1
|
Bentley JK, Kreger JE, Breckenridge HA, Singh S, Lei J, Li Y, Baker SC, Lumeng CN, Hershenson MB. Developing a mouse model of human coronavirus NL63 infection: comparison with rhinovirus-A1B and effects of prior rhinovirus infection. Am J Physiol Lung Cell Mol Physiol 2024; 327:L557-L573. [PMID: 39189801 DOI: 10.1152/ajplung.00149.2023] [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: 05/09/2023] [Revised: 08/02/2024] [Accepted: 08/04/2024] [Indexed: 08/28/2024] Open
Abstract
Human coronavirus (HCoV)-NL63 causes respiratory tract infections in humans and uses angiotensin-converting enzyme 2 (ACE2) as a receptor. We sought to establish a mouse model of HCoV-NL63 and determine whether prior rhinovirus (RV)-A1B infection affected HCoV-NL63 replication. HCoV-NL63 was propagated in LLC-MK2 cells expressing human ACE2. RV-A1B was grown in HeLa-H1 cells. C57BL6/J or transgenic mice expressing human ACE2 were infected intranasally with sham LLC-MK2 cell supernatant or 1 × 105 tissue culture infectious dose (TCID50) units HCoV-NL63. Wild-type mice were infected with 1 × 106 plaque-forming units (PFU) RV-A1B. Lungs were assessed for vRNA, bronchoalveolar lavage (BAL) cells, histology, HCoV-NL63 nonstructural protein 3 (nsp3), and host gene expression by next-generation sequencing and qPCR. To evaluate sequential infections, mice were infected with RV-A1B followed by HCoV-NL63 infection 4 days later. We report that hACE2 mice infected with HCoV-NL63 showed evidence of replicative infection with increased levels of vRNA, BAL neutrophils and lymphocytes, peribronchial and perivascular infiltrates, and expression of nsp3. Viral replication peaked 3 days after infection and inflammation persisted 6 days after infection. HCoV-NL63-infected hACE2 mice showed increased mRNA expression of IFNs, IFN-stimulated proteins, and proinflammatory cytokines. Infection with RV-A1B 4 days before HCoV-NL63 significantly decreased both HCoV-NL63 vRNA levels and airway inflammation. Mice infected with RV-A1B prior to HCoV-NL63 showed increased expression of antiviral proteins compared with sham-treated mice. In conclusion, we established a mouse model of HCoV-NL63 replicative infection characterized by relatively persistent viral replication and inflammation. Prior infection with RV-A1B reduced HCoV-NL63 replication and airway inflammation, indicative of viral interference.NEW & NOTEWORTHY We describe a mouse model of human coronavirus (HCoV) infection. Infection of transgenic mice expressing human angiotensin-converting enzyme 2 (ACE2) with HCoV-NL63 produced a replicative infection with peribronchial inflammation and nonstructural protein 3 expression. Mice infected with RV-A1B 4 days before HCoV-NL63 showed decreased HCoV-NL63 replication and airway inflammation and increased expression of antiviral proteins compared with sham-treated mice. This research may shed light on human coronavirus infections, viral interference, and viral-induced asthma exacerbations.
Collapse
Affiliation(s)
- J Kelley Bentley
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Jordan E Kreger
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Haley A Breckenridge
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Shilpi Singh
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Jing Lei
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Yiran Li
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Susan C Baker
- Department of Microbiology and Immunology, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois, United States
| | - Carey N Lumeng
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, United States
- Department Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Marc B Hershenson
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, United States
- Department Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| |
Collapse
|
2
|
Urbančič J, Košak Soklič T, Demšar Luzar A, Hočevar Boltežar I, Korošec P, Rijavec M. Transcriptomic Differentiation of Phenotypes in Chronic Rhinosinusitis and Its Implications for Understanding the Underlying Mechanisms. Int J Mol Sci 2023; 24:ijms24065541. [PMID: 36982612 PMCID: PMC10051401 DOI: 10.3390/ijms24065541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/04/2023] [Accepted: 03/12/2023] [Indexed: 03/16/2023] Open
Abstract
Chronic rhinosinusitis (CRS) is a multifaceted disease with variable clinical courses and outcomes. We aimed to determine CRS-associated nasal-tissue transcriptome in clinically well-characterized and phenotyped individuals, to gain a novel insight into the biological pathways of the disease. RNA-sequencing of tissue samples of patients with CRS with polyps (CRSwNP), without polyps (CRSsNP), and controls were performed. Characterization of differently expressed genes (DEGs) and functional and pathway analysis was undertaken. We identified 782 common CRS-associated nasal-tissue DEGs, while 375 and 328 DEGs were CRSwNP- and CRSsNP-specific, respectively. Common key DEGs were found to be involved in dendritic cell maturation, the neuroinflammation pathway, and the inhibition of the matrix metalloproteinases. Distinct CRSwNP-specific DEGs were involved in NF-kβ canonical pathways, Toll-like receptor signaling, HIF1α regulation, and the Th2 pathway. CRSsNP involved the NFAT pathway and changes in the calcium pathway. Our findings offer new insights into the common and distinct molecular mechanisms underlying CRSwNP and CRSsNP, providing further understanding of the complex pathophysiology of the CRS, with future research directions for novel treatment strategies.
Collapse
Affiliation(s)
- Jure Urbančič
- Department of Otorhinolaryngology and Cervicofacial Surgery, University Medical Centre Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
- Correspondence:
| | - Tanja Košak Soklič
- Department of Otorhinolaryngology and Cervicofacial Surgery, University Medical Centre Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - Ajda Demšar Luzar
- Laboratory for Clinical Immunology and Molecular Genetics, University Clinic of Respiratory and Allergic Diseases Golnik, Golnik 36, SI-4204 Golnik, Slovenia
| | - Irena Hočevar Boltežar
- Department of Otorhinolaryngology and Cervicofacial Surgery, University Medical Centre Ljubljana, Zaloska 2, SI-1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, SI-1000 Ljubljana, Slovenia
| | - Peter Korošec
- Laboratory for Clinical Immunology and Molecular Genetics, University Clinic of Respiratory and Allergic Diseases Golnik, Golnik 36, SI-4204 Golnik, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, SI-1000 Ljubljana, Slovenia
| | - Matija Rijavec
- Laboratory for Clinical Immunology and Molecular Genetics, University Clinic of Respiratory and Allergic Diseases Golnik, Golnik 36, SI-4204 Golnik, Slovenia
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
3
|
Chen R, Piao LZ, Liu L, Zhang XF. DNA methylation and gene expression profiles to identify childhood atopic asthma associated genes. BMC Pulm Med 2021; 21:292. [PMID: 34525985 PMCID: PMC8444351 DOI: 10.1186/s12890-021-01655-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 09/01/2021] [Indexed: 11/10/2022] Open
Abstract
Background Asthma is a chronic inflammatory disorder of the airways involving many different factors. This study aimed to screen for the critical genes using DNA methylation/CpGs and miRNAs involved in childhood atopic asthma. Methods DNA methylation and gene expression data (Access Numbers GSE40732 and GSE40576) were downloaded from the Gene Expression Omnibus database. Each set contains 194 peripheral blood mononuclear cell (PBMC) samples of 97 children with atopic asthma and 97 control children. Differentially expressed genes (DEGs) with DNA methylation changes were identified. Pearson correlation analysis was used to select genes with an opposite direction of expression and differences in methylation levels, and then Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed. Protein–protein interaction network and miRNA–target gene regulatory networks were then constructed. Finally, important genes related to asthma were screened. Results A total of 130 critical DEGs with DNA methylation changes were screened from children with atopic asthma and compared with control samples from healthy children. GO and KEGG pathway enrichment analysis found that critical genes were primarily related to 24 GO terms and 10 KEGG pathways. In the miRNA–target gene regulatory networks, 9 KEGG pathways were identified. Analysis of the miRNA–target gene network noted an overlapping KEGG signaling pathway, hsa04060: cytokine-cytokine receptor interaction, in which the gene CCL2, directly related to asthma, was involved. This gene is targeted by eight asthma related miRNAs (hsa-miR-206, hsa-miR-19a, hsa-miR-9,hsa-miR-22, hsa-miR-33b, hsa-miR-122, hsa-miR-1, and hsa-miR-23b). The genes IL2RG and CCl4 were also involved in this pathway. Conclusions The present study provides a novel insight into the underlying molecular mechanism of childhood atopic asthma.
Collapse
Affiliation(s)
- Rui Chen
- Department of Pediatrics, The Third Hospital of Jilin University, Changchun, 130033, Jilin, China
| | - Li-Zhen Piao
- Department of Pediatrics, The Third Hospital of Jilin University, Changchun, 130033, Jilin, China
| | - Ling Liu
- Department of Pediatrics, The Third Hospital of Jilin University, Changchun, 130033, Jilin, China.
| | - Xiao-Fei Zhang
- Department of Pediatrics, The Third Hospital of Jilin University, Changchun, 130033, Jilin, China.
| |
Collapse
|
4
|
Jazaeri S, Goldsmith AM, Jarman CR, Lee J, Hershenson MB, Lewis TC. Nasal interferon responses to community rhinovirus infections are similar in controls and children with asthma. Ann Allergy Asthma Immunol 2021; 126:690-695.e1. [PMID: 33515711 DOI: 10.1016/j.anai.2021.01.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/12/2020] [Accepted: 01/19/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Rhinovirus (RV) is the main cause of asthma exacerbations in children. Some studies reported that persons with asthma have attenuated interferon (IFN) responses to experimental RV infection compared with healthy individuals. However, responses to community-acquired RV infections in controls and children with asthma have not been compared. OBJECTIVE To evaluate nasal cytokine responses after natural RV infections in people with asthma and healthy children. METHODS We compared nasal cytokine expression among controls and children with asthma during healthy, virus-negative surveillance weeks and self-reported RV-positive sick weeks. A total of 14 controls and 21 patients with asthma were studied. Asthma disease severity was based on symptoms and medication use. Viral genome was detected by multiplex polymerase chain reaction. Nasal cytokine protein levels were determined by multiplex assays. RESULTS Two out of 47 surveillance weeks tested positive for RV, illustrating an asymptomatic infection rate of 5%. A total of 38 of 47 sick weeks (81%) tested positive for the respiratory virus. Of these, 33 (87%) were positive for RV. During well weeks, nasal interleukin 8 (IL-8), IL-12, and IL-1β levels were higher in children with asthma than controls. Compared with healthy virus-negative surveillance weeks, IL-8, IL-13, and interferon beta increased during colds only in patients with asthma. In both controls and children with asthma, the nasal levels of interferon gamma, interferon lambda-1, IL-1β, IL-8, and IL-10 increased during RV-positive sick weeks. During RV infection, IL-8, IL-1β, and tumor necrosis factor-α levels were strongly correlated. CONCLUSION In both controls and patients with asthma, natural RV infection results in robust type II and III IFN responses.
