of Cardiothoracic Anaesthesia and Intensive Care and Cardiothoracic Surgery and TransplantationSkåne University HospitalLund UniversitySweden
- Department of Clinical SciencesLund UniversitySweden
| | - Darcy E. Wagner
- Department of Experimental Medical SciencesLung Bioengineering and RegenerationLund UniversitySweden
- Wallenberg Center for Molecular MedicineLund UniversitySweden
- Lund Stem Cell CenterLund UniversitySweden
| | - Sandra Lindstedt
- Department of Cardiothoracic Anaesthesia and Intensive Care and Cardiothoracic Surgery and TransplantationSkåne University HospitalLund UniversitySweden
- Wallenberg Center for Molecular MedicineLund UniversitySweden
- Department of Clinical SciencesLund UniversitySweden
| |
Collapse
|
14
|
Ki KK, Millar JE, Langguth D, Passmore MR, McDonald CI, Shekar K, Shankar-Hari M, Cho HJ, Suen JY, Fraser JF. Current Understanding of Leukocyte Phenotypic and Functional Modulation During Extracorporeal Membrane Oxygenation: A Narrative Review. Front Immunol 2021; 11:600684. [PMID: 33488595 PMCID: PMC7821656 DOI: 10.3389/fimmu.2020.600684] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
A plethora of leukocyte modulations have been reported in critically ill patients. Critical illnesses such as acute respiratory distress syndrome and cardiogenic shock, which potentially require extracorporeal membrane oxygenation (ECMO) support, are associated with changes in leukocyte numbers, phenotype, and functions. The changes observed in these illnesses could be compounded by exposure of blood to the non-endothelialized surfaces and non-physiological conditions of ECMO. This can result in further leukocyte activation, increased platelet-leukocyte interplay, pro-inflammatory and pro-coagulant state, alongside features of immunosuppression. However, the effects of ECMO on leukocytes, in particular their phenotypic and functional signatures, remain largely overlooked, including whether these changes have attributable mortality and morbidity. The aim of our narrative review is to highlight the importance of studying leukocyte signatures to better understand the development of complications associated with ECMO. Increased knowledge and appreciation of their probable role in ECMO-related adverse events may assist in guiding the design and establishment of targeted preventative actions.
Collapse
Affiliation(s)
- Katrina K Ki
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Jonathan E Millar
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Daman Langguth
- Clinical Immunology and Allergy, and Sullivan Nicolaides Pathology, Wesley Hospital, Brisbane, QLD, Australia
| | - Margaret R Passmore
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Charles I McDonald
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Department of Anaesthesia and Perfusion, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Kiran Shekar
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Adult Intensive Care Service, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Manu Shankar-Hari
- Department of Intensive Care Unit, Guy's and St Thomas' Hospital NHS Foundation Trust, London, United Kingdom.,School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Hwa Jin Cho
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.,Department of Paediatrics, Chonnam National University Children's Hospital and Medical School, Gwangju, South Korea
| | - Jacky Y Suen
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - John F Fraser
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia.,Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
15
|
Noroozi R, Branicki W, Pyrc K, Łabaj PP, Pospiech E, Taheri M, Ghafouri-Fard S. Altered cytokine levels and immune responses in patients with SARS-CoV-2 infection and related conditions. Cytokine 2020; 133:155143. [PMID: 32460144 PMCID: PMC7241386 DOI: 10.1016/j.cyto.2020.155143] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/17/2020] [Accepted: 05/19/2020] [Indexed: 01/06/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a global pandemic in early 2020. The infection has been associated with a wide range of clinical symptoms. In the severely affected patients, it has caused dysregulation of immune responses including over-secretion of inflammatory cytokines and imbalances in the proportion of naïve helper T cells, memory helper T cells and regulatory T cells. Identification of the underlying mechanism of such aberrant function of immune system would help in the prediction of disease course and selection of susceptible patients for more intensive cares. In the current review, we summarize the results of studies which reported alterations in cytokine levels and immune cell functions in patients affected with SARS-CoV-2 and related viruses.
