1
|
Gerard L, Lecocq M, Detry B, Bouzin C, Hoton D, Pinto Pereira J, Carlier F, Plante-Bordeneuve T, Gohy S, Lacroix V, Laterre PF, Pilette C. Airway epithelium damage in acute respiratory distress syndrome. Crit Care 2024; 28:350. [PMID: 39478566 PMCID: PMC11523598 DOI: 10.1186/s13054-024-05127-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/08/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND The airway epithelium (AE) fulfils multiple functions to maintain pulmonary homeostasis, among which ensuring adequate barrier function, cell differentiation and polarization, and actively transporting immunoglobulin A (IgA), the predominant mucosal immunoglobulin in the airway lumen, via the polymeric immunoglobulin receptor (pIgR). Morphological changes of the airways have been reported in ARDS, while their detailed features, impact for mucosal immunity, and causative mechanisms remain unclear. Therefore, this study aimed to assess epithelial alterations in the distal airways of patients with ARDS. METHODS We retrospectively analyzed lung tissue samples from ARDS patients and controls to investigate and quantify structural and functional changes in the small airways, using multiplex fluorescence immunostaining and computer-assisted quantification on whole tissue sections. Additionally, we measured markers of mucosal immunity, IgA and pIgR, alongside with other epithelial markers, in the serum and the broncho-alveolar lavage fluid (BALF) prospectively collected from ARDS patients and controls. RESULTS Compared to controls, airways of ARDS were characterized by increased epithelial denudation (p = 0.0003) and diffuse epithelial infiltration by neutrophils (p = 0.0005). Quantitative evaluation of multiplex fluorescence immunostaining revealed a loss of ciliated cells (p = 0.0317) a trend towards decreased goblet cells (p = 0.056), and no change regarding cell progenitors (basal and club cells), indicating altered mucociliary differentiation. Increased epithelial permeability was also shown in ARDS with a significant decrease of tight (p < 0.0001) and adherens (p = 0.025) junctional proteins. Additionally, we observed a significant decrease of the expression of pIgR, (p < 0.0001), indicating impaired mucosal IgA immunity. Serum concentrations of secretory component (SC) and S-IgA were increased in ARDS (both p < 0.0001), along other lung-derived proteins (CC16, SP-D, sRAGE). However, their BALF concentrations remained unchanged, suggesting a spillover of airway and alveolar proteins through a damaged AE. CONCLUSION The airway epithelium from patients with ARDS exhibits multifaceted alterations leading to altered mucociliary differentiation, compromised defense functions and increased permeability with pneumoproteinemia.
Collapse
Affiliation(s)
- Ludovic Gerard
- Department of Critical Care Medicine, Cliniques universitaires Saint Luc, Université catholique de Louvain (UCLouvain), Avenue Hippocrate 10, 1200, Brussels, Belgium.
- Pôle de Pneumologie, O.R.L. et Dermatologie (LuNS, Lung-Nose-Skin), Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium.
| | - Marylene Lecocq
- Pôle de Pneumologie, O.R.L. et Dermatologie (LuNS, Lung-Nose-Skin), Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Bruno Detry
- Pôle de Pneumologie, O.R.L. et Dermatologie (LuNS, Lung-Nose-Skin), Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Caroline Bouzin
- IREC Imaging Platform (2IP, RRID:SCR_023378), Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Delphine Hoton
- Department of Pathology, Cliniques universitaires Saint Luc, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Joao Pinto Pereira
- Department of Critical Care Medicine, Cliniques universitaires Saint Luc, Université catholique de Louvain (UCLouvain), Avenue Hippocrate 10, 1200, Brussels, Belgium
| | - François Carlier
- Pôle de Pneumologie, O.R.L. et Dermatologie (LuNS, Lung-Nose-Skin), Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- Department of Pulmonology, CHU-UCL Namur, Yvoir, Belgium
| | - Thomas Plante-Bordeneuve
- Pôle de Pneumologie, O.R.L. et Dermatologie (LuNS, Lung-Nose-Skin), Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- Department of Pulmonology, CHU-UCL Namur, Yvoir, Belgium
| | - Sophie Gohy
- Pôle de Pneumologie, O.R.L. et Dermatologie (LuNS, Lung-Nose-Skin), Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- Department of Pulmonology, Cliniques universitaires Saint Luc, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Valérie Lacroix
- Department of Cardiovascular and Thoracic Surgery, Cliniques universitaires Saint Luc, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Pierre-François Laterre
- Department of Intensive Care Medicine, Centre Hospitalier Régional Mons-Hainaut, Mons, Belgium
| | - Charles Pilette
- Pôle de Pneumologie, O.R.L. et Dermatologie (LuNS, Lung-Nose-Skin), Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain (UCLouvain), Brussels, Belgium
- Department of Pulmonology, Cliniques universitaires Saint Luc, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
2
|
Reis APC, Correia FF, Celestrino GA, Pagliari C, Criado PR, Lalwani PJ, Benard G, Sousa MGT. In Situ Expression of TNF-α and IL-10 in Human Dermatophytosis Lesions due to Trichophyton rubrum. Mycopathologia 2024; 189:92. [PMID: 39420083 DOI: 10.1007/s11046-024-00895-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 09/13/2024] [Indexed: 10/19/2024]
Abstract
Dermatophytosis is a very common superficial mycosis, but there are few studies about the human immune response to dermatophytes. We aim to analyze the in situ expression of TNF-α and IL-10 in human dermatophytosis. Expression of TNF-α and IL-10 were evaluated in skin samples from 10 patients with dermatophytosis and 12 healthy subjects using an immunohistochemistry assay. TNF-α and IL-10 were significantly elevated in lesions from patients with dermatophytosis compared to healthy controls. These data illustrate the balance of pro- and anti-inflammatory cytokines suggesting Trichophyton rubrum infection could control the local immune response.
Collapse
Affiliation(s)
- Ana Paula Carvalho Reis
- Laboratório de Investigação Médica LIM 53, Divisão de Clínica Dermatológica, Instituto de Medicina Tropical, Faculdade de Medicina, Hospital das Clínicas, Universidade de São Paulo, São Paulo, São Paulo, Brasil
| | - Franciele Fernandes Correia
- Laboratório de Investigação Médica LIM 53, Divisão de Clínica Dermatológica, Instituto de Medicina Tropical, Faculdade de Medicina, Hospital das Clínicas, Universidade de São Paulo, São Paulo, São Paulo, Brasil
| | - Giovanna Azevedo Celestrino
- Laboratório de Investigação Médica LIM 53, Divisão de Clínica Dermatológica, Instituto de Medicina Tropical, Faculdade de Medicina, Hospital das Clínicas, Universidade de São Paulo, São Paulo, São Paulo, Brasil
| | - Carla Pagliari
- Laboratório da Disciplina de Patologia de Moléstias Transmissíveis, Departamento de Patologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, São Paulo, Brasil
| | | | | | - Gil Benard
- Laboratório de Investigação Médica LIM 53, Divisão de Clínica Dermatológica, Instituto de Medicina Tropical, Faculdade de Medicina, Hospital das Clínicas, Universidade de São Paulo, São Paulo, São Paulo, Brasil
| | - Maria Gloria Teixeira Sousa
- Laboratório de Investigação Médica LIM 53, Divisão de Clínica Dermatológica, Instituto de Medicina Tropical, Faculdade de Medicina, Hospital das Clínicas, Universidade de São Paulo, São Paulo, São Paulo, Brasil.
| |
Collapse
|
3
|
Kuzmanović J, Savić S, Bogdanović M, Martinović T, Bumbaširević V, Stevović TK. Micromorphological features and interleukin 6, 8, and 18 expressions in post-mortem lung tissue in cases with acute respiratory distress syndrome. Forensic Sci Med Pathol 2024; 20:1-7. [PMID: 36809485 DOI: 10.1007/s12024-022-00572-4] [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] [Accepted: 12/29/2022] [Indexed: 02/23/2023]
Abstract
The purpose of this study was to analyze the presence of interleukins 6, 8, and 18 in post-mortem lung tissue of subjects deceased due to polytrauma. In addition to this, we have described different micromorphological features of lung tissue in ARDS cases associated with fatal traffic trauma. A total of 18 autopsy cases with ARDS after polytrauma and 15 control autopsy cases were analyzed in this study. From every subject, we collected one sample for each lung lobe. All of the histological sections were analyzed by using light microscopy, and for the purpose of ultrastructural analysis, we used transmission electron microscopy. Representative sections were further processed by way of immunohistochemistry analysis. Quantification of IL-6, IL-8, and IL-18-positive cells was conducted by applying the IHC score. We noticed that all samples of ARDS cases exhibited elements of the proliferative phase. Immunohistochemical analysis of lung tissue in patients with ARDS showed strong positive staining for IL-6 (2.8 ± 0.7), IL-8 (2.2 ± 1.3), and IL-18 (2.7 ± 1.2), while staining of the control samples resulted in no positivity to low/moderate positivity (for IL-6 1.4 ± 0.5; for IL-8 0.1 ± 0.4; for IL-18 0.6 ± 0.9). Only IL-6 correlated negatively with the patients' age (r = -0.6805, p < 0.01). In this study, we described microstructural changes in lung sections of ARDS cases and control cases, as well as interleukins' expression, demonstrating that autopsy material is as informing as tissue samples collected by performing open lung biopsy.