Collapse
Affiliation(s)
| | - Adam M Goldsmith
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan
| | - Caitlin R Jarman
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan
| | - Julie Lee
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan
| | - Marc B Hershenson
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan
| | - Toby C Lewis
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan.
| |
Collapse
|
5
|
Skovbjerg S, Roos K, Andersson M, Rabe H, Nilsson S, Lindh M, Wold AE. Inflammatory Mediator Profiles in Secretory Otitis Media in Relationship to Viable Bacterial Pathogens and Bacterial and Viral Nucleic Acids. J Interferon Cytokine Res 2020; 40:555-569. [PMID: 33337936 DOI: 10.1089/jir.2020.0075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Secretory otitis media (SOM) is characterized by persistence of fluid in the middle ear, often following an episode of acute otitis media. Our hypothesis is that failure to eliminate bacterial or viral pathogens may result in persistent low-grade inflammation. In this study, we analyzed inflammatory mediators in middle ear fluids from 67 children with SOM. This was combined with determinations of viable bacteria by culture along with detection of bacterial and viral genetic material by real-time polymerase chain reaction (PCR). The inflammatory mediators found at the highest concentrations (>30 ng/mL) were stem cell growth factor-β (median 110 ng/mL), CXCL1, IL-16, IL-8, migration inhibitory factor, CXCL10, and CXCL9. Among bacterial pathogens, Moraxella catarrhalis and Haemophilus influenzae dominated, regardless of detection methods, while rhinovirus dominated among viral pathogens. Middle ear fluid levels of interleukin (IL)-1α, IL-17, IL-1β, fibroblast growth factor basic, and tumor necrosis factor correlated strongly with presence of bacteria detected either by culture or PCR, while IL-1RA, IL-3, IL-6, IL-8, CCL3, CCL4, and granulocyte-colony stimulating factor correlated significantly with real-time PCR values. CXCL10, CXCL9, CCL2, and TRAIL correlated significantly with viral nucleic acid levels. To conclude, persistence of viral and bacterial pathogens may fuel persistent inflammation in SOM. Bacteria caused a broad inflammatory response, while viruses chiefly elicited the interferon-induced chemokines CXCL9 and CXCL10.
Collapse
Affiliation(s)
- Susann Skovbjerg
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kristian Roos
- ENT Department, Capio Lundby Hospital, Gothenburg, Sweden
| | - Maria Andersson
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Hardis Rabe
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Staffan Nilsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Mathematical Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Magnus Lindh
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Agnes E Wold
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Clinical Microbiology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
6
|
He J, Yang Q, Xiao Q, Lei A, Li X, Zhou P, Liu T, Zhang L, Shi K, Yang Q, Dong J, Zhou J. IRF-7 Is a Critical Regulator of Type 2 Innate Lymphoid Cells in Allergic Airway Inflammation. Cell Rep 2020; 29:2718-2730.e6. [PMID: 31775040 DOI: 10.1016/j.celrep.2019.10.077] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 08/16/2019] [Accepted: 10/18/2019] [Indexed: 12/12/2022] Open
Abstract
Allergic asthma is a highly prevalent airway disease triggered by hyperresponsiveness to inhaled allergens. Interferon regulatory factor 7 (IRF7) has been shown to be highly expressed in nasal aspirates from children with asthma. Type 2 innate lymphoid cells (ILC2s) represent the major player in allergic airway inflammation. The role of IRF7 in ILC2-driven asthma remains to be explored. Here, we report that IRF7 expression in murine lung ILC2s is dramatically induced upon papain or interleukin-33 (IL-33) stimulation. ILC2s from asthma patients display a much higher level of IRF7 than those from healthy donors. Deficiency of IRF7 in mice significantly impairs the expansion and function of lung ILC2s in multiple models of allergic asthma. Furthermore, the regulation of ILC2s by IRF7 is cell intrinsic and mediated by the transcription factor Bcl11b. These observations identify IRF7 as a regulator of lung ILC2s, which may have immunotherapeutic value in allergic asthma.
Collapse
Affiliation(s)
- Juan He
- Joint Program in Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China; Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qiong Yang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qiang Xiao
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Aihua Lei
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xing Li
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Pan Zhou
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Ting Liu
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Lijuan Zhang
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Kun Shi
- Department of Gynaecology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Quan Yang
- Key Laboratory of Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Junchao Dong
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jie Zhou
- Joint Program in Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
7
|
Sinha A, Lutter R, Dekker T, Dierdorp B, J. Sterk P, Frey U, Delgado-Eckert E. Can Measurements of Inflammatory Biomarkers be Used to Spot Respiratory Viral Infections? Viruses 2020; 12:v12101175. [PMID: 33080844 PMCID: PMC7594027 DOI: 10.3390/v12101175] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 11/16/2022] Open
Abstract
Accurate detection of human respiratory viral infections is highly topical. We investigated how strongly inflammatory biomarkers (FeNO, eosinophils, neutrophils, and cytokines in nasal lavage fluid) and lung function parameters change upon rhinovirus 16 infection, in order to explore their potential use for infection detection. To this end, within a longitudinal cohort study, healthy and mildly asthmatic volunteers were experimentally inoculated with rhinovirus 16, and time series of these parameters/biomarkers were systematically recorded and compared between the pre- and post-infection phases of the study, which lasted two months and one month, respectively. We found that the parameters’/biomarkers’ ability to discriminate between the infected and the uninfected state varied over the observation time period. Consistently over time, the concentration of cytokines, in nasal lavage fluid, showed moderate to very good discrimination performance, thereby qualifying for disease progression monitoring, whereas lung function and FeNO, while quickly and non-invasively measurable using cheap portable devices (e.g., at airports), performed poorly.
Collapse
Affiliation(s)
- Anirban Sinha
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (R.L.); (P.J.S.)
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (T.D.); (B.D.)
- Correspondence: ; Tel.: +31-20-566-4356
| | - René Lutter
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (R.L.); (P.J.S.)
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (T.D.); (B.D.)
| | - Tamara Dekker
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (T.D.); (B.D.)
| | - Barbara Dierdorp
- Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (T.D.); (B.D.)
| | - Peter J. Sterk
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (R.L.); (P.J.S.)
| | - Urs Frey
- Computational Physiology and Biostatistics, Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, 4123 Allschwil, Switzerland
| | - Edgar Delgado-Eckert
- University Children’s Hospital (UKBB), University of Basel, Spitalstrasse 33, Postfach, 4031 Basel, Switzerland; (U.F.); (E.D.-E.)
- Computational Physiology and Biostatistics, Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, 4123 Allschwil, Switzerland
| |
Collapse
|
8
|
Ray PR, Wangzhou A, Ghneim N, Yousuf MS, Paige C, Tavares-Ferreira D, Mwirigi JM, Shiers S, Sankaranarayanan I, McFarland AJ, Neerukonda SV, Davidson S, Dussor G, Burton MD, Price TJ. A pharmacological interactome between COVID-19 patient samples and human sensory neurons reveals potential drivers of neurogenic pulmonary dysfunction. Brain Behav Immun 2020; 89:559-568. [PMID: 32497778 PMCID: PMC7263237 DOI: 10.1016/j.bbi.2020.05.078] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 12/21/2022] Open
Abstract
The SARS-CoV-2 virus infects cells of the airway and lungs in humans causing the disease COVID-19. This disease is characterized by cough, shortness of breath, and in severe cases causes pneumonia and acute respiratory distress syndrome (ARDS) which can be fatal. Bronchial alveolar lavage fluid (BALF) and plasma from mild and severe cases of COVID-19 have been profiled using protein measurements and bulk and single cell RNA sequencing. Onset of pneumonia and ARDS can be rapid in COVID-19, suggesting a potential neuronal involvement in pathology and mortality. We hypothesized that SARS-CoV-2 infection drives changes in immune cell-derived factors that then interact with receptors expressed by the sensory neuronal innervation of the lung to further promote important aspects of disease severity, including ARDS. We sought to quantify how immune cells might interact with sensory innervation of the lung in COVID-19 using published data from patients, existing RNA sequencing datasets from human dorsal root ganglion neurons and other sources, and a genome-wide ligand-receptor pair database curated for pharmacological interactions relevant for neuro-immune interactions. Our findings reveal a landscape of ligand-receptor interactions in the lung caused by SARS-CoV-2 viral infection and point to potential interventions to reduce the burden of neurogenic inflammation in COVID-19 pulmonary disease. In particular, our work highlights opportunities for clinical trials with existing or under development rheumatoid arthritis and other (e.g. CCL2, CCR5 or EGFR inhibitors) drugs to treat high risk or severe COVID-19 cases.