Collapse
Affiliation(s)
- Rezvan Noroozi
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Wojciech Branicki
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Krzysztof Pyrc
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Paweł P Łabaj
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Ewelina Pospiech
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Ghafouri-Fard S, Noroozi R, Vafaee R, Branicki W, Poṡpiech E, Pyrc K, Łabaj PP, Omrani MD, Taheri M, Sanak M. Effects of host genetic variations on response to, susceptibility and severity of respiratory infections. Biomed Pharmacother 2020; 128:110296. [PMID: 32480226 PMCID: PMC7258806 DOI: 10.1016/j.biopha.2020.110296] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/16/2020] [Accepted: 05/19/2020] [Indexed: 12/13/2022] Open
Abstract
The recent outbreak of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has created a global crisis, necessitating the identification of genetic factors that modulate the risk of disorder or its severity. The current data about the role of genetic risk factors in determination of rate of SARS-CoV-2 infection in each ethnic group and the severity of disorder is limited. Moreover, several confounding parameters such as the number of tests performed in each country, the structure of the population especially the age distribution, the presence of risk factors for respiratory disorders such as smoking and other environmental factors might be involved in the variability in disease course or prevalence of infection among different ethnic groups. However, assessment of the role of genetic variants in determination of the course of other respiratory infections might help in recognition of possible candidate for further analysis in patients affected with SARS-CoV-2. In the current review, we summarize the data showing the association between genomic variants and risk of acute respiratory distress syndrome, respiratory infections or severity of these conditions with an especial focus on the SARS-CoV-2.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rezvan Noroozi
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Reza Vafaee
- Proteomics Research Center, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Wojciech Branicki
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Ewelina Poṡpiech
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Krzysztof Pyrc
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Paweł P Łabaj
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Mir Davood Omrani
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Marek Sanak
- Department of Internal Medicine, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
17
|
Kotok D, Yang L, Evankovich JW, Bain W, Dunlap DG, Shah F, Zhang Y, Manatakis DV, Benos PV, Barbash IJ, Rapport SF, Lee JS, Morris A, McVerry BJ, Kitsios GD. The evolution of radiographic edema in ARDS and its association with clinical outcomes: A prospective cohort study in adult patients. J Crit Care 2020; 56:222-228. [PMID: 32028223 PMCID: PMC7136845 DOI: 10.1016/j.jcrc.2020.01.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 12/13/2019] [Accepted: 01/12/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE To assess the longitudinal evolution of radiographic edema using chest X-rays (CXR) in patients with Acute Respiratory Distress Syndrome (ARDS) and to examine its association with prognostic biomarkers, ARDS subphenotypes and outcomes. MATERIALS AND METHODS We quantified radiographic edema on CXRs from patients with ARDS or cardiogenic pulmonary edema (controls) using the Radiographic Assessment of Lung Edema (RALE) score on day of intubation and up to 10 days after. We measured baseline plasma biomarkers and recorded clinical variables. RESULTS The RALE score had good inter-rater agreement (r = 0.83, p < 0.0001) applied on 488 CXRs from 129 patients, with higher RALE scores in patients with ARDS (n = 108) compared to controls (n = 21, p = 0.01). Baseline RALE scores were positively correlated with levels of the receptor for end-glycation end products (RAGE) in ARDS patients (p < 0.05). Baseline RALE scores were not predictive of 30- or 90-day survival. Persistently elevated RALE scores were associated with prolonged need for mechanical ventilation (p = 0.002). CONCLUSIONS The RALE score is easily implementable with high inter-rater reliability. Longitudinal RALE scoring appears to be a reproducible approach to track the evolution of radiographic edema in patients with ARDS and can potentially predict prolonged need for mechanical ventilation.
Collapse
Affiliation(s)
- Daniel Kotok
- Internal Medicine Residency Program, University of Pittsburgh Medical Center McKeesport, USA
| | - Libing Yang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - John W Evankovich
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - William Bain
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Daniel G Dunlap
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Faraaz Shah
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Yingze Zhang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Dimitris V Manatakis
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Panayiotis V Benos
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ian J Barbash
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Sarah F Rapport
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Janet S Lee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Alison Morris
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA; Center for Medicine and the Microbiome, University of Pittsburgh, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Bryan J McVerry
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA; Center for Medicine and the Microbiome, University of Pittsburgh, USA
| | - Georgios D Kitsios
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA; Center for Medicine and the Microbiome, University of Pittsburgh, USA.