Collapse
Affiliation(s)
- Jelena Kuzmanović
- Pathology Department, University Medical Centre Zvezdara, Belgrade, Serbia
| | - Slobodan Savić
- Institute of Forensic Medicine "Dr Milovan Milovanovic", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Milenko Bogdanović
- Institute of Forensic Medicine "Dr Milovan Milovanovic", Faculty of Medicine, University of Belgrade, Belgrade, Serbia.
| | - Tamara Martinović
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vladimir Bumbaširević
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Tamara Kravić Stevović
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
4
|
Sanches Santos Rizzo Zuttion M, Moore SKL, Chen P, Beppu AK, Hook JL. New Insights into the Alveolar Epithelium as a Driver of Acute Respiratory Distress Syndrome. Biomolecules 2022; 12:biom12091273. [PMID: 36139112 PMCID: PMC9496395 DOI: 10.3390/biom12091273] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/02/2022] [Accepted: 09/08/2022] [Indexed: 11/29/2022] Open
Abstract
The alveolar epithelium serves as a barrier between the body and the external environment. To maintain efficient gas exchange, the alveolar epithelium has evolved to withstand and rapidly respond to an assortment of inhaled, injury-inducing stimuli. However, alveolar damage can lead to loss of alveolar fluid barrier function and exuberant, non-resolving inflammation that manifests clinically as acute respiratory distress syndrome (ARDS). This review discusses recent discoveries related to mechanisms of alveolar homeostasis, injury, repair, and regeneration, with a contemporary emphasis on virus-induced lung injury. In addition, we address new insights into how the alveolar epithelium coordinates injury-induced lung inflammation and review maladaptive lung responses to alveolar damage that drive ARDS and pathologic lung remodeling.
Collapse
Affiliation(s)
- Marilia Sanches Santos Rizzo Zuttion
- Women’s Guild Lung Institute, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sarah Kathryn Littlehale Moore
- Lung Imaging Laboratory, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Peter Chen
- Women’s Guild Lung Institute, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Andrew Kota Beppu
- Women’s Guild Lung Institute, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jaime Lynn Hook
- Lung Imaging Laboratory, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Correspondence:
| |
Collapse
|
5
|
Wang Y, Gao X, Li Y, Wang X, Li Y, Zhang S, Liu H, Guo H, Lu W, Sun D. Pulmonary surfactant-associated protein B regulates prostaglandin-endoperoxide synthase-2 and inflammation in chronic obstructive pulmonary disease. Exp Physiol 2021; 106:1303-1311. [PMID: 33729612 DOI: 10.1113/ep089244] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/10/2021] [Indexed: 02/06/2023]
Abstract
NEW FINDINGS What is the central question of this study? It is reported that polymorphism of the gene for pulmonary surfactant-associated protein B (SFTPB) is associated with chronic obstructive pulmonary disease (COPD): what are the function and mechanism of action of SFTPB in COPD? What is the main finding and its importance? Under stimulation of the risk factors of COPD, SFTPB expression is decreased, which may be involved in the formation of COPD. The progress of COPD induces an inflammatory response and reduces SFTPB expression. Levels of prostaglandin-endoperoxide synthase-2 (PTGS2) and inflammatory responses are changed by SFTPB, which indicates that SFTPB promotes the progression of COPD by PTGS2 and inflammation. ABSTRACT Pulmonary surfactant-associated protein B (SFTPB) is a critical protein for lung homeostasis, and polymorphism of its gene is associated with chronic obstructive pulmonary disease (COPD). However, few studies have so far confirmed the functional involvement of SFTPB in COPD. Serum SFTPB and inflammatory cytokine levels were measured in 54 patients with acute exacerbation of COPD and 29 healthy controls. A549 cells were induced using 10% cigarette smoke extract (CSE) and treated with dexamethasone to investigate the effect of inflammation on SFTPB expression, and the effect of SFTPB overexpression and silencing on inflammatory cytokines was measured using real-time PCR and enzyme-linked immunosorbent assay. SFTPB expression was assessed in mouse lung tissues using immunofluorescence. Serum levels of SFTPB were significantly lower in COPD patients than in controls (P = 0.009). Conversely, levels of interleukin (IL)-6 and prostaglandin-endoperoxide synthase-2 (PTGS2) were increased in COPD patients (IL-6: P = 0.006; PTGS2: P = 0.043). After CSE treatment, SFTPB mRNA and protein levels were significantly decreased compared to controls (mRNA: P = 0.002; protein: P = 0.011), while IL-6, IL-8 and PTGS2 were elevated. Dexamethasone treatment increased SFTPB levels. Following overexpression of SFTPB in A549 cells, mRNA and protein levels of IL-6, IL-8 and PTGS2 were significantly reduced, while gene silencing induced the opposite effect. SFTPB levels were significantly reduced in the lung tissue of a mouse model of COPD compared to controls. Reduced SFTPB levels may induce PTGS2 and inflammatory responses in COPD and SFTPB could be a key protein for evaluation of COPD progression.
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of National Health Commission for the Diagnosis & Treatment of COPD, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, China.,Graduate School, Baotou Medical College, Baotou, Inner Mongolia, China
| | - Xiaoyu Gao
- Key Laboratory of National Health Commission for the Diagnosis & Treatment of COPD, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, China
| | - Yuan Li
- Key Laboratory of National Health Commission for the Diagnosis & Treatment of COPD, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, China
| | - Xiao Wang
- Key Laboratory of National Health Commission for the Diagnosis & Treatment of COPD, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, China
| | - Yuanyuan Li
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Sainan Zhang
- Key Laboratory of National Health Commission for the Diagnosis & Treatment of COPD, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, China.,Graduate School, Baotou Medical College, Baotou, Inner Mongolia, China
| | - Hongyan Liu
- Key Laboratory of National Health Commission for the Diagnosis & Treatment of COPD, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, China.,Graduate School, Baotou Medical College, Baotou, Inner Mongolia, China
| | - Hui Guo
- Key Laboratory of National Health Commission for the Diagnosis & Treatment of COPD, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, China.,Graduate School, Baotou Medical College, Baotou, Inner Mongolia, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dejun Sun
- Key Laboratory of National Health Commission for the Diagnosis & Treatment of COPD, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, China.,Graduate School, Baotou Medical College, Baotou, Inner Mongolia, China
| |
Collapse
|
6
|
Galván-Román JM, Rodríguez-García SC, Roy-Vallejo E, Marcos-Jiménez A, Sánchez-Alonso S, Fernández-Díaz C, Alcaraz-Serna A, Mateu-Albero T, Rodríguez-Cortes P, Sánchez-Cerrillo I, Esparcia L, Martínez-Fleta P, López-Sanz C, Gabrie L, Del Campo Guerola L, Suárez-Fernández C, Ancochea J, Canabal A, Albert P, Rodríguez-Serrano DA, Aguilar JM, Del Arco C, de Los Santos I, García-Fraile L, de la Cámara R, Serra JM, Ramírez E, Alonso T, Landete P, Soriano JB, Martín-Gayo E, Fraile Torres A, Zurita Cruz ND, García-Vicuña R, Cardeñoso L, Sánchez-Madrid F, Alfranca A, Muñoz-Calleja C, González-Álvaro I. IL-6 serum levels predict severity and response to tocilizumab in COVID-19: An observational study. J Allergy Clin Immunol 2020; 147:72-80.e8. [PMID: 33010257 PMCID: PMC7525244 DOI: 10.1016/j.jaci.2020.09.018] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/07/2020] [Accepted: 09/17/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Patients with coronavirus disaese 2019 (COVID-19) can develop a cytokine release syndrome that eventually leads to acute respiratory distress syndrome requiring invasive mechanical ventilation (IMV). Because IL-6 is a relevant cytokine in acute respiratory distress syndrome, the blockade of its receptor with tocilizumab (TCZ) could reduce mortality and/or morbidity in severe COVID-19. OBJECTIVE We sought to determine whether baseline IL-6 serum levels can predict the need for IMV and the response to TCZ. METHODS A retrospective observational study was performed in hospitalized patients diagnosed with COVID-19. Clinical information and laboratory findings, including IL-6 levels, were collected approximately 3 and 9 days after admission to be matched with preadministration and postadministration of TCZ. Multivariable logistic and linear regressions and survival analysis were performed depending on outcomes: need for IMV, evolution of arterial oxygen tension/fraction of inspired oxygen ratio, or mortality. RESULTS One hundred forty-six patients were studied, predominantly males (66%); median age was 63 years. Forty-four patients (30%) required IMV, and 58 patients (40%) received treatment with TCZ. IL-6 levels greater than 30 pg/mL was the best predictor for IMV (odds ratio, 7.1; P < .001). Early administration of TCZ was associated with improvement in oxygenation (arterial oxygen tension/fraction of inspired oxygen ratio) in patients with high IL-6 (P = .048). Patients with high IL-6 not treated with TCZ showed high mortality (hazard ratio, 4.6; P = .003), as well as those with low IL-6 treated with TCZ (hazard ratio, 3.6; P = .016). No relevant serious adverse events were observed in TCZ-treated patients. CONCLUSIONS Baseline IL-6 greater than 30 pg/mL predicts IMV requirement in patients with COVID-19 and contributes to establish an adequate indication for TCZ administration.