Collapse
Affiliation(s)
- Pradipta R Ray
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA.
| | - Andi Wangzhou
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Nizar Ghneim
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Muhammad S Yousuf
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Candler Paige
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Diana Tavares-Ferreira
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Juliet M Mwirigi
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Stephanie Shiers
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Ishwarya Sankaranarayanan
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Amelia J McFarland
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Sanjay V Neerukonda
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Steve Davidson
- University of Cincinnati, College of Medicine, Department of Anesthesiology, USA
| | - Gregory Dussor
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA
| | - Michael D Burton
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Neuroimmunology and Behavior Research Group, USA
| | - Theodore J Price
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies, Pain Neurobiology Research Group, USA.
| |
Collapse
|
9
|
Ray PR, Wangzhou A, Ghneim N, Yousuf MS, Paige C, Tavares-Ferreira D, Mwirigi JM, Shiers S, Sankaranarayanan I, McFarland AJ, Neerukonda SV, Davidson S, Dussor G, Burton MD, Price TJ. A pharmacological interactome between COVID-19 patient samples and human sensory neurons reveals potential drivers of neurogenic pulmonary dysfunction. SSRN 2020:3581446. [PMID: 32714114 PMCID: PMC7366818 DOI: 10.2139/ssrn.3581446] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/04/2020] [Indexed: 11/15/2022]
Abstract
The SARS-CoV-2 virus infects cells of the airway and lungs in humans causing the disease COVID-19. This disease is characterized by cough, shortness of breath, and in severe cases causes pneumonia and acute respiratory distress syndrome (ARDS) which can be fatal. Bronchial alveolar lavage fluid (BALF) and plasma from mild and severe cases of COVID-19 have been profiled using protein measurements and bulk and single cell RNA sequencing. Onset of pneumonia and ARDS can be rapid in COVID-19, suggesting a potential neuronal involvement in pathology and mortality. We sought to quantify how immune cells might interact with sensory innervation of the lung in COVID-19 using published data from patients, existing RNA sequencing datasets from human dorsal root ganglion neurons and other sources, and a genome-wide ligand-receptor pair database curated for pharmacological interactions relevant for neuro-immune interactions. Our findings reveal a landscape of ligand-receptor interactions in the lung caused by SARS-CoV-2 viral infection and point to potential interventions to reduce the burden of neurogenic inflammation in COVID-19 disease. In particular, our work highlights opportunities for clinical trials with existing or under development rheumatoid arthritis and other (e.g. CCL2, CCR5 or EGFR inhibitors) drugs to treat high risk or severe COVID-19 cases.
Collapse
Affiliation(s)
- Pradipta R. Ray
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Andi Wangzhou
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Nizar Ghneim
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Muhammad S. Yousuf
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Candler Paige
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Diana Tavares-Ferreira
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Juliet M. Mwirigi
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Stephanie Shiers
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Ishwarya Sankaranarayanan
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Amelia J. McFarland
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Sanjay V. Neerukonda
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Steve Davidson
- University of Cincinnati, College of Medicine, Department of Anesthesiology
| | - Gregory Dussor
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Michael D. Burton
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| | - Theodore J. Price
- University of Texas at Dallas, School of Behavioral and Brain Sciences and Center for Advanced Pain Studies
| |
Collapse
|
10
|
Halpin DMG, Singh D, Hadfield RM. Inhaled corticosteroids and COVID-19: a systematic review and clinical perspective. Eur Respir J 2020; 55:2001009. [PMID: 32341100 PMCID: PMC7236828 DOI: 10.1183/13993003.01009-2020] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 04/16/2020] [Indexed: 11/05/2022]
Abstract
The current coronavirus 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) infection, raises important questions as to whether pre-morbid use or continued administration of inhaled corticosteroids (ICS) affects the outcomes of acute respiratory infections due to coronavirus. Many physicians are concerned about whether individuals positive for SARS-CoV-2 and taking ICS should continue them or stop them, given that ICS are often regarded as immunosuppressive. A number of key questions arise. Are people with asthma or COPD at increased risk of developing COVID-19? Do ICS modify this risk, either increasing or decreasing it? Do ICS influence the clinical course of COVID-19? (figure 1). Whether ICS modify the risk of developing COVID-19 or the clinical course of COVID-19 in people who do not have lung disease should also be considered (figure 1). There is no evidence on benefits or harms of inhaled steroids in COVID-19. It is essential that epidemiological studies of COVID-19 include detailed information on comorbidities and prior medication to help answer this question. https://bit.ly/2XVwIsa
Collapse
Affiliation(s)
- David M G Halpin
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter, UK
- Global Initiative for Chronic Obstructive Lung Disease (GOLD)
| | - Dave Singh
- Global Initiative for Chronic Obstructive Lung Disease (GOLD)
- University of Manchester, Manchester University NHS Foundation Trust, Manchester, UK
| | - Ruth M Hadfield
- Global Initiative for Chronic Obstructive Lung Disease (GOLD)
- Macquarie University, Sydney, Australia
| |
Collapse
|
11
|
|
12
|
Jha A, Dunning J, Tunstall T, Thwaites RS, Hoang LT, Kon OM, Zambon MC, Hansel TT, Openshaw PJ. Patterns of systemic and local inflammation in patients with asthma hospitalised with influenza. Eur Respir J 2019; 54:13993003.00949-2019. [PMID: 31391224 DOI: 10.1183/13993003.00949-2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/25/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Patients with asthma are at risk of hospitalisation with influenza, but the reasons for this predisposition are unknown. STUDY SETTING A prospective observational study of adults with PCR-confirmed influenza in 11 UK hospitals, measuring nasal, nasopharyngeal and systemic immune mediators and whole-blood gene expression. RESULTS Of 133 admissions, 40 (30%) had previous asthma; these were more often female (70% versus 38.7%, OR 3.69, 95% CI 1.67-8.18; p=0.0012), required less mechanical ventilation (15% versus 37.6%, Chi-squared 6.78; p=0.0338) and had shorter hospital stays (mean 8.3 versus 15.3 days, p=0.0333) than those without. In patients without asthma, severe outcomes were more frequent in those given corticosteroids (OR 2.63, 95% CI 1.02-6.96; p=0.0466) or presenting >4 days after disease onset (OR 5.49, 95% CI 2.28-14.03; p=0.0002). Influenza vaccination in at-risk groups (including asthma) were lower than intended by national policy and the early use of antiviral medications were less than optimal. Mucosal immune responses were equivalent between groups. Those with asthma had higher serum interferon (IFN)-α, but lower serum tumour necrosis factor, interleukin (IL)-5, IL-6, CXCL8, CXCL9, IL-10, IL-17 and CCL2 levels (all p<0.05); both groups had similar serum IL-13, total IgE, periostin and blood eosinophil gene expression levels. Asthma diagnosis was unrelated to viral load, IFN-α, IFN-γ, IL-5 or IL-13 levels. CONCLUSIONS Asthma is common in those hospitalised with influenza, but may not represent classical type 2-driven disease. Those admitted with influenza tend to be female with mild serum inflammatory responses, increased serum IFN-α levels and good clinical outcomes.
Collapse
Affiliation(s)
- Akhilesh Jha
- National Heart and Lung Institute, Imperial College London, St Mary's Campus, London, UK.,Dept of Medicine, University of Cambridge, Cambridge, UK
| | - Jake Dunning
- National Heart and Lung Institute, Imperial College London, St Mary's Campus, London, UK.,Public Health England (formerly Health Protection Agency), London, UK
| | - Tanushree Tunstall
- National Heart and Lung Institute, Imperial College London, St Mary's Campus, London, UK
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, St Mary's Campus, London, UK
| | - Long T Hoang
- National Heart and Lung Institute, Imperial College London, St Mary's Campus, London, UK
| | | | - Onn Min Kon
- National Heart and Lung Institute, Imperial College London, St Mary's Campus, London, UK
| | - Maria C Zambon
- Public Health England (formerly Health Protection Agency), London, UK
| | - Trevor T Hansel
- National Heart and Lung Institute, Imperial College London, St Mary's Campus, London, UK
| | - Peter J Openshaw
- National Heart and Lung Institute, Imperial College London, St Mary's Campus, London, UK
| |
Collapse
|
13
|
Southworth T, Pattwell C, Khan N, Mowbray SF, Strieter RM, Erpenbeck VJ, Singh D. Increased type 2 inflammation post rhinovirus infection in patients with moderate asthma. Cytokine 2019; 125:154857. [PMID: 31557636 DOI: 10.1016/j.cyto.2019.154857] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 01/15/2023]
Abstract
Rhinovirus (RV) infections are a major cause of exacerbations in patients with asthma. Experimental RV challenges can provide insight into the pathophysiology of viral exacerbations. Previous reports, investigating mild or moderate asthma patients, have shown an upregulation in type 2 inflammation post RV infection, however, studies specifically involving asthma patients taking inhaled corticosteroids have concentrated on symptoms and lung function, rather than the inflammatory response. Eleven moderate asthma patients were inoculated with RV. Cold symptoms and asthma control were assessed at baseline and post infection. Nasal epithelial lining fluid and bronchial alveolar lavage (BAL) fluid were collected at baseline and 4 days post infection for assessment of inflammatory proteins. Patients suffered increased cold symptoms and decreased asthma control within 7 days of infection. Antiviral mechanisms were induced following inoculation, with increases in interferon -α, β, γ and λ, as well as CXCL10 and CXCL11. Type 2 inflammatory cytokines were also significantly elevated post RV infection in both nasal and bronchial samples. In BAL, epithelial derived IL-25 and IL-33 levels strongly correlated with Th2 cytokines, IL-4, IL-5 and IL-13. We show how experimental rhinovirus challenge regulates lung and nasal biomarkers in asthma patients taking inhaled corticosteroids. These biomarkers could be used to evaluate the effects of novel drugs for asthma.
Collapse
Affiliation(s)
- Thomas Southworth
- University of Manchester & Medicines Evaluation Unit, Manchester, UK.
| | | | | | - Sarah F Mowbray
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | | | | | - Dave Singh
- University of Manchester & Medicines Evaluation Unit, Manchester, UK
| |
Collapse
|
14
|
Carvajal JJ, Avellaneda AM, Salazar-Ardiles C, Maya JE, Kalergis AM, Lay MK. Host Components Contributing to Respiratory Syncytial Virus Pathogenesis. Front Immunol 2019; 10:2152. [PMID: 31572372 PMCID: PMC6753334 DOI: 10.3389/fimmu.2019.02152] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 08/27/2019] [Indexed: 12/22/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the most prevalent viral etiological agent of acute respiratory tract infection. Although RSV affects people of all ages, the disease is more severe in infants and causes significant morbidity and hospitalization in young children and in the elderly. Host factors, including an immature immune system in infants, low lymphocyte levels in patients under 5 years old, and low levels of RSV-specific neutralizing antibodies in the blood of adults over 65 years of age, can explain the high susceptibility to RSV infection in these populations. Other host factors that correlate with severe RSV disease include high concentrations of proinflammatory cytokines such as interleukins (IL)-6, IL-8, tumor necrosis factor (TNF)-α, and thymic stromal lymphopoitein (TSLP), which are produced in the respiratory tract of RSV-infected individuals, accompanied by a strong neutrophil response. In addition, data from studies of RSV infections in humans and in animal models revealed that this virus suppresses adaptive immune responses that could eliminate it from the respiratory tract. Here, we examine host factors that contribute to RSV pathogenesis based on an exhaustive review of in vitro infection in humans and in animal models to provide insights into the design of vaccines and therapeutic tools that could prevent diseases caused by RSV.