| |
Collapse
|
18
|
Stenlo M, Hyllén S, Silva IAN, Bölükbas DA, Pierre L, Hallgren O, Wagner DE, Lindstedt S. Increased particle flow rate from airways precedes clinical signs of ARDS in a porcine model of LPS-induced acute lung injury. Am J Physiol Lung Cell Mol Physiol 2020; 318:L510-L517. [PMID: 31994907 PMCID: PMC7191636 DOI: 10.1152/ajplung.00524.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common cause of death in the intensive care unit, with mortality rates of ~30-40%. To reduce invasive diagnostics such as bronchoalveolar lavage and time-consuming in-hospital transports for imaging diagnostics, we hypothesized that particle flow rate (PFR) pattern from the airways could be an early detection method and contribute to improving diagnostics and optimizing personalized therapies. Porcine models were ventilated mechanically. Lipopolysaccharide (LPS) was administered endotracheally and in the pulmonary artery to induce ARDS. PFR was measured using a customized particles in exhaled air (PExA 2.0) device. In contrast to control animals undergoing mechanical ventilation and receiving saline administration, animals who received LPS developed ARDS according to clinical guidelines and histologic assessment. Plasma levels of TNF-α and IL-6 increased significantly compared with baseline after 120 and 180 min, respectively. On the other hand, the PFR significantly increased and peaked 60 min after LPS administration, i.e., ~30 min before any ARDS stage was observed with other well-established outcome measurements such as hypoxemia, increased inspiratory pressure, and lower tidal volumes or plasma cytokine levels. The present results imply that PFR could be used to detect early biomarkers or as a clinical indicator for the onset of ARDS.
Collapse
Affiliation(s)
- Martin Stenlo
- Department of Cardiothoracic Anaesthesia and Intensive Care and Cardiothoracic Surgery and Transplantation, Skåne University Hospital, Lund University, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.,Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Snejana Hyllén
- Department of Cardiothoracic Anaesthesia and Intensive Care and Cardiothoracic Surgery and Transplantation, Skåne University Hospital, Lund University, Lund, Sweden.,Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Iran A N Silva
- Department of Experimental Medical Sciences, Lung Bioengineering and Regeneration, Lund University, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Deniz A Bölükbas
- Department of Experimental Medical Sciences, Lung Bioengineering and Regeneration, Lund University, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Leif Pierre
- Department of Cardiothoracic Anaesthesia and Intensive Care and Cardiothoracic Surgery and Transplantation, Skåne University Hospital, Lund University, Lund, Sweden.,Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Oskar Hallgren
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.,Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Darcy E Wagner
- Department of Experimental Medical Sciences, Lung Bioengineering and Regeneration, Lund University, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Sandra Lindstedt
- Department of Cardiothoracic Anaesthesia and Intensive Care and Cardiothoracic Surgery and Transplantation, Skåne University Hospital, Lund University, Lund, Sweden.,Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden.,Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
19
|
Kitsios GD, Yang L, Manatakis DV, Nouraie M, Evankovich J, Bain W, Dunlap DD, Shah F, Barbash IJ, Rapport SF, Zhang Y, DeSensi RS, Weathington NM, Chen BB, Ray P, Mallampalli RK, Benos PV, Lee JS, Morris A, McVerry BJ. Host-Response Subphenotypes Offer Prognostic Enrichment in Patients With or at Risk for Acute Respiratory Distress Syndrome. Crit Care Med 2019; 47:1724-1734. [PMID: 31634231 PMCID: PMC6865808 DOI: 10.1097/ccm.0000000000004018] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVES Classification of patients with acute respiratory distress syndrome into hyper- and hypoinflammatory subphenotypes using plasma biomarkers may facilitate more effective targeted therapy. We examined whether established subphenotypes are present not only in patients with acute respiratory distress syndrome but also in patients at risk for acute respiratory distress syndrome (ARFA) and then assessed the prognostic information of baseline subphenotyping on the evolution of host-response biomarkers and clinical outcomes. DESIGN Prospective, observational cohort study. SETTING Medical ICU at a tertiary academic medical center. PATIENTS Mechanically ventilated patients with acute respiratory distress syndrome or ARFA. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS We performed longitudinal measurements of 10 plasma biomarkers of host injury and inflammation. We applied unsupervised latent class analysis methods utilizing baseline clinical and biomarker variables and demonstrated that two-class models (hyper- vs hypoinflammatory subphenotypes) offered improved fit compared with one-class models in both patients with acute respiratory distress syndrome and ARFA. Baseline assignment to the hyperinflammatory subphenotype (39/104 [38%] acute respiratory distress syndrome and 30/108 [28%] ARFA patients) was associated with higher severity of illness by Sequential Organ Failure Assessment scores and incidence of acute kidney injury in patients with acute respiratory distress syndrome, as well as higher 30-day mortality and longer duration of mechanical ventilation in ARFA patients (p < 0.0001). Hyperinflammatory patients exhibited persistent elevation of biomarkers of innate immunity for up to 2 weeks postintubation. CONCLUSIONS Our results suggest that two distinct subphenotypes are present not only in patients with established acute respiratory distress syndrome but also in patients at risk for its development. Hyperinflammatory classification at baseline is associated with higher severity of illness, worse clinical outcomes, and trajectories of persistently elevated biomarkers of host injury and inflammation during acute critical illness compared with hypoinflammatory patients. Our findings provide strong rationale for examining treatment effect modifications by subphenotypes in randomized clinical trials to inform precision therapeutic approaches in critical care.