Collapse
Affiliation(s)
- José María Galván-Román
- Internal Medicine Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Sebastián C Rodríguez-García
- Rheumatology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Emilia Roy-Vallejo
- Internal Medicine Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Ana Marcos-Jiménez
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Santiago Sánchez-Alonso
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Carlos Fernández-Díaz
- Rheumatology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Ana Alcaraz-Serna
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Tamara Mateu-Albero
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Pablo Rodríguez-Cortes
- Internal Medicine Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Ildefonso Sánchez-Cerrillo
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Laura Esparcia
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Pedro Martínez-Fleta
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Celia López-Sanz
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Ligia Gabrie
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Luciana Del Campo Guerola
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Carmen Suárez-Fernández
- Internal Medicine Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Julio Ancochea
- Pneumology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Alfonso Canabal
- Intensive Care Unit, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Patricia Albert
- Intensive Care Unit, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Diego A Rodríguez-Serrano
- Intensive Care Unit, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Juan Mariano Aguilar
- Emergency Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Carmen Del Arco
- Emergency Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Ignacio de Los Santos
- Internal Medicine Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Lucio García-Fraile
- Internal Medicine Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Rafael de la Cámara
- Hematology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - José María Serra
- Hospital Pharmacy Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Esther Ramírez
- Hospital Pharmacy Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Tamara Alonso
- Pneumology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Pedro Landete
- Pneumology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Joan B Soriano
- Pneumology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Enrique Martín-Gayo
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Arturo Fraile Torres
- Microbiology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Nelly Daniela Zurita Cruz
- Microbiology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Rosario García-Vicuña
- Rheumatology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Laura Cardeñoso
- Microbiology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Francisco Sánchez-Madrid
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain; Centro de Investigación Biomédica en Red - Enfermedades Cardiovasculares (CIBER CV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Arantzazu Alfranca
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Cecilia Muñoz-Calleja
- Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Isidoro González-Álvaro
- Rheumatology Service, Hospital Universitario de la Princesa, Universidad Autónoma Madrid, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain.
| | | |
Collapse
|
7
|
Stilhano RS, Costa AJ, Nishino MS, Shams S, Bartolomeo CS, Breithaupt-Faloppa AC, Silva EA, Ramirez AL, Prado CM, Ureshino RP. SARS-CoV-2 and the possible connection to ERs, ACE2, and RAGE: Focus on susceptibility factors. FASEB J 2020; 34:14103-14119. [PMID: 32965736 PMCID: PMC7537138 DOI: 10.1096/fj.202001394rr] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/15/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has provoked major stresses on the health-care systems of several countries, and caused the death of more than a quarter of a million people globally, mainly in the elderly population with preexisting pathologies. Previous studies with coronavirus (SARS-CoV) point to gender differences in infection and disease progression with increased susceptibility in male patients, indicating that estrogens may be associated with physiological protection against the coronavirus. Therefore, the objectives of this work are threefold. First, we aim to summarize the SARS-CoV-2 infection pathway and the roles both the virus and patient play in COVID-19 (Coronavirus disease 2019) progression, clinical symptomatology, and mortality. Second, we detail the effect estrogen has on viral infection and host infection response, including its role in both the regulation of key viral receptor expression and the mediation of inflammatory activity. Finally, we describe how ERs (estrogen receptors) and RAGE (receptor for advanced glycation end-products) play a critical role in metabolic pathways, which we envisage could maintain a close interplay with SARS-CoV and COVID-19 mortality rates, despite a current lack of research directly determining how. Taken together, we present the current state of the field regarding SARS-CoV-2 research and illuminate where research is needed to better define the role both estrogen and metabolic comorbidities have in the COVID-19 disease state, which can be key in screening potential therapeutic options as the search for effective treatments continue.
Collapse
Affiliation(s)
- Roberta Sessa Stilhano
- Department of Physiological Sciences, Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| | - Angelica Jardim Costa
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Michelle Sayuri Nishino
- Department of Biological Sciences, Universidade Federal de São Paulo, Diadema, Brazil.,Laboratory of Molecular and Translational Endocrinology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Shahin Shams
- Department of Biomedical Engineering, University of California, Davis, CA, USA
| | - Cynthia Silva Bartolomeo
- Department of Physiological Sciences, Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil.,Department of Biosciences, Universidade Federal de São Paulo, Santos, Brazil
| | - Ana Cristina Breithaupt-Faloppa
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulação (LIM-11), Instituto do Coração (InCor), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | | | - Carla Maximo Prado
- Department of Biosciences, Universidade Federal de São Paulo, Santos, Brazil
| | - Rodrigo Portes Ureshino
- Department of Biological Sciences, Universidade Federal de São Paulo, Diadema, Brazil.,Laboratory of Molecular and Translational Endocrinology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
8
|
Guérin C, Terzi N, Galerneau LM, Mezidi M, Yonis H, Baboi L, Kreitmann L, Turbil E, Cour M, Argaud L, Louis B. Lung and chest wall mechanics in patients with acute respiratory distress syndrome, expiratory flow limitation, and airway closure. J Appl Physiol (1985) 2020; 128:1594-1603. [PMID: 32352339 DOI: 10.1152/japplphysiol.00059.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tidal expiratory flow limitation (EFL), which may herald airway closure (AC), is a mechanism of loss of aeration in ARDS. In this prospective, short-term, two-center study, we measured static and dynamic chest wall (Est,cw and Edyn,cw) and lung (Est,L and Edyn,L) elastance with esophageal pressure, EFL, and AC at 5 cmH2O positive end-expiratory pressure (PEEP) in intubated, sedated, and paralyzed ARDS patients. For EFL determination, we used the atmospheric method and a new device allowing comparison of tidal flow during expiration to PEEP and to atmosphere. AC was validated when airway opening pressure (AOP) assessed from volume-pressure curve was found greater than PEEP by at least 1 cmH2O. EFL was defined whenever flow did not increase between exhalation to PEEP and to atmosphere over all or part of expiration. Elastance values were expressed as percentage of normal predicted values (%N). Among the 25 patients included, eight had EFL (32%) and 13 AOP (52%). Between patients with and without EFL Edyn,cw [median (1st to 3rd quartiles)] was 70 (16-127) and 102 (70-142) %N (P = 0.32) and Edyn,L338 (332-763) and 224 (160-275) %N (P < 0.001). The corresponding values for Est,cw and Est,L were 70 (56-88) and 85 (64-103) %N (P = 0.35) and 248 (206-348) and 170 (144-195) (P = 0.02), respectively. Dynamic EL had an area receiver operating characteristic curve of 0.88 [95% confidence intervals 0.83-0.92] for EFL and 0.74[0.68-0.79] for AOP. Higher Edyn,L was accurate to predict EFL in ARDS patients; AC can occur independently of EFL, and both should be assessed concurrently in ARDS patients.NEW & NOTEWORTHY Expiratory flow limitation (EFL) and airway closure (AC) were observed in 32% and 52%, respectively, of 25 patients with ARDS investigated during mechanical ventilation in supine position with a positive end-expiratory pressure of 5 cmH2O. The performance of dynamic lung elastance to detect expiratory flow limitation was good and better than that to detect airway closure. The vast majority of patients with EFL also had AC; however, AC can occur in the absence of EFL.
Collapse
Affiliation(s)
- Claude Guérin
- Medecine Intensive-Réanimation, Groupement Hospitalier Centre, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France.,Université de Lyon, Lyon, France.,Institut Mondor de Recherches Biomédicales INSERM 955 CNRS ERL 7000, Créteil, France
| | - Nicolas Terzi
- Medecine Intensive-Réanimation, CHU Grenoble-Alpes, Grenoble, France.,Université de Grenoble-Alpes, Grenoble, France
| | - Louis-Marie Galerneau
- Medecine Intensive-Réanimation, CHU Grenoble-Alpes, Grenoble, France.,Université de Grenoble-Alpes, Grenoble, France
| | - Mehdi Mezidi
- Université de Lyon, Lyon, France.,Médecine Intensive-Réanimation, Groupement Hospitalier Nord, Hospices Civils de Lyon, Lyon, France
| | - Hodane Yonis
- Médecine Intensive-Réanimation, Groupement Hospitalier Nord, Hospices Civils de Lyon, Lyon, France
| | - Loredana Baboi
- Médecine Intensive-Réanimation, Groupement Hospitalier Nord, Hospices Civils de Lyon, Lyon, France
| | - Louis Kreitmann
- Medecine Intensive-Réanimation, Groupement Hospitalier Centre, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France.,Université de Lyon, Lyon, France
| | - Emanuele Turbil
- Department of Anesthesia and Critical Care, University of Sassari, Sassari, Italy
| | - Martin Cour
- Medecine Intensive-Réanimation, Groupement Hospitalier Centre, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France.,Université de Lyon, Lyon, France
| | - Laurent Argaud
- Medecine Intensive-Réanimation, Groupement Hospitalier Centre, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France.,Université de Lyon, Lyon, France
| | - Bruno Louis
- Institut Mondor de Recherches Biomédicales INSERM 955 CNRS ERL 7000, Créteil, France
| |
Collapse
|
9
|
Breithaupt-Faloppa AC, Correia CDJ, Prado CM, Stilhano RS, Ureshino RP, Moreira LFP. 17β-Estradiol, a potential ally to alleviate SARS-CoV-2 infection. Clinics (Sao Paulo) 2020; 75:e1980. [PMID: 32490931 PMCID: PMC7233687 DOI: 10.6061/clinics/2020/e1980] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 05/05/2020] [Indexed: 01/08/2023] Open
Abstract
Considering that female sexual hormones may modulate the inflammatory response and also exhibit direct effects on the cells of the immune system, herein, we intend to discuss the sex differences and the role of estradiol in modulating the lung and systemic inflammatory response, focusing on its possible application as a treatment modality for SARS-CoV-2 patients. COVID-19 patients develop severe hypoxemia early in the course of the disease, which is silent most of the time. Small fibrinous thrombi in pulmonary arterioles and a tumefaction of endothelial were observed in the autopsies of fatal COVID-19 cases. Studies showed that the viral infection induces a vascular process in the lung, which included vasodilation and endothelial dysfunction. Further, the proportions of CD4+ T and CD8+ T lymphocytes were strongly reduced in patients with severe SARS-CoV-2 infection. Estradiol is connected with CD4+ T cell numbers and increases T-reg cell populations, affecting immune responses to infection. It is known that estradiol exerts a protective effect on endothelial function, activating the generation of nitric oxide (NO) via endothelial nitric oxide synthase. Estrogen attenuates the vasoconstrictor response to various stimuli and induces vasodilation in the pulmonary vasculature during stress situations like hypoxia. It exerts a variety of rapid actions, which are initiated after its coupling with membrane receptors, which in turn, may positively modulate vascular responses in pulmonary disease and help to maintain microvascular flow. Direct and indirect mechanisms underlying the effects of estradiol were investigated, and the results point to a possible protective effect of estradiol against COVID-19, indicating that it may be considered as an adjuvant therapeutic element for the treatment of patients affected by the novel coronavirus.