Collapse
Affiliation(s)
- Jonatan J. Carvajal
- Departamento de Biotecnología, Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
| | - Andrea M. Avellaneda
- Departamento de Biotecnología, Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
| | - Camila Salazar-Ardiles
- Departamento de Biotecnología, Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
| | - Jorge E. Maya
- Departamento de Biotecnología, Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Margarita K. Lay
- Departamento de Biotecnología, Facultad de Ciencias del Mar y Recursos Biológicos, Universidad de Antofagasta, Antofagasta, Chile
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad de Chile, Santiago, Chile
| |
Collapse
|
15
|
Han M, Rajput C, Hershenson MB. Rhinovirus Attributes that Contribute to Asthma Development. Immunol Allergy Clin North Am 2019; 39:345-359. [PMID: 31284925 PMCID: PMC6624084 DOI: 10.1016/j.iac.2019.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Early-life wheezing-associated infections with human rhinovirus (HRV) are strongly associated with the inception of asthma. The immune system of immature mice and humans is skewed toward a type 2 cytokine response. Thus, HRV-infected 6-day-old mice but not adult mice develop augmented type 2 cytokine expression, eosinophilic inflammation, mucous metaplasia, and airway hyperresponsiveness. This asthma phenotype depends on interleukin (IL)-13-producing type 2 innate lymphoid cells, the expansion of which in turn depends on release of the innate cytokines IL-25, IL-33, and thymic stromal lymphopoietin from the airway epithelium. In humans, certain genetic variants may predispose to HRV-induced childhood asthma.
Collapse
Affiliation(s)
- Mingyuan Han
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Medical Sciences Research Building II, 1150 West Medical Center Drive, Ann Arbor, MI, USA
| | - Charu Rajput
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Medical Sciences Research Building II, 1150 West Medical Center Drive, Ann Arbor, MI, USA
| | - Marc B Hershenson
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Medical Sciences Research Building II, 1150 West Medical Center Drive, Ann Arbor, MI, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Medical Sciences Research Building II, 1150 West Medical Center Drive, Ann Arbor, MI, USA.
| |
Collapse
|
16
|
Lewis TC, Metitiri EE, Mentz GB, Ren X, Carpenter AR, Goldsmith AM, Wicklund KE, Eder BN, Comstock AT, Ricci JM, Brennan SR, Washington GL, Owens KB, Mukherjee B, Robins TG, Batterman SA, Hershenson MB. Influence of viral infection on the relationships between airway cytokines and lung function in asthmatic children. Respir Res 2018; 19:228. [PMID: 30463560 PMCID: PMC6249926 DOI: 10.1186/s12931-018-0922-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 10/24/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Few longitudinal studies examine inflammation and lung function in asthma. We sought to determine the cytokines that reduce airflow, and the influence of respiratory viral infections on these relationships. METHODS Children underwent home collections of nasal lavage during scheduled surveillance periods and self-reported respiratory illnesses. We studied 53 children for one year, analyzing 392 surveillance samples and 203 samples from 85 respiratory illnesses. Generalized estimated equations were used to evaluate associations between nasal lavage biomarkers (7 mRNAs, 10 proteins), lung function and viral infection. RESULTS As anticipated, viral infection was associated with increased cytokines and reduced FVC and FEV1. However, we found frequent and strong interactions between biomarkers and virus on lung function. For example, in the absence of viral infection, CXCL10 mRNA, MDA5 mRNA, CXCL10, IL-4, IL-13, CCL4, CCL5, CCL20 and CCL24 were negatively associated with FVC. In contrast, during infection, the opposite relationship was frequently found, with IL-4, IL-13, CCL5, CCL20 and CCL24 levels associated with less severe reductions in both FVC and FEV1. CONCLUSIONS In asthmatic children, airflow obstruction is driven by specific pro-inflammatory cytokines. In the absence of viral infection, higher cytokine levels are associated with decreasing lung function. However, with infection, there is a reversal in this relationship, with cytokine abundance associated with reduced lung function decline. While nasal samples may not reflect lower airway responses, these data suggest that some aspects of the inflammatory response may be protective against viral infection. This study may have ramifications for the treatment of viral-induced asthma exacerbations.
Collapse
Affiliation(s)
- Toby C. Lewis
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
- Environmental Health Sciences, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
- Health Behavior/Health Education, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Ediri E. Metitiri
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
| | - Graciela B. Mentz
- Health Behavior/Health Education, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Xiaodan Ren
- Environmental Health Sciences, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Ashley R. Carpenter
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
| | - Adam M. Goldsmith
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
| | - Kyra E. Wicklund
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
- Epidemiology, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Breanna N. Eder
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
| | - Adam T. Comstock
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
| | - Jeannette M. Ricci
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
| | - Sean R. Brennan
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
| | - Ginger L. Washington
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
| | - Kendall B. Owens
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
| | - Bhramar Mukherjee
- Departments of Biostatistics, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Thomas G. Robins
- Environmental Health Sciences, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Stuart A. Batterman
- Environmental Health Sciences, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Marc B. Hershenson
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
- Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, USA
| | - the Community Action Against Asthma Steering Committee
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
- Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, USA
- Departments of Biostatistics, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
- Environmental Health Sciences, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
- Epidemiology, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
- Health Behavior/Health Education, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| |
Collapse
|
17
|
Lewis TC, Metitiri EE, Mentz GB, Ren X, Goldsmith AM, Eder BN, Wicklund KE, Walsh MP, Comstock AT, Ricci JM, Brennan SR, Washington GL, Owens KB, Mukherjee B, Robins TG, Batterman SA, Hershenson MB. Impact of community respiratory viral infections in urban children with asthma. Ann Allergy Asthma Immunol 2018; 122:175-183.e2. [PMID: 30385348 PMCID: PMC6360098 DOI: 10.1016/j.anai.2018.10.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/13/2018] [Accepted: 10/21/2018] [Indexed: 12/13/2022]
Abstract
Background Upper respiratory tract viral infections cause asthma exacerbations in children. However, the impact of natural colds on children with asthma in the community, particularly in the high-risk urban environment, is less well defined. Objective We hypothesized that children with high-symptom upper respiratory viral infections have reduced airway function and greater respiratory tract inflammation than children with virus-positive low-symptom illnesses or virus-negative upper respiratory tract symptoms. Methods We studied 53 children with asthma from Detroit, Michigan, during scheduled surveillance periods and self-reported respiratory illnesses for 1 year. Symptom score, spirometry, fraction of exhaled nitric oxide (FeNO), and nasal aspirate biomarkers, and viral nucleic acid and rhinovirus (RV) copy number were assessed. Results Of 658 aspirates collected, 22.9% of surveillance samples and 33.7% of respiratory illnesses were virus-positive. Compared with the virus-negative asymptomatic condition, children with severe colds (symptom score ≥5) showed reduced forced expiratory flow at 25% to 75% of the pulmonary volume (FEF25%-75%), higher nasal messenger RNA expression of C-X-C motif chemokine ligand (CXCL)-10 and melanoma differentiation-associated protein 5, and higher protein abundance of CXCL8, CXCL10 and C-C motif chemokine ligands (CCL)-2, CCL4, CCL20, and CCL24. Children with mild (symptom score, 1-4) and asymptomatic infections showed normal airway function and fewer biomarker elevations. Virus-negative cold-like illnesses demonstrated increased FeNO, minimal biomarker elevation, and normal airflow. The RV copy number was associated with nasal chemokine levels but not symptom score. Conclusion Urban children with asthma with high-symptom respiratory viral infections have reduced FEF25%-75% and more elevations of nasal biomarkers than children with mild or symptomatic infections, or virus-negative illnesses.
Collapse
Affiliation(s)
- Toby C Lewis
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan; Department of Environmental Health Sciences, University of Michigan School of Public Health; University of Michigan, Ann Arbor, Michigan; Department of Health Behavior/Health Education, University of Michigan School of Public Health; University of Michigan, Ann Arbor, Michigan
| | - Ediri E Metitiri
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Graciela B Mentz
- Department of Health Behavior/Health Education, University of Michigan School of Public Health; University of Michigan, Ann Arbor, Michigan
| | - Xiaodan Ren
- Department of Environmental Health Sciences, University of Michigan School of Public Health; University of Michigan, Ann Arbor, Michigan
| | - Adam M Goldsmith
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Breanna N Eder
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Kyra E Wicklund
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan; Department of Epidemiology, University of Michigan School of Public Health; University of Michigan, Ann Arbor, Michigan
| | - Megan P Walsh
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan; Department of Epidemiology, University of Michigan School of Public Health; University of Michigan, Ann Arbor, Michigan
| | - Adam T Comstock
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jeannette M Ricci
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Sean R Brennan
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Ginger L Washington
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Kendall B Owens
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Bhramar Mukherjee
- Department of Biostatistics, University of Michigan School of Public Health; University of Michigan, Ann Arbor, Michigan
| | - Thomas G Robins
- Department of Environmental Health Sciences, University of Michigan School of Public Health; University of Michigan, Ann Arbor, Michigan
| | - Stuart A Batterman
- Department of Environmental Health Sciences, University of Michigan School of Public Health; University of Michigan, Ann Arbor, Michigan
| | - Marc B Hershenson
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan.
| | | |
Collapse
|
18
|
Xi Y, Troy NM, Anderson D, Pena OM, Lynch JP, Phipps S, Bosco A, Upham JW. Critical Role of Plasmacytoid Dendritic Cells in Regulating Gene Expression and Innate Immune Responses to Human Rhinovirus-16. Front Immunol 2017; 8:1351. [PMID: 29118754 PMCID: PMC5660993 DOI: 10.3389/fimmu.2017.01351] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/03/2017] [Indexed: 11/16/2022] Open
Abstract
Though human rhinoviruses (HRVs) are usually innocuous viruses, they can trigger serious consequences in certain individuals, especially in the setting of impaired interferon (IFN) synthesis. Plasmacytoid dendritic cells (pDCs) are key IFN producing cells, though we know little about the role of pDC in HRV-induced immune responses. Herein, we used gene expression microarrays to examine HRV-activated peripheral blood mononuclear cells (PBMCs) from healthy people, in combination with pDC depletion, to assess whether observed gene expression patterns were pDC dependent. As expected, pDC depletion led to a major reduction in IFN-α release. This was associated with profound differences in gene expression between intact PBMC and pDC-depleted PBMC, and major changes in upstream regulators: 70–80% of the HRV activated genes appeared to be pDC dependent. Real-time PCR confirmed key changes in gene expression, in which the following selected genes were shown to be highly pDC dependent: the transcription factor IRF7, both IL-27 chains (IL-27p28 and EBI3), the alpha chain of the IL-15 receptor (IL-15RA) and the IFN-related gene IFI27. HRV-induced IL-6, IFN-γ, and IL-27 protein synthesis were also highly pDC dependent. Supplementing pDC-depleted cultures with recombinant IL-15, IFN-γ, IL-27, or IL-6 was able to restore the IFN-α response, thereby compensating for the absence of pDC. Though pDC comprise only a minority population of migratory leukocytes, our findings highlight the profound extent to which these cells contribute to the immune response to HRV.