Collapse
Affiliation(s)
- Georgios D. Kitsios
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Center for Medicine and the Microbiome, University of Pittsburgh
| | - Libing Yang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Dimitris V. Manatakis
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Mehdi Nouraie
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - John Evankovich
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - William Bain
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Daniel D. Dunlap
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Faraaz Shah
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Ian J Barbash
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Sarah F. Rapport
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Yingze Zhang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Rebecca S. DeSensi
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Nathaniel M. Weathington
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Bill B. Chen
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Prabir Ray
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Rama K. Mallampalli
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Veterans Affairs Pittsburgh Healthcare System
| | - Panayiotis V. Benos
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Janet S. Lee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Alison Morris
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Center for Medicine and the Microbiome, University of Pittsburgh
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Bryan J. McVerry
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Center for Medicine and the Microbiome, University of Pittsburgh
| |
Collapse
|
20
|
Esawy MM, Shabana MA, Baioumy SA, Ismail NA. Diagnostic and prognostic roles of peripheral blood Toll-like receptor-4 and stanniocalcin-1 genes expression in acute lung injury. Immunobiology 2019; 224:734-738. [PMID: 31515080 DOI: 10.1016/j.imbio.2019.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/16/2019] [Accepted: 09/03/2019] [Indexed: 01/16/2023]
Abstract
Acute lung injury (ALI) is an acute inflammatory disorder. Toll-like receptor-4 (TLR-4) and Stanniocalcin -1 (STC-1) had roles in lung endothelial protection. This study aims to assess TLR-4 and SCT-1 genes expressions in peripheral blood of ALI patients. Total RNA was extracted from peripheral blood of 48 subjects (20 healthy controls, 28 ALI patients) and expressions of genes were assessed by real-Time qRT-PCR. The expression levels of TLR-4 and SCT-1 genes were significantly lower in ALI patients compared to controls (P < 0.0001). After 10 days, the expression levels of TLR-4 and SCT-1 were increased compared to their baseline levels (p = 0.012 and 0.024, respectively). SCT-1 has 92.9% sensitivity and 100% specificity in ALI detection. SCT-1 gene expression was negatively correlated with severity score (r= -0.54, p = 0.003). The mortality pattern was higher in ALI patients with lower TLR-4 gene expression (p = 0.014). In conclusion, the peripheral blood expressions of TLR-4 and STC-1 genes were decreased in ALI patients. Both genes expressions were increased with patients' recovery. SCT-1 had higher sensitivity and specificity in ALI diagnosis. The peripheral blood expressions of SCT-1 and TLR-4 genes seem to be diagnostic and prognostic markers in ALI.