Collapse
Affiliation(s)
- Ana Cristina Breithaupt-Faloppa
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR
- *Corresponding author. E-mail:
| | - Cristiano de Jesus Correia
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Carla Máximo Prado
- Instituto de Saude e Sociedade (ISS), Universidade Federal de Sao Paulo (UNIFESP), Santos, SP, BR
| | | | - Rodrigo Portes Ureshino
- Departamento de Ciencias Biologicas, Universidade Federal de Sao Paulo (UNIFESP), Diadema, SP, BR
- Laboratorio de Endocrinologia Molecular e Translacional, Escola Paulista de Medicina, Universidade Federal de Sao Paulo (UNIFESP), Sao Paulo, SP, BR
| | - Luiz Felipe Pinho Moreira
- Laboratorio de Cirurgia Cardiovascular e Fisiopatologia da Circulacao (LIM-11), Instituto do Coracao (InCor), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, BR
| |
Collapse
|
10
|
Spengler D, Rintz N, Krause MF. An Unsettled Promise: The Newborn Piglet Model of Neonatal Acute Respiratory Distress Syndrome (NARDS). Physiologic Data and Systematic Review. Front Physiol 2019; 10:1345. [PMID: 31736777 PMCID: PMC6831728 DOI: 10.3389/fphys.2019.01345] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022] Open
Abstract
Despite great advances in mechanical ventilation and surfactant administration for the newborn infant with life-threatening respiratory failure no specific therapies are currently established to tackle major pro-inflammatory pathways. The susceptibility of the newborn infant with neonatal acute respiratory distress syndrome (NARDS) to exogenous surfactant is linked with a suppression of most of the immunologic responses by the innate immune system, however, additional corticosteroids applied in any severe pediatric lung disease with inflammatory background do not reduce morbidity or mortality and may even cause harm. Thus, the neonatal piglet model of acute lung injury serves as an excellent model to study respiratory failure and is the preferred animal model for reasons of availability, body size, similarities of porcine and human lung, robustness, and costs. In addition, similarities to the human toll-like receptor 4, the existence of intraalveolar macrophages, the sensitivity to lipopolysaccharide, and the production of nitric oxide make the piglet indispensable in anti-inflammatory research. Here we present the physiologic and immunologic data of newborn piglets from three trials involving acute lung injury secondary to repeated airway lavage (and others), mechanical ventilation, and a specific anti-inflammatory intervention via the intratracheal route using surfactant as a carrier substance. The physiologic data from many organ systems of the newborn piglet—but with preference on the lung—are presented here differentiating between baseline data from the uninjured piglet, the impact of acute lung injury on various parameters (24 h), and the follow up data after 72 h of mechanical ventilation. Data from the control group and the intervention groups are listed separately or combined. A systematic review of the newborn piglet meconium aspiration model and the repeated airway lavage model is finally presented. While many studies assessed lung injury scores, leukocyte infiltration, and protein/cytokine concentrations in bronchoalveolar fluid, a systematic approach to tackle major upstream pro-inflammatory pathways of the innate immune system is still in the fledgling stages. For the sake of newborn infants with life-threatening NARDS the newborn piglet model still is an unsettled promise offering many options to conquer neonatal physiology/immunology and to establish potent treatment modalities.
Collapse
Affiliation(s)
- Dietmar Spengler
- Department of Pediatrics, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | - Nele Rintz
- Department of Pediatrics, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | - Martin F Krause
- Department of Pediatrics, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
11
|
Structural alterations and markers of endothelial activation in pulmonary and bronchial arteries in fatal asthma. Allergy Asthma Clin Immunol 2019; 15:50. [PMID: 31485240 PMCID: PMC6714380 DOI: 10.1186/s13223-019-0363-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 08/19/2019] [Indexed: 12/15/2022] Open
Abstract
Background There is interest in better understanding vessel pathology in asthma, given the findings of loss of peripheral vasculature associated with disease severity by imaging and altered markers of endothelial activation. To date, vascular changes in asthma have been described mainly at the submucosal capillary level of the bronchial microcirculation, with sparse information available on the pathology of bronchial and pulmonary arteries. The aim of this study was to describe structural and endothelial activation markers in bronchial arteries (BAs) and pulmonary arteries (PAs) of asthma patients who died during a fatal asthma attack. Methods Autopsy lung tissue was obtained from 21 smoking and non-smoking patients who died of an asthma attack and nine non-smoking control patients. Verhoeff–Masson trichrome staining was used to analyse the structure of arteries. Using immuno-histochemistry and image analyses, we quantified extracellular matrix (ECM) components (collagen I, collagen III, versican, tenascin, fibronectin, elastic fibres), adhesion molecules [vascular cell adhesion molecule 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1)] and markers of vascular tone/dysfunction [endothelin-1 (ET-1) and angiotensin II type 2 receptor (AT2)] in PAs and BAs. Results There were no significant differences in ECM components, ICAM-1, ET-1 or AT2 between asthma patients and controls. Smoking asthma patients presented with decreased content of collagen III in both BA (p = 0.046) and PA (p = 0.010) walls compared to non-smoking asthma patients. Asthma patients had increased VCAM-1 content in the BA wall (p = 0.026) but not in the PA wall. Conclusion Our data suggest that the mechanisms linking asthma and arterial functional abnormalities might involve systemic rather than local mediators. Loss of collagen III in the PA was observed in smoking asthma patients, and this was compatible with the degradative environment induced by cigarette smoking. Our data also reinforce the idea that the mechanisms of leukocyte efflux via adhesion molecules differ between bronchial and pulmonary circulation, which might be relevant to understanding and treating the distal lung in asthma.
Collapse
|
12
|
The Effects of Leukocyte Filtration on Cell Salvaged Autologous Blood Transfusion on Lung Function and Lung Inflammatory and Oxidative Stress Reactions in Elderly Patients Undergoing Lumbar Spinal Surgery. J Neurosurg Anesthesiol 2019; 31:36-42. [DOI: 10.1097/ana.0000000000000495] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
13
|
Roca O, García-de-Acilu M, Soriano-Navarro T, Griffiths MJD. Airway Alterations and Diffuse Alveolar Damage in Acute Respiratory Distress Syndrome: Is There Any Association? Arch Bronconeumol 2018; 55:3-4. [PMID: 29861071 DOI: 10.1016/j.arbres.2018.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Oriol Roca
- Critical Care Department, Hospital Universitari Vall d'Hebron, Institut de Recerca Vall d'Hebron, Barcelona, Spain; Ciber Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.
| | - Marina García-de-Acilu
- Critical Care Department, Hospital Universitari Vall d'Hebron, Institut de Recerca Vall d'Hebron, Barcelona, Spain
| | | | - Mark J D Griffiths
- Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom; Imperial College London, London, United Kingdom
| |
Collapse
|
14
|
Ortiz G, Garay M, Capelozzi V, Cardinal-Fernández P. Airway Pathological Alterations Selectively Associated With Acute Respiratory Distress Syndrome and Diffuse Alveolar Damage - Narrative Review. Arch Bronconeumol 2018; 55:31-37. [PMID: 29853259 DOI: 10.1016/j.arbres.2018.03.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 02/16/2018] [Accepted: 03/07/2018] [Indexed: 12/12/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is a frequent and life-threatening entity. Recently, it has been demonstrated that diffuse alveolar damage (DAD), which is considered the histological hallmark in spite of presenting itself in only half of living patients with ARDS, exerts a relevant effect in the ARDS outcome. Despite the fact that the bronchial tree constitutes approximately 1% of the lung volume, discovering a relation between DAD and bronchial tree findings could be of paramount importance for a few reasons; (a) it could improve the description of ARDS with DAD as a clinical-pathological entity, (b) it could subrogate DAD findings with the advantage of their more accessible and safer analysis and (c) it could allow the discovery of new therapeutic targets. This narrative review is focused on pathological airway changes associated to Diffuse Alveolar Damage in the context of Acute Respiratory Distress Syndrome. It is organized into five sections: main anatomical and functional features of the human airway, why it is necessary to study airway features associated to DAD in patients with ARDS, pathological airway changes associated with DAD in animal models of ARDS, pathological airway changes associated with DAD in patients with ARDS, and the newest techniques for studying the histology of the bronchial tree and lung parenchyma.