Collapse
Affiliation(s)
- Yang Xi
- Lung and Allergy Research Center, Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Niamh M Troy
- Systems Immunology, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Denise Anderson
- Systems Immunology, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - Olga M Pena
- Lung and Allergy Research Center, Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Jason P Lynch
- Respiratory Immunology Group, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Simon Phipps
- Respiratory Immunology Group, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Anthony Bosco
- Systems Immunology, Telethon Kids Institute, The University of Western Australia, Perth, WA, Australia
| | - John W Upham
- Lung and Allergy Research Center, Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia.,Department of Respiratory Medicine, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| |
Collapse
|
19
|
Reza Etemadi M, Ling KH, Zainal Abidin S, Chee HY, Sekawi Z. Gene expression patterns induced at different stages of rhinovirus infection in human alveolar epithelial cells. PLoS One 2017; 12:e0176947. [PMID: 28558071 PMCID: PMC5448745 DOI: 10.1371/journal.pone.0176947] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 04/19/2017] [Indexed: 12/30/2022] Open
Abstract
Human rhinovirus (HRV) is the common virus that causes acute respiratory infection (ARI) and is frequently associated with lower respiratory tract infections (LRTIs). We aimed to investigate whether HRV infection induces a specific gene expression pattern in airway epithelial cells. Alveolar epithelial cell monolayers were infected with HRV species B (HRV-B). RNA was extracted from both supernatants and infected monolayer cells at 6, 12, 24 and 48 hours post infection (hpi) and transcriptional profile was analyzed using Affymetrix GeneChip and the results were subsequently validated using quantitative Real-time PCR method. HRV-B infects alveolar epithelial cells which supports implication of the virus with LRTIs. In total 991 genes were found differentially expressed during the course of infection. Of these, 459 genes were up-regulated whereas 532 genes were down-regulated. Differential gene expression at 6 hpi (187 genes up-regulated vs. 156 down-regulated) were significantly represented by gene ontologies related to the chemokines and inflammatory molecules indicating characteristic of viral infection. The 75 up-regulated genes surpassed the down-regulated genes (35) at 12 hpi and their enriched ontologies fell into discrete functional entities such as regulation of apoptosis, anti-apoptosis, and wound healing. At later time points of 24 and 48 hpi, predominated down-regulated genes were enriched for extracellular matrix proteins and airway remodeling events. Our data provides a comprehensive image of host response to HRV infection. The study suggests the underlying molecular regulatory networks genes which might be involved in pathogenicity of the HRV-B and potential targets for further validations and development of effective treatment.
Collapse
Affiliation(s)
- Mohammad Reza Etemadi
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Sciences, University Putra Malaysia, UPM Serdang, Selangor DE, Malaysia
| | - King-Hwa Ling
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor DE, Serdang, Selangor, Malaysia
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor DE, Serdang, Selangor, Malaysia
| | - Shahidee Zainal Abidin
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor DE, Serdang, Selangor, Malaysia
- Genetics and Regenerative Medicine Research Centre (GRMRC), Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor DE, Serdang, Selangor, Malaysia
| | - Hui-Yee Chee
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Sciences, University Putra Malaysia, UPM Serdang, Selangor DE, Malaysia
| | - Zamberi Sekawi
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Sciences, University Putra Malaysia, UPM Serdang, Selangor DE, Malaysia
- * E-mail:
| |
Collapse
|
20
|
Hansel TT, Tunstall T, Trujillo-Torralbo MB, Shamji B, Del-Rosario A, Dhariwal J, Kirk PDW, Stumpf MPH, Koopmann J, Telcian A, Aniscenko J, Gogsadze L, Bakhsoliani E, Stanciu L, Bartlett N, Edwards M, Walton R, Mallia P, Hunt TM, Hunt TL, Hunt DG, Westwick J, Edwards M, Kon OM, Jackson DJ, Johnston SL. A Comprehensive Evaluation of Nasal and Bronchial Cytokines and Chemokines Following Experimental Rhinovirus Infection in Allergic Asthma: Increased Interferons (IFN-γ and IFN-λ) and Type 2 Inflammation (IL-5 and IL-13). EBioMedicine 2017; 19:128-138. [PMID: 28373098 PMCID: PMC5440599 DOI: 10.1016/j.ebiom.2017.03.033] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/09/2017] [Accepted: 03/24/2017] [Indexed: 01/04/2023] Open
Abstract
Background Rhinovirus infection is a major cause of asthma exacerbations. Objectives We studied nasal and bronchial mucosal inflammatory responses during experimental rhinovirus-induced asthma exacerbations. Methods We used nasosorption on days 0, 2–5 and 7 and bronchosorption at baseline and day 4 to sample mucosal lining fluid to investigate airway mucosal responses to rhinovirus infection in patients with allergic asthma (n = 28) and healthy non-atopic controls (n = 11), by using a synthetic absorptive matrix and measuring levels of 34 cytokines and chemokines using a sensitive multiplex assay. Results Following rhinovirus infection asthmatics developed more upper and lower respiratory symptoms and lower peak expiratory flows compared to controls (all P < 0.05). Asthmatics also developed higher nasal lining fluid levels of an anti-viral pathway (including IFN-γ, IFN-λ/IL-29, CXCL11/ITAC, CXCL10/IP10 and IL-15) and a type 2 inflammatory pathway (IL-4, IL-5, IL-13, CCL17/TARC, CCL11/eotaxin, CCL26/eotaxin-3) (area under curve day 0–7, all P < 0.05). Nasal IL-5 and IL-13 were higher in asthmatics at day 0 (P < 0.01) and levels increased by days 3 and 4 (P < 0.01). A hierarchical correlation matrix of 24 nasal lining fluid cytokine and chemokine levels over 7 days demonstrated expression of distinct interferon-related and type 2 pathways in asthmatics. In asthmatics IFN-γ, CXCL10/IP10, CXCL11/ITAC, IL-15 and IL-5 increased in bronchial lining fluid following viral infection (all P < 0.05). Conclusions Precision sampling of mucosal lining fluid identifies robust interferon and type 2 responses in the upper and lower airways of asthmatics during an asthma exacerbation. Nasosorption and bronchosorption have potential to define asthma endotypes in stable disease and at exacerbation. Following rhinovirus infection asthmatics have increased interferons and type 2 inflammation in airway mucosal lining fluid. Nasosorption cytokines and chemokines showed distinct pathways of interferon and type 2 inflammation in asthma. Precision mucosal sampling has potential for stratifying molecular endotypes of asthma. Validation of nasosorption and bronchosorption will be required for selection of asthmatics for therapy with biologics.
Experimental human rhinovirus (HRV) infection causes more severe upper and lower respiratory tract symptoms in allergic asthmatics than in healthy controls. There is greater induction of cytokines and chemokines in nasal and bronchial mucosal lining fluid (MLF) of asthmatics: with distinct pathways of type 2 and anti-viral/regulatory inflammation. Subject to further validation, analysis of MLF may prove useful in stratification of patients with asthma, and the definition of molecular endotypes. Interpretation Nasosorption and bronchosorption are precision sampling methods with potential for widespread application in respiratory and other mucosal diseases (e.g. gastrointestinal diseases). Biomarkers identified in nasosorption and bronchosorption samples will need to be validated compared to established airway sampling methods, in a range of asthma phenotypes, and with current and novel therapies.
Collapse
Affiliation(s)
- Trevor T Hansel
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Imperial College Healthcare NHS Trust, UK; Imperial Clinical Respiratory Research Unit (ICRRU), UK.
| | - Tanushree Tunstall
- Imperial College Healthcare NHS Trust, UK; Imperial Clinical Respiratory Research Unit (ICRRU), UK
| | - Maria-Belen Trujillo-Torralbo
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Imperial College Healthcare NHS Trust, UK
| | - Betty Shamji
- Novartis Institute for Biomedical Research, Horsham, UK
| | - Ajerico Del-Rosario
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Imperial College Healthcare NHS Trust, UK
| | - Jaideep Dhariwal
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Imperial College Healthcare NHS Trust, UK
| | - Paul D W Kirk
- MRC Biostatistics Unit, Cambridge Institute of Public Health, Cambridge, UK
| | | | - Jens Koopmann
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; Medimmune, Cambridge, UK
| | - Aurica Telcian
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Julia Aniscenko
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Leila Gogsadze
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Eteri Bakhsoliani
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Luminita Stanciu
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Nathan Bartlett
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Michael Edwards
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Ross Walton
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Patrick Mallia
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Imperial College Healthcare NHS Trust, UK
| | - Toby M Hunt
- Hunt Developments (UK) Ltd, Midhurst, West Sussex, UK
| | - Trevor L Hunt
- Hunt Developments (UK) Ltd, Midhurst, West Sussex, UK
| | - Duncan G Hunt
- Hunt Developments (UK) Ltd, Midhurst, West Sussex, UK
| | - John Westwick
- Novartis Institute for Biomedical Research, Horsham, UK
| | | | - Onn Min Kon
- Imperial College Healthcare NHS Trust, UK; Imperial Clinical Respiratory Research Unit (ICRRU), UK
| | - David J Jackson
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Guy's and St Thomas' NHS Trust
| | - Sebastian L Johnston
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Imperial College Healthcare NHS Trust, UK
| |
Collapse
|
21
|
Hayney MS, Henriquez KM, Barnet JH, Ewers T, Champion HM, Flannery S, Barrett B. Serum IFN-γ-induced protein 10 (IP-10) as a biomarker for severity of acute respiratory infection in healthy adults. J Clin Virol 2017; 90:32-37. [PMID: 28334685 PMCID: PMC5408957 DOI: 10.1016/j.jcv.2017.03.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 02/20/2017] [Accepted: 03/05/2017] [Indexed: 01/08/2023]
Abstract
Serum IP-10 concentrations from 225 ARI episodes correlated with ARI severity for the episode. IP-10 concentrations varied with the viral pathogen that was identified. IP-10 may be a biomarker for ARI severity and for presence of a viral pathogen.