Collapse
Affiliation(s)
- Marwa M Esawy
- Clinical Pathology Department, Faculty of Human Medicine, Zagazig University, Egypt.
| | - Marwa A Shabana
- Clinical Pathology Department, Faculty of Human Medicine, Zagazig University, Egypt
| | - Shereen A Baioumy
- Microbiology and Immunology Department, Faculty of Human Medicine, Zagazig University, Egypt
| | - Nagwan A Ismail
- Chest Department, Faculty of Human Medicine, Zagazig University, Egypt
| |
Collapse
|
21
|
Al-Fares A, Pettenuzzo T, Del Sorbo L. Extracorporeal life support and systemic inflammation. Intensive Care Med Exp 2019; 7:46. [PMID: 31346840 PMCID: PMC6658641 DOI: 10.1186/s40635-019-0249-y] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 04/22/2019] [Indexed: 01/10/2023] Open
Abstract
Extracorporeal life support (ECLS) encompasses a wide range of extracorporeal modalities that offer short- and intermediate-term mechanical support to the failing heart or lung. Apart from the daily use of cardiopulmonary bypass (CPB) in the operating room, there has been a resurgence of interest and utilization of veno-arterial and veno-venous extracorporeal membrane oxygenation (VA- and VV-ECMO, respectively) and extracorporeal carbon dioxide removal (ECCO2R) in recent years. This might be attributed to the advancement in technology, nonetheless the morbidity and mortality associated with the clinical application of this technology is still significant. The initiation of ECLS triggers a systemic inflammatory response, which involves the activation of the coagulation cascade, complement systems, endothelial cells, leukocytes, and platelets, thus potentially contributing to morbidity and mortality. This is due to the release of cytokines and other biomarkers of inflammation, which have been associated with multiorgan dysfunction. On the other hand, ECLS can be utilized as a therapy to halt the inflammatory response associated with critical illness and ICU therapeutic intervention, such as facilitating ultra-protective mechanical ventilation. In addition to addressing the impact on outcome of the relationship between inflammation and ECLS, two different but complementary pathophysiological perspectives will be developed in this review: ECLS as the cause of inflammation and ECLS as the treatment of inflammation. This framework may be useful in guiding the development of novel therapeutic strategies to improve the outcome of critical illness.
Collapse
Affiliation(s)
- Abdulrahman Al-Fares
- Adult Critical Care Medicine Fellowship Program, University of Toronto, Toronto, Canada.,Al-Amiri Hospital, Ministry of Health, Kuwait City, Kuwait.,Interdepartmental Division of Critical Care Medicine, Toronto General Hospital, University of Toronto, Toronto, Canada
| | - Tommaso Pettenuzzo
- Adult Critical Care Medicine Fellowship Program, University of Toronto, Toronto, Canada.,Interdepartmental Division of Critical Care Medicine, Toronto General Hospital, University of Toronto, Toronto, Canada
| | - Lorenzo Del Sorbo
- Interdepartmental Division of Critical Care Medicine, Toronto General Hospital, University of Toronto, Toronto, Canada. .,Toronto General Hospital, 585 University Avenue, PMB 11-122, Toronto, Ontario, M5G 2 N2, Canada.
| |
Collapse
|
22
|
Husain-Syed F, Ricci Z, Brodie D, Vincent JL, Ranieri VM, Slutsky AS, Taccone FS, Gattinoni L, Ronco C. Extracorporeal organ support (ECOS) in critical illness and acute kidney injury: from native to artificial organ crosstalk. Intensive Care Med 2018; 44:1447-1459. [PMID: 30043276 DOI: 10.1007/s00134-018-5329-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 07/18/2018] [Indexed: 12/11/2022]
Abstract
The complex nature of single organ failure potentially leading to multiple organ dysfunction syndrome (MODS) in critically ill patients necessitates integrated supportive therapy. Rather than a primary disease, acute kidney injury (AKI) is considered a window to a potentially serious underlying systemic disease, which may partially explain the high morbidity and mortality rates associated with the condition. Renal replacement therapy (RRT) has been routinely used for more than a decade in various intensive care settings and there has also been an increase in the use of extracorporeal membrane oxygenation and extracorporeal carbon dioxide removal. When these renal and cardiopulmonary modalities are used together, a multidisciplinary approach is necessary to minimize negative interactions and unwanted adverse effects. In this review, we describe the patterns of organ crosstalk between the native and artificial organs, the incidence of AKI and need for RRT and associated mortality after extracorporeal organ support (ECOS) therapy, including the potential short- and long-term advantages and disadvantages of organ support in terms of renal function. We also review potential indications of RRT outside its conventional indications in patients with MODS, as well as technical considerations when RRT is used alongside other organ support therapies. Overall, available literature has not definitely established the ideal timing of these interventions, and whether early implementation impacts organ recovery and optimizes resource utilization is still a matter of open debate: it is possible that future research will be devoted to identify patient groups that may benefit from short- and long-term multiple organ support.