Collapse
Affiliation(s)
- Guillermo Ortiz
- Universidad del Bosque, Bogotá, Colombia; Universidad de Barcelona, Barcelona, Spain
| | | | - Vera Capelozzi
- Department of Pathology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Pablo Cardinal-Fernández
- Emergency Department, Hospital Universitario HM Sanchinarro, Madrid, Spain; HM Research Foundation, Madrid, Spain.
| |
Collapse
|
15
|
Nascimento ECTD, Baldi BG, Mariani AW, Annoni R, Kairalla RA, Pimenta SP, da Silva LFF, Carvalho CRR, Dolhnikoff M. Immunohistological features related to functional impairment in lymphangioleiomyomatosis. Respir Res 2018; 19:83. [PMID: 29739412 PMCID: PMC5941479 DOI: 10.1186/s12931-018-0797-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 04/29/2018] [Indexed: 12/25/2022] Open
Abstract
Background Lymphangioleiomyomatosis (LAM) is a low-grade neoplasm characterized by the pulmonary infiltration of smooth muscle-like cells (LAM cells) and cystic destruction. Patients usually present with airway obstruction in pulmonary function tests (PFTs). Previous studies have shown correlations among histological parameters, lung function abnormalities and prognosis in LAM. We investigated the lung tissue expression of proteins related to the mTOR pathway, angiogenesis and enzymatic activity and its correlation with functional parameters in LAM patients. Methods We analyzed morphological and functional parameters of thirty-three patients. Two groups of disease severity were identified according to FEV1 values. Lung tissue from open biopsies or lung transplants was immunostained for SMA, HMB-45, mTOR, VEGF-D, MMP-9 and D2-40. Density of cysts, density of nodules and protein expression were measured by image analysis and correlated with PFT parameters. Results There was no difference in the expression of D2-40 between the more severe and the less severe groups. All other immunohistological parameters showed significantly higher values in the more severe group (p ≤ 0.002). The expression of VEGF-D, MMP-9 and mTOR in LAM cells was associated with the density of both cysts and nodules. The density of cysts and nodules as well as the expression of MMP-9 and VEGF-D were associated with the impairment of PFT parameters. Conclusions Severe LAM represents an active phase of the disease with high expression of VEGF-D, mTOR, and MMP-9, as well as LAM cell infiltration. Our findings suggest that the tissue expression levels of VEGF-D and MMP-9 are important parameters associated with the loss of pulmonary function and could be considered as potential severity markers in open lung biopsies of LAM patients.
Collapse
Affiliation(s)
- Ellen Caroline Toledo do Nascimento
- Departamento de Patologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Av. Dr. Arnaldo, 455, room 1155, Cerqueira Cesar, CEP 01246-903, Sao Paulo, SP, Brazil.
| | - Bruno Guedes Baldi
- Divisao de Pneumologia, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Alessandro Wasum Mariani
- Departamento de Cirurgia Toracica, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Raquel Annoni
- Departamento de Patologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Av. Dr. Arnaldo, 455, room 1155, Cerqueira Cesar, CEP 01246-903, Sao Paulo, SP, Brazil
| | - Ronaldo Adib Kairalla
- Divisao de Pneumologia, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Suzana Pinheiro Pimenta
- Divisao de Pneumologia, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Luiz Fernando Ferraz da Silva
- Departamento de Patologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Av. Dr. Arnaldo, 455, room 1155, Cerqueira Cesar, CEP 01246-903, Sao Paulo, SP, Brazil
| | - Carlos Roberto Ribeiro Carvalho
- Divisao de Pneumologia, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Marisa Dolhnikoff
- Departamento de Patologia, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Av. Dr. Arnaldo, 455, room 1155, Cerqueira Cesar, CEP 01246-903, Sao Paulo, SP, Brazil
| |
Collapse
|
16
|
Surfactant Proteins SP-A and SP-D Ameliorate Pneumonia Severity and Intestinal Injury in a Murine Model of Staphylococcus Aureus Pneumonia. Shock 2018; 46:164-72. [PMID: 26849628 DOI: 10.1097/shk.0000000000000587] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
UNLABELLED Staphylococcus aureus pneumonia is an important cause of sepsis which causes gut injury, inflammation, and apoptosis. The surfactant proteins surfactant protein A (SP-A) and surfactant protein D (SP-D) bind bacterial pathogens and facilitate clearance of pathogens, apoptotic bodies, and modulate immune responses. SP-A and SP-D are expressed in both lung and gut epithelia. We hypothesize SP-A and SP-D regulate pneumonia severity and gut injury during pneumonia. METHODS Wild-type (WT) and SP-A and SP-D double knockout (SP-A/D KO) mice were subjected to S. aureus or sham pneumonia. Bronchoalveolar lavage and tissue harvest were performed 24 h later. Pneumonia severity, gut mucosal injury, inflammation, and apoptosis were measured using a combination of histology, immunohistochemistry, cytokine assay, TUNEL assay, quantitative real-time polymerase chain reaction, and Western blot analyses. RESULTS Pneumonia increased gut inflammation, apoptosis, and mucosal injury in both groups. Pneumonia histology and bacterial growth in bronchoalveolar lavage fluid demonstrate more severe infection in SP-A/D KO mice compared with WT controls. SP-A/D KO mice with pneumonia also demonstrate more severe histologic gut mucosal injury, increased gut apoptosis, elevated caspase-3 levels, and Bax/Bcl-2 mRNA expression compared with WT pneumonia mice. Nuclear factor κB (NF-κB) p65 expression and its nuclear translocation, gut levels of tumor necrosis factor α and interleukin-1β were all increased in SP-A/D KO mice with pneumonia compared with WT controls. CONCLUSIONS These data provide evidence SP-A and SP-D attenuate S. aureus pneumonia severity resulting in decreased intestinal mucosal injury, apoptosis, and inflammation. Improved pulmonary clearance of S. aureus decreased caspase-3 and Bax/Bcl-2 expressions and decreased activation of the NF-κB signaling pathway in intestine represent potential mechanisms for the effects of SP-A and SP-D on gut injury during pneumonia.
Collapse
|
17
|
Buttignol M, Pires-Neto RC, Rossi E Silva RC, Albino MB, Dolhnikoff M, Mauad T. Airway and parenchyma immune cells in influenza A(H1N1)pdm09 viral and non-viral diffuse alveolar damage. Respir Res 2017; 18:147. [PMID: 28774302 PMCID: PMC5543730 DOI: 10.1186/s12931-017-0630-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 07/26/2017] [Indexed: 12/31/2022] Open
Abstract
Background Diffuse alveolar damage (DAD), which is the histological surrogate for acute respiratory distress syndrome (ARDS), has a multifactorial aetiology. Therefore it is possible that the immunopathology differs among the various presentations of DAD. The aim of this study is to compare lung immunopathology of viral (influenza A(H1N1)pdm09) to non-viral, extrapulmonary aetiologies in autopsy cases with DAD. Methods The lung tissue of 44 patients, was divided in the H1N1 group (n = 15) characterized by severe pulmonary injury due to influenza A(H1N1)pdm09 infection; the ARDS group (n = 13), characterized by patients with DAD due to non-pulmonary causes; and the Control group (n = 16), consisting of patients with non-pulmonary causes of death. Immunohistochemistry and image analysis were used to quantify, in the parenchyma and small airways, several immune cell markers. Results Both DAD groups had higher expression of neutrophils and macrophages in parenchyma and small airways. However, there was a higher expression of CD4+ and CD8+ T lymphocytes, CD83+ dendritic cells, granzyme A+ and natural killer + cell density in the lung parenchyma of the H1N1 group (p < 0.05). In the small airways, there was a lower cell density of tryptase + mast cells and dendritic + cells and an increase of IL-17 in both DAD groups (p < 0.05). Conclusion DAD due to viral A(H1N1)pdm09 is associated with a cytotoxic inflammatory phenotype, with partially divergent responses in the parenchyma relative to the small airways. In non-viral DAD, main immune cell alterations were found at the small airway level, reinforcing the role of the small airways in the pathogenesis of the exudative phase of DAD. Electronic supplementary material The online version of this article (doi:10.1186/s12931-017-0630-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Monique Buttignol
- Departament of Pathology, University of São Paulo - School of Medicine (FMUSP), Av. Dr. Arnaldo, 455 - 1 andar, sala 1155, São Paulo, SP, 01246903, Brazil.