Background The inflammatory chemokine, interferon-gamma inducible protein of 10 kDa (IP-10), is a biomarker associated with several conditions. Objectives This study investigated serum concentrations of IP-10 in healthy individuals who developed acute respiratory infection (ARI). The hypothesis is that serum IP-10 concentrations correlate with ARI severity and detection of viral pathogens. Study design Data come from a randomized controlled trial measuring the effects of mindfulness meditation or exercise on ARI (Clinical Trials ID: NCT01654289). Healthy adults ages 30–69 were followed for a single season for ARI incidence and severity. This trial is ongoing, and the investigators are still blinded. When a participant reported ARI symptoms, nasal swab and lavage for PCR-based viral identification and blood samples were collected within the first 72 h of ARI symptoms. Serum IP-10 concentrations were measured by ELISA (R&D Systems, Inc., Quantikine ELISA, Minneapolis, MN). ARI severity was measured using the validated Wisconsin Upper Respiratory Symptom Survey (WURSS-24) until the ARI episode resolved. Results Serum IP-10 concentrations from 225 ARI episodes correlated with ARI global severity (rho 0.28 [95% CI: 0.15–0.39]; p < 0.001). IP-10 concentrations were higher with an ARI in which a viral pathogen was detected compared to no viral pathogen detected (median 366 pg/ml [IQR: 227–486] vs 163 pg/ml [IQR: 127–295], p < 0.0001). Influenza infections had higher IP-10 concentrations than coronavirus, enterovirus or rhinovirus, and paramyxovirus. Conclusion Serum IP-10 concentration correlates with ARI global severity. Also, IP-10 concentration measured early in the course of the ARI correlates with the daily severity, duration, and illness symptoms.
Collapse
Affiliation(s)
- Mary S Hayney
- School of Pharmacy, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States.
| | - Kelsey M Henriquez
- School of Pharmacy, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
| | - Jodi H Barnet
- Department of Family Medicine, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
| | - Tola Ewers
- Department of Family Medicine, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
| | - Heather M Champion
- School of Pharmacy, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
| | - Sean Flannery
- School of Pharmacy, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
| | - Bruce Barrett
- Department of Family Medicine, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
22
|
Song DJ. Rhinovirus and childhood asthma: an update. KOREAN JOURNAL OF PEDIATRICS 2016; 59:432-439. [PMID: 27895690 PMCID: PMC5118502 DOI: 10.3345/kjp.2016.59.11.432] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 10/18/2015] [Accepted: 10/23/2015] [Indexed: 01/26/2023]
Abstract
Asthma is recognized as a complex disease resulting from interactions between multiple genetic and environmental factors. Accumulating evidence suggests that respiratory viral infections in early life constitute a major environmental risk factor for the development of childhood asthma. Respiratory viral infections have also been recognized as the most common cause of asthma exacerbation. The advent of molecular diagnostics to detect respiratory viruses has provided new insights into the role of human rhinovirus (HRV) infections in the pathogenesis of asthma. However, it is still unclear whether HRV infections cause asthma or if wheezing with HRV infection is simply a predictor of childhood asthma. Recent clinical and experimental studies have identified plausible pathways by which HRV infection could cause asthma, particularly in a susceptible host, and exacerbate disease. Airway epithelial cells, the primary site of infection and replication of HRV, play a key role in these processes. Details regarding the role of genetic factors, including ORMDL3, are beginning to emerge. This review discusses recent clinical and experimental evidence for the role of HRV infection in the development and exacerbation of childhood asthma and the potential underlying mechanisms that have been proposed.
Collapse
Affiliation(s)
- Dae Jin Song
- Department of Pediatrics, Korea University College of Medicine, Seoul, Korea.; Environmental Health Center for Childhood Asthma, Korea University Anam Hospital, Seoul, Korea
| |
Collapse
|
23
|
Abstract
Allergy and viral respiratory infections have long been recognized as two of the most important risk factors for exacerbations of asthma. These observations have raised questions regarding potential interactions between these two important risk factors. For example, does allergy diminish the antiviral response, thereby promoting exacerbations of asthma? Alternately, do viral respiratory infections potentiate ongoing allergic inflammation in the airway? The answers to these questions are likely to have implications regarding the prevention and treatment of exacerbations of asthma. This article reviews that clinical evidence linking viral infections and allergy to exacerbations of asthma, reviews potential interactions between these two risk factors, and discusses possible application of new insights in virus/allergen interactions to the prevention and treatment of exacerbations of asthma.
Collapse
|
24
|
Nguyen TH, Maltby S, Simpson JL, Eyers F, Baines KJ, Gibson PG, Foster PS, Yang M. TNF-α and Macrophages Are Critical for Respiratory Syncytial Virus-Induced Exacerbations in a Mouse Model of Allergic Airways Disease. THE JOURNAL OF IMMUNOLOGY 2016; 196:3547-58. [PMID: 27036916 DOI: 10.4049/jimmunol.1502339] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 02/29/2016] [Indexed: 12/27/2022]
Abstract
Viral respiratory infections trigger severe exacerbations of asthma, worsen disease symptoms, and impair lung function. To investigate the mechanisms underlying viral exacerbation, we established a mouse model of respiratory syncytial virus (RSV)-induced exacerbation after allergen sensitization and challenge. RSV infection of OVA-sensitized/challenged BALB/c mice resulted in significantly increased airway hyperresponsiveness (AHR) and macrophage and neutrophil lung infiltration. Exacerbation was accompanied by increased levels of inflammatory cytokines (including TNF-α, MCP-1, and keratinocyte-derived protein chemokine [KC]) compared with uninfected OVA-treated mice or OVA-treated mice exposed to UV-inactivated RSV. Dexamethasone treatment completely inhibited all features of allergic disease, including AHR and eosinophil infiltration, in uninfected OVA-sensitized/challenged mice. Conversely, dexamethasone treatment following RSV-induced exacerbation only partially suppressed AHR and failed to dampen macrophage and neutrophil infiltration or inflammatory cytokine production (TNF-α, MCP-1, and KC). This mimics clinical observations in patients with exacerbations, which is associated with increased neutrophils and often poorly responds to corticosteroid therapy. Interestingly, we also observed increased TNF-α levels in sputum samples from patients with neutrophilic asthma. Although RSV-induced exacerbation was resistant to steroid treatment, inhibition of TNF-α and MCP-1 function or depletion of macrophages suppressed features of disease, including AHR and macrophage and neutrophil infiltration. Our findings highlight critical roles for macrophages and inflammatory cytokines (including TNF-α and MCP-1) in viral-induced exacerbation of asthma and suggest examination of these pathways as novel therapeutic approaches for disease management.
Collapse
Affiliation(s)
- Thi Hiep Nguyen
- Priority Research Centre for Asthma and Respiratory Diseases, Faculty of Health, University of Newcastle, Callaghan, New South Wales 2300, Australia; Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales 2300, Australia; and
| | - Steven Maltby
- Priority Research Centre for Asthma and Respiratory Diseases, Faculty of Health, University of Newcastle, Callaghan, New South Wales 2300, Australia; Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales 2300, Australia; and
| | - Jodie L Simpson
- Priority Research Centre for Asthma and Respiratory Diseases, Faculty of Health, University of Newcastle, Callaghan, New South Wales 2300, Australia; Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales 2300, Australia; and Department of Respiratory and Sleep Medicine, Hunter New England Area Health Service, Newcastle, New South Wales 2305, Australia
| | - Fiona Eyers
- Priority Research Centre for Asthma and Respiratory Diseases, Faculty of Health, University of Newcastle, Callaghan, New South Wales 2300, Australia; Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales 2300, Australia; and
| | - Katherine J Baines
- Priority Research Centre for Asthma and Respiratory Diseases, Faculty of Health, University of Newcastle, Callaghan, New South Wales 2300, Australia; Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales 2300, Australia; and Department of Respiratory and Sleep Medicine, Hunter New England Area Health Service, Newcastle, New South Wales 2305, Australia
| | - Peter G Gibson
- Priority Research Centre for Asthma and Respiratory Diseases, Faculty of Health, University of Newcastle, Callaghan, New South Wales 2300, Australia; Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales 2300, Australia; and Department of Respiratory and Sleep Medicine, Hunter New England Area Health Service, Newcastle, New South Wales 2305, Australia
| | - Paul S Foster
- Priority Research Centre for Asthma and Respiratory Diseases, Faculty of Health, University of Newcastle, Callaghan, New South Wales 2300, Australia; Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales 2300, Australia; and
| | - Ming Yang
- Priority Research Centre for Asthma and Respiratory Diseases, Faculty of Health, University of Newcastle, Callaghan, New South Wales 2300, Australia; Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales 2300, Australia; and
| |
Collapse
|
25
|
Bosco A, Wiehler S, Proud D. Interferon regulatory factor 7 regulates airway epithelial cell responses to human rhinovirus infection. BMC Genomics 2016; 17:76. [PMID: 26810609 PMCID: PMC4727386 DOI: 10.1186/s12864-016-2405-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/18/2016] [Indexed: 12/11/2022] Open
Abstract
Background Human rhinoviruses (HRV) cause the majority of colds and trigger exacerbations of chronic lower airway diseases. Airway epithelial cells are the primary site for HRV infection and replication, and the initiation of host inflammatory responses. At present, the molecular mechanisms that underpin HRV responses in airway epithelial cells are incompletely understood. The aim of this study was to employ microarray profiling, upstream regulator analysis, and siRNA mediated gene silencing to further our understanding of the role of interferon regulatory factor 7 (IRF7) in this response. Methods Primary human bronchial epithelial cells (HBE) where transfected with siRNA that targets IRF7 or a non-silencing control (all-star control) using Lipofectamine. The cells were allowed to recover, and then cultured in the presence or absence of HRV-16 for 24 h. Global patterns of gene expression were profiled on microarrays. A subset of genes identified in the microarray study were validated at the mRNA and/or protein level using real time RT-qPCR, ELISA, and western blots. Results Hundreds of genes were upregulated in HBE during HRV infection. Pathways analysis demonstrated that these genes were mainly involved in type I and II interferon signaling, RIG-I/MDA5 signaling, antigen processing and presentation, and apoptosis. Upstream regulator analysis of these data suggested that IRF7 was a major molecular driver of this response. Knockdown of IRF7 reduced the HRV-driven upregulation of genes involved in antiviral responses (interferon signaling, Toll-like receptor signaling, NOD-like receptor signaling, RIG-I/MDA5 signaling), and increased the expression of genes that promote inflammation (e.g. CXCL5, IL-33, IL1RL1) and the response to oxidative stress. However, the majority of genes that were perturbed by HRV in HBE cells including those that are known to be regulated by IRF7 were insensitive to IRF7 knockdown. Upstream regulator analysis of the part of the response that was insensitive to IRF7 knockdown suggested it was driven by NF-κB, STAT1, STAT3, and IRF1. Conclusions Our findings demonstrate that IRF7 regulates the expression of genes involved in antiviral immunity, inflammation, and the response to oxidative stress during HRV infections in HBE cells, and also suggests that other transcription factors play a major role in this response. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2405-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anthony Bosco
- Telethon Kids Institute, University of Western Australia, PO Box 855, West Perth, WA, 6872, Australia.