Collapse
Affiliation(s)
- Faeq Husain-Syed
- Department of Internal Medicine II, Division of Nephrology, Pulmonology and Critical Care Medicine, University Clinic Giessen and Marburg-Campus Giessen, Klinikstrasse 33, 35392, Giessen, Germany.
| | - Zaccaria Ricci
- Department of Cardiology and Cardiac Surgery, Pediatric Cardiac Intensive Care Unit, Bambino Gesu Children's Hospital, IRCCS, Piazza di Sant'Onofrio 4, 00165, Rome, Italy
| | - Daniel Brodie
- Department of Medicine, Columbia University College of Physicians and Surgeons/New York-Presbyterian Hospital, 630 West 168th Street, PH8 East, Room 101, New York, NY, 10032, USA
| | - Jean-Louis Vincent
- Department of Intensive Care, Erasme University Hospital, Route de Lennik 808, 1070, Brussels, Belgium
- Université Libre de Bruxelles, Brussels, Brussels, Belgium
| | - V Marco Ranieri
- Anesthesia and Intensive Care Medicine, Sapienza University of Rome, Policlinico Umberto I Hospital, Viale DEL Policlinico 155, 00161, Rome, Italy
| | - Arthur S Slutsky
- Interdepartmental Division of Critical Care Medicine, Departments of Medicine, Surgery and Biomedical Engineering, University of Toronto, Toronto, Canada
- Keenan Research Center for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
| | - Fabio Silvio Taccone
- Department of Intensive Care, Erasme University Hospital, Route de Lennik 808, 1070, Brussels, Belgium
- Université Libre de Bruxelles, Brussels, Brussels, Belgium
| | - Luciano Gattinoni
- Department of Anesthesiology, Emergency and Intensive Care Medicine, University of Göttingen, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
| | - Claudio Ronco
- Department of Nephrology, Dialysis and Transplantation, San Bortolo Hospital, Via Rodolfi, 37, 36100, Vicenza, Italy
- International Renal Research Institute of Vicenza (IRRIV), Via Rodolfi, 37, 36100, Vicenza, Italy
| |
Collapse
|
23
|
Wang N, Wang J, Jiang R. Effects of IL-10 on OX62, MHC-II and CD86 in bone marrow DCs in rats with organophosphate poisoning. Exp Ther Med 2017; 15:1906-1909. [PMID: 29434782 PMCID: PMC5776626 DOI: 10.3892/etm.2017.5629] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 11/28/2017] [Indexed: 12/25/2022] Open
Abstract
This study investigated the effect of interleukin (IL)-10 on the expression of OX62, MHC-II and CD86 in bone marrow dendritic cells (DCs) in rats with organophosphorus poisoning. Sixty adult SD rats were randomly divided into normal control group (group A) (20 rats), 20 rats with organophosphorus pesticide poisoning (group B), 20 rats with organophosphorus poisoning, and IL-10 treated (group C). Group A was not treated with special treatment. Group B was treated with 4% omethoate by gavage to establish the model of organophosphate poisoning. Group C was treated with omethoate to establish the model of organophosphate poisoning, then the rats were given intraperitoneal injection of IL-10 for 3 continuous days. Rats were sacrificed after 3 days, bone marrow lymphocytes were extracted, DCs were collected and cultured for 7 days, the expression of DC surface antigen OX62, MHC-II, CD86 and related proteins was detected by flow cytometry and western blotting after cell maturation. The expression of DC surface antigen and corresponding protein increased in group B, and decreased in group C, the difference was statistically significant (P<0.05). The results showed that the expression of OX62, MHC-II and CD86 in bone marrow DCs is enhanced and the cellular immune function is enhanced after organophosphate poisoning. IL-10 can down-regulate the antigen presenting function of DCs, achieve anti-inflammatory effect and assist the treatment of organophosphorus pesticide poisoning.
Collapse
Affiliation(s)
- Ning Wang
- Department of Electrophysiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Jinfeng Wang
- Department of Electrophysiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Ronggang Jiang
- Department of Electrophysiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| |
Collapse
|