| | - Ruy Camargo Pires-Neto
- Departament of Pathology, University of São Paulo - School of Medicine (FMUSP), Av. Dr. Arnaldo, 455 - 1 andar, sala 1155, São Paulo, SP, 01246903, Brazil
| | - Renata Calciolari Rossi E Silva
- Departament of Pathology, University of São Paulo - School of Medicine (FMUSP), Av. Dr. Arnaldo, 455 - 1 andar, sala 1155, São Paulo, SP, 01246903, Brazil
| | - Marina Ballarin Albino
- Departament of Pathology, University of São Paulo - School of Medicine (FMUSP), Av. Dr. Arnaldo, 455 - 1 andar, sala 1155, São Paulo, SP, 01246903, Brazil
| | - Marisa Dolhnikoff
- Departament of Pathology, University of São Paulo - School of Medicine (FMUSP), Av. Dr. Arnaldo, 455 - 1 andar, sala 1155, São Paulo, SP, 01246903, Brazil
| | - Thais Mauad
- Departament of Pathology, University of São Paulo - School of Medicine (FMUSP), Av. Dr. Arnaldo, 455 - 1 andar, sala 1155, São Paulo, SP, 01246903, Brazil
| |
Collapse
|
18
|
Chang HY, Li F, Li FS, Zheng CZ, Lei YZ, Wang J. Genetic Polymorphisms of SP-A, SP-B, and SP-D and Risk of Respiratory Distress Syndrome in Preterm Neonates. Med Sci Monit 2016; 22:5091-5100. [PMID: 28011976 PMCID: PMC5207009 DOI: 10.12659/msm.898553] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Background We examined selected polymorphisms in 3 pulmonary surfactant-associated proteins (SP) for their influence on serum SP levels and risk of respiratory distress syndrome (RDS) in preterm neonates. Material/Methods Premature infants from a Han population were enrolled, including 100 premature infants with RDS (case group) and 120 premature infants without RDS (control group). SNP genotyping for SP-A (+186A/G and +655C/T), SP-B (−18A/C and 1580C/T), and SP-D (Met11ThrT/C and Ala160ThrG/A) used polymerase chain reaction-restriction fragment length polymorphism. Haplotypes were calculated with Shesis software and serum SP-A/B/D levels were quantified by ELISA. Results Case and control groups exhibited significant differences in genotype and allele frequencies of SP-A (+186A/G, +655C/T) and SP-B (1580C/T). However, no statistically significant differences were observed in the allele and genotype frequencies of SP-B −18A/C, SP-D Met11ThrT/C, and SP-D Ala160ThrG/A. Importantly, serum SP-A and SP-B levels were reduced in RDS patients carrying SP-A (+186A/G, +655C/T) and SP-B (1580C/T) polymorphisms. AA genotype of +186A/G, SP-A level, and CC genotype of 1580C/T were independently correlated with increased RDS risk. Conclusions SP-A (+186A/G) and SP-B (1580C/T) polymorphisms are strongly associated with the risk of RDS in preterm infants. Notably, reduced serum SP-A levels were correlated with a high risk of RDS and may serve as novel biomarkers for RDS detection and monitoring.
Collapse
Affiliation(s)
- Hong-Yu Chang
- Department of Pediatrics, The General Hospital of the PLA Rocket Force, Beijing, China (mainland)
| | - Fang Li
- Department of Pediatrics, The General Hospital of the PLA Rocket Force, Beijing, China (mainland)
| | - Feng-Sheng Li
- Central Laboratory, The General Hospital of the PLA Rocket Force, Beijing, China (mainland)
| | - Cheng-Zhong Zheng
- Department of Pediatrics, The 306 Hospital of PLA, Beijing, China (mainland)
| | - Yan-Zhe Lei
- Department of Neonatology, Haidian Maternal and Child Healthcare Hospital, Beijing, China (mainland)
| | - Jing Wang
- Department of Neonatology, Nanjing Tongren Hospital, Nanjing, China (mainland)
| |
Collapse
|
19
|
Pires-Neto RC, Del Carlo Bernardi F, Alves de Araujo P, Mauad T, Dolhnikoff M. The Expression of Water and Ion Channels in Diffuse Alveolar Damage Is Not Dependent on DAD Etiology. PLoS One 2016; 11:e0166184. [PMID: 27835672 PMCID: PMC5106024 DOI: 10.1371/journal.pone.0166184] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 10/24/2016] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Aquaporins and ion channels are membrane proteins that facilitate the rapid movement of water and solutes across biological membranes. Experimental and in vitro studies reported that the function of these channels and pulmonary edema resolution are impaired in acute lung injury (ALI). Although current evidence indicates that alveolar fluid clearance is impaired in patients with ALI/diffuse alveolar damage (DAD), few human studies have addressed the alterations in pulmonary channels in this clinical condition. Additionally, it is not known whether the primary cause of DAD is a relevant variable for the channel dysfunction. METHODS Autopsied lungs of 43 patients with acute respiratory failure (ARF) due to DAD of three different etiologies, non-pulmonary sepsis, H1N1 viral infection and leptospirosis, were compared to 18 normal lungs. We quantified the expression of aquaporin (AQP) 1, AQP3, AQP5, epithelial Na+ channel (ENaC) and sodium potassium ATPase (Na-K-ATPase) in the alveolar septum using immunohistochemistry and image analysis. RESULTS The DAD group presented with increased expression of AQP3, AQP5 and Na-K-ATPase and decreased expression of ENaC compared to controls. However, there was no difference in protein expression within the DAD groups of different etiologies. CONCLUSION Water and ion channels are altered in patients with ARF due to DAD. The cause of DAD does not seem to influence the level of impairment of these channels.
Collapse
Affiliation(s)
- Ruy Camargo Pires-Neto
- Departamento de Patologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | | | - Priscila Alves de Araujo
- Departamento de Patologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - Thais Mauad
- Departamento de Patologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| | - Marisa Dolhnikoff
- Departamento de Patologia da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
20
|
Blondonnet R, Constantin JM, Sapin V, Jabaudon M. A Pathophysiologic Approach to Biomarkers in Acute Respiratory Distress Syndrome. DISEASE MARKERS 2016; 2016:3501373. [PMID: 26980924 PMCID: PMC4766331 DOI: 10.1155/2016/3501373] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 01/10/2016] [Indexed: 01/10/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is an acute-onset hypoxic condition with radiographic bilateral lung infiltration. It is characterized by an acute exudative phase combining diffuse alveolar damage and lung edema followed by a later fibroproliferative phase. Despite an improved understanding of ARDS pathobiology, our ability to predict the development of ARDS and risk-stratify patients with the disease remains limited. Biomarkers may help to identify patients at the highest risk of developing ARDS, assess response to therapy, predict outcome, and optimize enrollment in clinical trials. After a short description of ARDS pathobiology, here, we review the scientific evidence that supports the value of various ARDS biomarkers with regard to their major biological roles in ARDS-associated lung injury and/or repair. Ongoing research aims at identifying and characterizing novel biomarkers, in order to highlight relevant mechanistic explorations of lung injury and repair, and to ultimately develop innovative therapeutic approaches for ARDS patients. This review will focus on the pathophysiologic, diagnostic, and therapeutic implications of biomarkers in ARDS and on their utility to ultimately improve patient care.
Collapse
Affiliation(s)
- Raiko Blondonnet
- CHU Clermont-Ferrand, Intensive Care Unit, Department of Perioperative Medicine, Estaing University Hospital, 63000 Clermont-Ferrand, France
- Clermont Université, Université d'Auvergne, EA 7281, R2D2, 63000 Clermont-Ferrand, France
| | - Jean-Michel Constantin
- CHU Clermont-Ferrand, Intensive Care Unit, Department of Perioperative Medicine, Estaing University Hospital, 63000 Clermont-Ferrand, France
- Clermont Université, Université d'Auvergne, EA 7281, R2D2, 63000 Clermont-Ferrand, France
| | - Vincent Sapin
- Clermont Université, Université d'Auvergne, EA 7281, R2D2, 63000 Clermont-Ferrand, France
- Department of Medical Biochemistry and Molecular Biology, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - Matthieu Jabaudon
- CHU Clermont-Ferrand, Intensive Care Unit, Department of Perioperative Medicine, Estaing University Hospital, 63000 Clermont-Ferrand, France
- Clermont Université, Université d'Auvergne, EA 7281, R2D2, 63000 Clermont-Ferrand, France
| |
Collapse
|
21
|
Shah RJ, Diamond JM, Cantu E, Flesch J, Lee JC, Lederer DJ, Lama VN, Orens J, Weinacker A, Wilkes DS, Roe D, Bhorade S, Wille KM, Ware LB, Palmer SM, Crespo M, Demissie E, Sonnet J, Shah A, Kawut SM, Bellamy SL, Localio AR, Christie JD. Objective Estimates Improve Risk Stratification for Primary Graft Dysfunction after Lung Transplantation. Am J Transplant 2015; 15:2188-96. [PMID: 25877792 PMCID: PMC4721238 DOI: 10.1111/ajt.13262] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 02/02/2015] [Accepted: 02/07/2015] [Indexed: 01/25/2023]
Abstract
Primary graft dysfunction (PGD) is a major cause of early mortality after lung transplant. We aimed to define objective estimates of PGD risk based on readily available clinical variables, using a prospective study of 11 centers in the Lung Transplant Outcomes Group (LTOG). Derivation included 1255 subjects from 2002 to 2010; with separate validation in 382 subjects accrued from 2011 to 2012. We used logistic regression to identify predictors of grade 3 PGD at 48/72 h, and decision curve methods to assess impact on clinical decisions. 211/1255 subjects in the derivation and 56/382 subjects in the validation developed PGD. We developed three prediction models, where low-risk recipients had a normal BMI (18.5-25 kg/m(2) ), chronic obstructive pulmonary disease/cystic fibrosis, and absent or mild pulmonary hypertension (mPAP<40 mmHg). All others were considered higher-risk. Low-risk recipients had a predicted PGD risk of 4-7%, and high-risk a predicted PGD risk of 15-18%. Adding a donor-smoking lung to a higher-risk recipient significantly increased PGD risk, although risk did not change in low-risk recipients. Validation demonstrated that probability estimates were generally accurate and that models worked best at baseline PGD incidences between 5% and 25%. We conclude that valid estimates of PGD risk can be produced using readily available clinical variables.