| | - Shahina Wiehler
- Airway Inflammation Research Group, Snyder Institute for Chronic Diseases, and the Department of Physiology & Pharmacology, University of Calgary Faculty of Medicine, Calgary, AB, Canada.
| | - David Proud
- Airway Inflammation Research Group, Snyder Institute for Chronic Diseases, and the Department of Physiology & Pharmacology, University of Calgary Faculty of Medicine, Calgary, AB, Canada.
| |
Collapse
|
26
|
Wang Q, Chen D, Xie H, Lin X, Wang X, Yao Q, Zheng X, Xu C, Chen L, He S, Zhang H. Altered Expression of IFN-λ2 in Allergic Airway Disorders and Identification of Its Cell Origins. Mediators Inflamm 2016; 2016:5759496. [PMID: 27057098 PMCID: PMC4737047 DOI: 10.1155/2016/5759496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 11/06/2015] [Accepted: 11/22/2015] [Indexed: 02/05/2023] Open
Abstract
This study investigated the expression levels of interferon- (IFN-) λ2 in peripheral blood and tissues. The results showed that the levels of IFN-λ2 were elevated by 17.9% and 14.2% in the plasma of allergic rhinitis (AR) and combined rhinitis with asthma (AR + AS), which was positively correlated with the level of tryptase but negatively correlated with the level of IL-10. IFN-λ2 was predominately expressed in the CD16+ cells and CD14+ cells in healthy control subjects (HC) but upregulated only in CD8+ cells of AR and in eosinophils of asthma. It was observed that approximately 6.6% and 7.0% dispersed tonsil cells and 5.8% and 0.44% dispersed lung cells are IFN-λ2+ mast cells and macrophages. Moreover, tryptase and agonist peptides of PAR-2 induced enhanced IFN-λ2 mRNA expression in A549 cells. In conclusion, the elevated levels of IFN-λ2 in the plasma of AR and AR + AS indicate that IFN-λ2 is likely to contribute to the pathogenesis of allergic airway disorders. The potential origins of the elevated plasma IFN-λ2 include mast cells, macrophages, and epithelial cells in tissues, neutrophils, monocytes, CD8+ T cells, and eosinophils in peripheral blood. Development of IFN-λ2 related therapy may help to treat or prevent allergic airway disorders.
Collapse
Affiliation(s)
- Qiuli Wang
- Department of ENT, Allergy and Clinical Immunology Research Centre, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning 121001, China
| | - Dong Chen
- Department of ENT, Allergy and Clinical Immunology Research Centre, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning 121001, China
| | - Hua Xie
- Department of Respiratory Medicine, The General Hospital of Shenyang Military Region, Shenyang, Liaoning 110016, China
| | - Xiaoping Lin
- Department of Respiratory Medicine, The General Hospital of Shenyang Military Region, Shenyang, Liaoning 110016, China
| | - Xuefeng Wang
- Department of ENT, Allergy and Clinical Immunology Research Centre, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning 121001, China
| | - Qijian Yao
- Allergy and Inflammation Research Institute, Shantou University Medical College, Shantou 515031, China
| | - Xiaoxuan Zheng
- Allergy and Inflammation Research Institute, Shantou University Medical College, Shantou 515031, China
| | - Chiyan Xu
- Allergy and Inflammation Research Institute, Shantou University Medical College, Shantou 515031, China
| | - Lingfei Chen
- Allergy and Inflammation Research Institute, Shantou University Medical College, Shantou 515031, China
| | - Shaoheng He
- Department of ENT, Allergy and Clinical Immunology Research Centre, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning 121001, China
| | - Huiyun Zhang
- Department of ENT, Allergy and Clinical Immunology Research Centre, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning 121001, China
| |
Collapse
|
27
|
Pancham K, Perez GF, Huseni S, Jain A, Kurdi B, Rodriguez-Martinez CE, Preciado D, Rose MC, Nino G. Premature infants have impaired airway antiviral IFNγ responses to human metapneumovirus compared to respiratory syncytial virus. Pediatr Res 2015; 78:389-94. [PMID: 26086642 PMCID: PMC5529168 DOI: 10.1038/pr.2015.113] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 03/09/2015] [Indexed: 11/09/2022]
Abstract
BACKGROUND It is unknown why human metapneumovirus (HMPV) and respiratory syncytial virus (RSV) cause severe respiratory infection in children, particularly in premature infants. Our aim was to investigate if there are defective airway antiviral responses to these viruses in young children with history of prematurity. METHODS Nasal airway secretions were collected from 140 children ≤ 3 y old without detectable virus (n = 80) or with PCR-confirmed HMPV or RSV infection (n = 60). Nasal protein levels of IFNγ, CCL5/RANTES, IL-10, IL-4, and IL-17 were determined using a multiplex magnetic bead immunoassay. RESULTS Full-term children with HMPV and RSV infection had increased levels of nasal airway IFNγ, CCL5, and IL-10 along with an elevation in Th1 (IFNγ)/Th2 (IL-4) ratios, which is expected during antiviral responses. In contrast, HMPV-infected premature children (< 32 wk gestation) did not exhibit increased Th1/Th2 ratios or elevated nasal airway secretion of IFNγ, CCL5, and IL-10 relative to uninfected controls. CONCLUSION Our study is the first to demonstrate that premature infants have defective IFNγ, CCL5/RANTES, and IL-10 airway responses during HMPV infection and provides novel insights about the potential reason why HMPV causes severe respiratory disease in children with history of prematurity.
Collapse
Affiliation(s)
- Krishna Pancham
- Division of Pulmonary and Sleep Medicine, Children’s National Medical Center, Washington, DC
| | - Geovanny F. Perez
- Division of Pulmonary and Sleep Medicine, Children’s National Medical Center, Washington, DC,Department of Pediatrics, George Washington University, Washington, DC,Department of Integrative Systems Biology, George Washington University, Washington, DC,Center for Genetic Research Medicine, Children’s National Medical Center, Washington, DC
| | - Shehlanoor Huseni
- Division of Pulmonary and Sleep Medicine, Children’s National Medical Center, Washington, DC
| | - Amisha Jain
- Division of Pulmonary and Sleep Medicine, Children’s National Medical Center, Washington, DC
| | - Bassem Kurdi
- Department of Pediatrics, George Washington University, Washington, DC
| | - Carlos E. Rodriguez-Martinez
- Department of Pediatrics, School of Medicine, Universidad Nacional de Colombia, Bogota, Colombia,Department of Pediatric Pulmonology and Pediatric Critical Care Medicine, School of Medicine, Universidad El Bosque, Bogota, Colombia,Research Unit, Military Hospital of Colombia, Bogota, Colombia
| | - Diego Preciado
- Department of Pediatrics, George Washington University, Washington, DC,Department of Integrative Systems Biology, George Washington University, Washington, DC,Center for Genetic Research Medicine, Children’s National Medical Center, Washington, DC,Division of Pediatric Otorhinolaryngology, Department of Surgery, George Washington University, Washington, DC,Division of Pediatric Otorhinolaryngology, Department of Pediatrics, George Washington University, Washington, DC
| | - Mary C. Rose
- Division of Pulmonary and Sleep Medicine, Children’s National Medical Center, Washington, DC,Department of Pediatrics, George Washington University, Washington, DC,Department of Integrative Systems Biology, George Washington University, Washington, DC,Center for Genetic Research Medicine, Children’s National Medical Center, Washington, DC,Department of Biochemistry and Molecular Medicine, George Washington University, Washington, DC
| | - Gustavo Nino
- Division of Pulmonary and Sleep Medicine, Children’s National Medical Center, Washington, DC,Department of Pediatrics, George Washington University, Washington, DC,Department of Integrative Systems Biology, George Washington University, Washington, DC,Center for Genetic Research Medicine, Children’s National Medical Center, Washington, DC
| |
Collapse
|
28
|
Bertrand P, Lay MK, Piedimonte G, Brockmann PE, Palavecino CE, Hernández J, León MA, Kalergis AM, Bueno SM. Elevated IL-3 and IL-12p40 levels in the lower airway of infants with RSV-induced bronchiolitis correlate with recurrent wheezing. Cytokine 2015; 76:417-423. [PMID: 26299549 DOI: 10.1016/j.cyto.2015.07.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 07/17/2015] [Accepted: 07/20/2015] [Indexed: 12/30/2022]
Abstract
Respiratory Syncytial Virus (RSV) is the first cause of hospitalization due to bronchiolitis in infants. RSV bronchiolitis has been linked to asthma and recurrent wheezing, however the mechanisms behind this association have not been elucidated. Here, we evaluated the cytokine and chemokine profiles in the airways in infants with RSV bronchiolitis. Nasopharyngeal Aspirates (NPA) and Bronchoalveolar Lavage Fluids (BALF) from infants hospitalized due to RSV bronchiolitis and healthy controls were analyzed for cytokine and chemokine production. We observed elevated levels of Th2 cytokines (IL-3, IL-4, IL-10 and IL-13), pro-inflammatory cytokines and chemokines (IL-1β, IL-6, TNF-β, MCP-1/CCL2, MIP-1α/CCL3 and IL-8/CXCL8) in BALF from infants with RSV bronchiolitis, as compared to controls. We found a direct correlation of IL-3 and IL-12p40 levels with the development of recurrent wheezing later in life. These results suggest that IL-3 and IL-12p40 could be considered as molecular predictors for recurrent wheezing due to RSV infection.