Collapse
Affiliation(s)
- Rupal J. Shah
- Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania School of Medicine, Philadelphia, PA
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Joshua M. Diamond
- Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania School of Medicine, Philadelphia, PA
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Edward Cantu
- Division of Cardiovascular Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA, Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University College of Physicians and Surgeons, New York, New York
| | - Judd Flesch
- Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania School of Medicine, Philadelphia, PA
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - James C. Lee
- Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - David J. Lederer
- Division of Cardiovascular Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA, Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University College of Physicians and Surgeons, New York, New York
| | - Vibha N. Lama
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan
| | - Jonathon Orens
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Johns Hopkins University Hospital, Baltimore, Maryland
| | - Ann Weinacker
- Department of Pulmonary and Critical Care, Stanford University, Palo Alto, CA
| | - David S. Wilkes
- Division of Pulmonary, Allergy, and Critical Care Medicine, Indiana University School of Medicine, Indianapolis, IN
| | | | - Sangeeta Bhorade
- Division of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, Illinois
| | - Keith M. Wille
- Division of Pulmonary and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Lorraine B. Ware
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Scott M. Palmer
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University, Raleigh-Durham, North Carolina
| | - Maria Crespo
- Division of Pulmonary, Allergy, and Critical Care, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ejigayehu Demissie
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Joshua Sonnet
- Department Surgery, Columbia University College of Physicians and Surgeons, New York, New York
| | - Ashish Shah
- Department of Surgery, Johns Hopkins University Hospital, Baltimore, Maryland
| | - Steven M. Kawut
- Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania School of Medicine, Philadelphia, PA
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Scarlett L. Bellamy
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - A. Russell Localio
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, PA
| | - Jason D. Christie
- Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania School of Medicine, Philadelphia, PA
- Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania School of Medicine, Philadelphia, PA
| |
Collapse
|
22
|
Oliveira CBD, Vasconcellos C, Sakai-Valente NY, Sotto MN, Luiz FG, Belda Júnior W, Sousa MDGTD, Benard G, Criado PR. Toll-like receptors (TLR) 2 and 4 expression of keratinocytes from patients with localized and disseminated dermatophytosis. Rev Inst Med Trop Sao Paulo 2015; 57:57-61. [PMID: 25651327 PMCID: PMC4325524 DOI: 10.1590/s0036-46652015000100008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 08/06/2014] [Indexed: 12/21/2022] Open
Abstract
There are few studies on the role of innate immune response in
dermatophytosis. An investigation was conducted to define the involvement of
Toll-Like Receptors (TLRs) 2 and 4 in localized (LD) and disseminated (DD)
dermatophytosis due to T. rubrum. Fifteen newly diagnosed patients,
eight patients with LD and seven with DD, defined by involvement of at least three
body segments were used in this study. Controls comprised twenty skin samples from
healthy individuals undergoing plastic surgery. TLR2 and TLR4 were quantified in skin
lesions by immunohistochemistry. A reduced expression of TLR4 in the lower and upper
epidermis of both LD and DD patients was found compared to controls; TLR2 expression
was preserved in the upper and lower epidermis of all three groups. As TLR4 signaling
induces the production of inflammatory cytokines and neutrophils recruitment, its
reduced expression likely contributed to the lack of resolution of the infection and
the consequent chronic nature of the dermatophytosis. As TLR2 expression acts to
limit the inflammatory process and preserves the epidermal structure, its preserved
expression may also contribute to the persistent infection and limited inflammation
that are characteristic of dermatophytic infections.
Collapse
Affiliation(s)
| | - Cídia Vasconcellos
- Department of Dermatology, Medical School, University of Sao Paulo, São Paulo, SP, Brazil
| | - Neusa Y Sakai-Valente
- Laboratory of Medical Investigation Unit 53, Division of Clinical Dermatology, Medical School, University of Sao Paulo, São Paulo, SP, Brazil
| | - Mirian Nacagami Sotto
- Department of Dermatology, Medical School, University of Sao Paulo, São Paulo, SP, Brazil
| | - Fernanda Guedes Luiz
- Instituto Pasteur de São Paulo, Laboratório de Diagnóstico da Raiva, São Paulo, SP, Brazil
| | - Walter Belda Júnior
- Department of Dermatology, Medical School, University of Sao Paulo, São Paulo, SP, Brazil
| | - Maria da Gloria Teixeira de Sousa
- Laboratory of Medical Investigation Unit 53, Division of Clinical Dermatology, Medical School, University of Sao Paulo, São Paulo, SP, Brazil
| | - Gil Benard
- Laboratory of Medical Investigation Unit 53, Division of Clinical Dermatology, Medical School, University of Sao Paulo, São Paulo, SP, Brazil
| | - Paulo Ricardo Criado
- Department of Dermatology, Medical School, University of Sao Paulo, São Paulo, SP, Brazil
| |
Collapse
|
23
|
Zhang X, Gao F, Li Q, Dong Z, Sun B, Hou L, Li Z, Liu Z. MSCs with ACE II gene affect apoptosis pathway of acute lung injury induced by bleomycin. Exp Lung Res 2014; 41:32-43. [DOI: 10.3109/01902148.2014.963901] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
24
|
Abstract
PURPOSE OF REVIEW The article provides an overview of efforts to identify and validate biomarkers in acute respiratory distress syndrome (ARDS) and a discussion of the challenges confronting researchers in this area. RECENT FINDINGS Although various putative biomarkers have been investigated in ARDS, the data have been largely disappointing and the 'troponin' of ARDS remains elusive. Establishing a relationship between measurable biological processes and clinical outcomes is vital to advancing clinical trials in ARDS and expanding our arsenal of treatments for this complex syndrome. SUMMARY This article summarizes the current status of ARDS biomarker research and provides a framework for future investigation.
Collapse
|
25
|
Early inflammation mainly affects normally and poorly aerated lung in experimental ventilator-induced lung injury*. Crit Care Med 2014; 42:e279-87. [PMID: 24448197 DOI: 10.1097/ccm.0000000000000161] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVE The common denominator in most forms of ventilator-induced lung injury is an intense inflammatory response mediated by neutrophils. PET with [(18)F]fluoro-2-deoxy-D-glucose can be used to image cellular metabolism, which, during lung inflammatory processes, mainly reflects neutrophil activity, allowing the study of regional lung inflammation in vivo. The aim of this study was to assess the location and magnitude of lung inflammation using PET imaging of [(18)F]fluoro-2-deoxy-D-glucose in a porcine experimental model of early acute respiratory distress syndrome. DESIGN Prospective laboratory investigation. SETTING A university animal research laboratory. SUBJECTS Seven piglets submitted to experimental ventilator-induced lung injury and five healthy controls. INTERVENTIONS Lung injury was induced by lung lavages and 210 minutes of injurious mechanical ventilation using low positive end-expiratory pressure and high inspiratory pressures. All animals were subsequently studied with dynamic PET imaging of [(18)F]fluoro-2-deoxy-D-glucose. CT scans were acquired at end expiration and end inspiration. MEASUREMENTS AND MAIN RESULTS [(18)F]fluoro-2-deoxy-D-glucose uptake rate was computed for the whole lung, four isogravitational regions, and regions grouping voxels with similar density. Global and intermediate gravitational zones [(18)F]fluoro-2-deoxy-D-glucose uptakes were higher in ventilator-induced lung injury piglets compared with controls animals. Uptake of normally and poorly aerated regions was also higher in ventilator-induced lung injury piglets compared with control piglets, whereas regions suffering tidal recruitment or tidal hyperinflation had [(18)F]fluoro-2-deoxy-D-glucose uptakes similar to controls. CONCLUSIONS The present findings suggest that normally and poorly aerated regions--corresponding to intermediate gravitational zones--are the primary targets of the inflammatory process accompanying early experimental ventilator-induced lung injury. This may be attributed to the small volume of the aerated lung, which receives most of ventilation.
Collapse
|
26
|
Shah RJ, Wickersham N, Lederer DJ, Palmer SM, Cantu E, Diamond JM, Kawut SM, Lama VN, Bhorade S, Crespo M, Demissie E, Sonett J, Wille K, Orens J, Weinacker A, Shah P, Arcasoy S, Wilkes DS, Christie JD, Ware LB. Preoperative plasma club (clara) cell secretory protein levels are associated with primary graft dysfunction after lung transplantation. Am J Transplant 2014; 14:446-52. [PMID: 24400993 PMCID: PMC3946770 DOI: 10.1111/ajt.12541] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 09/09/2013] [Accepted: 09/23/2013] [Indexed: 01/25/2023]
Abstract
Inherent recipient factors, including pretransplant diagnosis, obesity and elevated pulmonary pressures, are established primary graft dysfunction (PGD) risks. We evaluated the relationship between preoperative lung injury biomarkers and PGD to gain further mechanistic insight in recipients. We performed a prospective cohort study of recipients in the Lung Transplant Outcomes Group enrolled between 2002 and 2010. Our primary outcome was Grade 3 PGD on Day 2 or 3. We measured preoperative plasma levels of five biomarkers (CC-16, sRAGE, ICAM-1, IL-8 and Protein C) that were previously associated with PGD when measured at the postoperative time point. We used multivariable logistic regression to adjust for potential confounders. Of 714 subjects, 130 (18%) developed PGD. Median CC-16 levels were elevated in subjects with PGD (10.1 vs. 6.0, p<0.001). CC-16 was associated with PGD in nonidiopathic pulmonary fibrosis (non-IPF) subjects (OR for highest quartile of CC-16: 2.87, 95% CI: 1.37, 6.00, p=0.005) but not in subjects with IPF (OR 1.38, 95% CI: 0.43, 4.45, p=0.59). After adjustment, preoperative CC-16 levels remained associated with PGD (OR: 3.03, 95% CI: 1.26, 7.30, p=0.013) in non-IPF subjects. Our study suggests the importance of preexisting airway epithelial injury in PGD. Markers of airway epithelial injury may be helpful in pretransplant risk stratification in specific recipients.
Collapse
Affiliation(s)
- Rupal J. Shah
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Nancy Wickersham
- Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center, Nashville, Tennessee
| | - David J. Lederer
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University College of Physicians and Surgeons, New York, New York
| | - Scott M. Palmer
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University, Raleigh-Durham, North Carolina
| | - Edward Cantu
- Division of Cardiovascular Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Joshua M. Diamond
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Steven M. Kawut
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia,Penn Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Vibha N. Lama
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan
| | - Sangeeta Bhorade
- Division of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, Illinois
| | - Maria Crespo
- Division of Pulmonary, Allergy, and Critical Care, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ejigayehu Demissie
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Joshua Sonett
- Department of Surgery, Columbia University College of Physicians and Surgeons, New York, New York
| | - Keith Wille
- Division of Pulmonary and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jonathan Orens
- Department of Pulmonary and Critical Care, Stanford University, Palo Alto, CA
| | - Ann Weinacker
- Department of Surgery, Johns Hopkins University Hospital, Baltimore, Maryland
| | - Pali Shah
- Department of Pulmonary and Critical Care, Stanford University, Palo Alto, CA
| | - Selim Arcasoy
- Division of Cardiovascular Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - David S. Wilkes
- Division of Pulmonary, Allergy, and Critical Care Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Jason D. Christie
- Pulmonary, Allergy, and Critical Care Division, Perelman School of Medicine at the University of Pennsylvania, Philadelphia,Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia,Penn Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Lorraine B. Ware
- Division of Allergy, Pulmonary, and Critical Care Medicine Vanderbilt University Medical Center, Nashville, Tennessee,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | | |
Collapse
|
27
|
Vincent JL. Dynamics of Regional Lung Inflammation: New Questions and Answers Using PET. ANNUAL UPDATE IN INTENSIVE CARE AND EMERGENCY MEDICINE 2014 2014. [PMCID: PMC7176157 DOI: 10.1007/978-3-319-03746-2_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The meaning of the term ‘inflammation’ has undergone considerable evolution. It was originally defined around the year 25 A.D. by Aulus Cornelius Celsus [1] and described the body’s acute reaction following a traumatic event, such as a microscopic tear of a ligament or muscle. His original wording: “Notae vero inflammationis sunt quatour: rubor et tumor cum calore et dolore” (true signs of inflammation are four: redness and swelling with heat and pain) still holds. Disturbance of function (functio laesa) is the legendary fifth cardinal sign of inflammation and was added by Galen in the second century A.D. [2]. Recent articles [3] highlight the complicated role that inflammation plays in chronic illnesses, including metabolic, cardiovascular and neurodegenerative diseases. In addition to these difficult-to-treat diseases, more research and research tools are needed to illuminate therapeutic strategies in another difficulty-to-treat inflammatory malady, the acute respiratory distress syndrome (ARDS).
Collapse
|
28
|
Derosa S, Borges JB, Segelsjö M, Tannoia A, Pellegrini M, Larsson A, Perchiazzi G, Hedenstierna G. Reabsorption atelectasis in a porcine model of ARDS: regional and temporal effects of airway closure, oxygen, and distending pressure. J Appl Physiol (1985) 2013; 115:1464-73. [DOI: 10.1152/japplphysiol.00763.2013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Little is known about the small airways dysfunction in acute respiratory distress syndrome (ARDS). By computed tomography (CT) imaging in a porcine experimental model of early ARDS, we aimed at studying the location and magnitude of peripheral airway closure and alveolar collapse under high and low distending pressures and high and low inspiratory oxygen fraction (FIO2). Six piglets were mechanically ventilated under anesthesia and muscle relaxation. Four animals underwent saline-washout lung injury, and two served as healthy controls. Beyond the site of assumed airway closure, gas was expected to be trapped in the injured lungs, promoting alveolar collapse. This was tested by ventilation with an FIO2 of 0.25 and 1 in sequence during low and high distending pressures. In the most dependent regions, the gas/tissue ratio of end-expiratory CT, after previous ventilation with FIO2 0.25 low-driving pressure, was significantly higher than after ventilation with FIO2 1; with high-driving pressure, this difference disappeared. Also, significant reduction in poorly aerated tissue and a correlated increase in nonaerated tissue in end-expiratory CT with FIO2 1 low-driving pressure were seen. When high-driving pressure was applied or after previous ventilation with FIO2 0.25 and low-driving pressure, this pattern disappeared. The findings suggest that low distending pressures produce widespread dependent airway closure and with high FIO2, subsequent absorption atelectasis. Low FIO2 prevented alveolar collapse during the study period because of slow absorption of gas behind closed airways.
Collapse
Affiliation(s)
- Savino Derosa
- Department of Emergency and Organ Transplant, Bari University, Bari, Italy
- Hedenstierna Laboratory, Department of Surgical Sciences, Section of Anaesthesiology and Critical Care, Uppsala University, Uppsala, Sweden
| | - João Batista Borges
- Hedenstierna Laboratory, Department of Surgical Sciences, Section of Anaesthesiology and Critical Care, Uppsala University, Uppsala, Sweden
- Pulmonary Divison, Heart Institute (Incor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Monica Segelsjö
- Department of Radiology, Oncology and Radiation Science, Section of Radiology, Uppsala University, Uppsala, Sweden; and
| | - Angela Tannoia
- Department of Emergency and Organ Transplant, Bari University, Bari, Italy
| | | | - Anders Larsson
- Hedenstierna Laboratory, Department of Surgical Sciences, Section of Anaesthesiology and Critical Care, Uppsala University, Uppsala, Sweden
| | - Gaetano Perchiazzi
- Department of Emergency and Organ Transplant, Bari University, Bari, Italy
| | - Göran Hedenstierna
- Hedenstierna Laboratory, Department of Medical Sciences, Clinical Physiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
29
|
Chiaretti A, Pulitanò S, Conti G, Barone G, Buonsenso D, Manni L, Capozzi D, Ria F, Riccardi R. Interleukin and neurotrophin up-regulation correlates with severity of H1N1 infection in children: a case-control study. Int J Infect Dis 2013; 17:e1186-93. [PMID: 24094526 DOI: 10.1016/j.ijid.2013.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 07/08/2013] [Accepted: 07/11/2013] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE To evaluate the correlation between cytokine and neurotrophin expression and clinical findings, disease severity, and outcome of children with H1N1 influenza infection. METHODS A prospective observational clinical study was performed on 15 children with H1N1 infection, 15 controls with lower respiratory tract infections (LRTI), and 15 non-infected children. Plasma levels of interleukin (IL)-1β, IL-6, and neurotrophic factor (nerve growth factor (NGF), brain derived neurotrophic factor (BDNF), and glial derived neurotrophic factor (GDNF)) were measured using immunoenzymatic assays. RESULTS Significantly higher levels of IL-1β, IL-6, BDNF, and NGF were detected in patients with H1N1 infection compared to LRTI controls, while there was no significant variation in GDNF in the two groups. IL-1β, IL-6, BDNF, and NGF levels were significantly higher in H1N1 patients with more severe clinical manifestations compared to H1N1 patients with mild clinical manifestations. Of note, IL-6 was significantly correlated with the severity of respiratory compromise and fever, while NGF up-regulation was associated with the duration of cough. No correlation was found between interleukin and neurotrophic factor expression and outcome. CONCLUSIONS H1N1 infection induces an early and significant IL-1β, IL-6, BDNF, and NGF up-regulation. The over-expression of these molecular markers is likely to play a neuroimmunomodulatory role in H1N1 infection and may contribute to airway inflammation and bronchial hyper-reactivity in infected children.
Collapse
Affiliation(s)
- Antonio Chiaretti
- Department of Pediatrics, Catholic University of the Sacred Heart, A. Gemelli Hospital, Largo Gemelli, 1, 00168, Rome, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|