Collapse
Affiliation(s)
- Pablo Bertrand
- División de Pediatría, Unidad de Enfermedades Respiratorias Pediátricas, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Margarita K Lay
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Giovanni Piedimonte
- The Cleveland Clinic Pediatric Institute and Children's Hospital, Cleveland, OH, United States
| | - Pablo E Brockmann
- División de Pediatría, Unidad de Enfermedades Respiratorias Pediátricas, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Christian E Palavecino
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jury Hernández
- División de Pediatría, Unidad de Enfermedades Respiratorias Pediátricas, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Miguel A León
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Reumatología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; INSERM U1064, Nantes, France
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Reumatología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
29
|
Perez GF, Rodriguez-Martinez CE, Nino G. Rhinovirus-Induced Airway Disease: A Model to Understand the Antiviral and Th2 Epithelial Immune Dysregulation in Childhood Asthma. J Investig Med 2015; 63:792-5. [PMID: 26057561 PMCID: PMC4512841 DOI: 10.1097/jim.0000000000000209] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Rhinovirus (RV) infections account for most asthma exacerbations among children and adults, yet the fundamental mechanism responsible for why asthmatics are more susceptible to RV than otherwise healthy individuals remains largely unknown. Nonetheless, the use of models to understand the mechanisms of RV-induced airway disease in asthma has dramatically expanded our knowledge about the cellular and molecular pathogenesis of the disease. For instance, ground-breaking studies have recently established that the susceptibility to RV in asthmatic subjects is associated with a dysfunctional airway epithelial inflammatory response generated after innate recognition of viral-related molecules, such as double-stranded RNA. This review summarizes the novel cardinal features of the asthmatic condition identified in the past few years through translational and experimental RV-based approaches. Specifically, we discuss the evidence demonstrating the presence of an abnormal innate antiviral immunity (airway epithelial secretion of types I and III interferons), exaggerated production of the master Th2 molecule thymic stromal lymphopoietin, and altered antimicrobial host defense in the airways of asthmatic individuals with acute RV infection.
Collapse
Affiliation(s)
- Geovanny F. Perez
- Division of Pulmonary and Sleep Medicine, Children’s National Medical Center, Washington, DC
- Departments of Pediatrics and Integrative Systems Biology, George Washington University, Washington, DC
- Center for Genetic Research Medicine, Children’s National Medical Center, Washington, DC
| | - Carlos E. Rodriguez-Martinez
- Department of Pediatrics, School of Medicine, Universidad Nacional de Colombia, Bogota, Colombia
- Department of Pediatric Pulmonology and Pediatric Critical Care Medicine, School of Medicine, Universidad El Bosque, Bogota, Colombia
- Research Unit, Military Hospital of Colombia, Bogota, Colombia
| | - Gustavo Nino
- Division of Pulmonary and Sleep Medicine, Children’s National Medical Center, Washington, DC
- Departments of Pediatrics and Integrative Systems Biology, George Washington University, Washington, DC
- Center for Genetic Research Medicine, Children’s National Medical Center, Washington, DC
| |
Collapse
|
30
|
Girkin J, Hatchwell L, Foster P, Johnston SL, Bartlett N, Collison A, Mattes J. CCL7 and IRF-7 Mediate Hallmark Inflammatory and IFN Responses following Rhinovirus 1B Infection. THE JOURNAL OF IMMUNOLOGY 2015; 194:4924-30. [PMID: 25847975 DOI: 10.4049/jimmunol.1401362] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 03/13/2015] [Indexed: 12/22/2022]
Abstract
Rhinovirus (RV) infections are common and have the potential to exacerbate asthma. We have determined the lung transcriptome in RV strain 1B-infected naive BALB/c mice (nonallergic) and identified CCL7 and IFN regulatory factor (IRF)-7 among the most upregulated mRNA transcripts in the lung. To investigate their roles we employed anti-CCL7 Abs and an IRF-7-targeting small interfering RNA in vivo. Neutralizing CCL7 or inhibiting IRF-7 limited neutrophil and macrophage influx and IFN responses in nonallergic mice. Neutralizing CCL7 also reduced activation of NF-κB p65 and p50 subunits, as well as airway hyperreactivity (AHR) in nonallergic mice. However, neither NF-κB subunit activation nor AHR was abolished with infection of allergic mice after neutralizing CCL7, despite a reduction in the number of neutrophils, macrophages, and eosinophils. IRF-7 small interfering RNA primarily suppressed IFN-α and IFN-β levels during infection of allergic mice. Our data highlight a pivotal role of CCL7 and IRF-7 in RV-induced inflammation and IFN responses and link NF-κB signaling to the development of AHR.
Collapse
Affiliation(s)
- Jason Girkin
- Experimental and Translational Respiratory Medicine Group, University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales 2305, Australia; Priority Research Centre for Asthma and Respiratory Diseases, University of Newcastle, Newcastle, New South Wales 2305, Australia
| | - Luke Hatchwell
- Experimental and Translational Respiratory Medicine Group, University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales 2305, Australia; Priority Research Centre for Asthma and Respiratory Diseases, University of Newcastle, Newcastle, New South Wales 2305, Australia
| | - Paul Foster
- Priority Research Centre for Asthma and Respiratory Diseases, University of Newcastle, Newcastle, New South Wales 2305, Australia
| | - Sebastian L Johnston
- Airway Disease Infection Section, National Heart and Lung Institute, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London W2 1PG, United Kingdom; and
| | - Nathan Bartlett
- Airway Disease Infection Section, National Heart and Lung Institute, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London W2 1PG, United Kingdom; and
| | - Adam Collison
- Experimental and Translational Respiratory Medicine Group, University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales 2305, Australia; Priority Research Centre for Asthma and Respiratory Diseases, University of Newcastle, Newcastle, New South Wales 2305, Australia
| | - Joerg Mattes
- Experimental and Translational Respiratory Medicine Group, University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales 2305, Australia; Priority Research Centre for Asthma and Respiratory Diseases, University of Newcastle, Newcastle, New South Wales 2305, Australia; Paediatric Respiratory and Sleep Medicine Unit, Newcastle Children's Hospital, Kaleidoscope, Newcastle, New South Wales 2305, Australia
| |
Collapse
|
31
|
Feldman AS, He Y, Moore ML, Hershenson MB, Hartert TV. Toward primary prevention of asthma. Reviewing the evidence for early-life respiratory viral infections as modifiable risk factors to prevent childhood asthma. Am J Respir Crit Care Med 2015; 191:34-44. [PMID: 25369458 DOI: 10.1164/rccm.201405-0901pp] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
A first step in primary disease prevention is identifying common, modifiable risk factors that contribute to a significant proportion of disease development. Infant respiratory viral infection and childhood asthma are the most common acute and chronic diseases of childhood, respectively. Common clinical features and links between these diseases have long been recognized, with early-life respiratory syncytial virus (RSV) and rhinovirus (RV) lower respiratory tract infections (LRTIs) being strongly associated with increased asthma risk. However, there has long been debate over the role of these respiratory viruses in asthma inception. In this article, we systematically review the evidence linking early-life RSV and RV LRTIs with asthma inception and whether they could therefore be targets for primary prevention efforts.
Collapse
Affiliation(s)
- Amy S Feldman
- 1 Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, and Center for Asthma Research, Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | | | | | | |
Collapse
|
32
|
Perez GF, Pancham K, Huseni S, Jain A, Rodriguez-Martinez CE, Preciado D, Rose MC, Nino G. Rhinovirus-induced airway cytokines and respiratory morbidity in severely premature children. Pediatr Allergy Immunol 2015; 26:145-52. [PMID: 25640734 PMCID: PMC5542573 DOI: 10.1111/pai.12346] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/27/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND Rhinovirus (RV) has been linked to the pathogenesis of asthma. Prematurity is a risk factor for severe RV infection in early life, but is unknown if RV elicits enhanced pro-asthmatic airway cytokine responses in premature infants. This study investigated whether young children born severely premature (<32 wks gestation) exhibit airway secretion of Th2 and Th17 cytokines during natural RV infections and whether RV-induced Th2-Th17 responses are linked to more respiratory morbidity in premature children during the first 2 yrs of life. METHODS We measured Th2 and Th17 nasal airway cytokines in a retrospective cohort of young children aged 0-2 yrs with PCR-confirmed RV infection or non-detectable virus. Protein levels of IL-4, IL-13, TSLP, and IL-17 were determined with multiplex immunoassays. Demographic and clinical variables were obtained by electronic medical record (EMR) review. RESULTS The study comprised 214 children born full term (n = 108), preterm (n = 44) or severely premature (n = 62). Natural RV infection in severely premature children was associated with elevated airway secretion of Th2 (IL-4 and IL-13) and Th17 (IL-17) cytokines, particularly in subjects with history of bronchopulmonary dysplasia. Severely premature children with high RV-induced airway IL-4 had recurrent respiratory hospitalizations (median 3.65 hosp/yr; IQR 2.8-4.8) and were more likely to have at least one pediatric intensive care unit admission during the first 2 yrs of life (OR 8.72; 95% CI 1.3-58.7; p = 0.02). CONCLUSIONS Severely premature children have increased airway secretion of Th2 and Th17 cytokines during RV infections, which is associated with more respiratory morbidity in the first 2 yrs of life.
Collapse
Affiliation(s)
- Geovanny F Perez
- Division of Pulmonary and Sleep Medicine, Children's National Medical Center, Washington, DC, USA; Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, USA; Department of Integrative Systems Biology, George Washington University, Washington, DC, USA; Center for Genetic Research Medicine, Children's National Medical Center, Washington, DC, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Thomson NC, Charron CE, Chaudhuri R, Spears M, Ito K, McSharry C. Atorvastatin in combination with inhaled beclometasone modulates inflammatory sputum mediators in smokers with asthma. Pulm Pharmacol Ther 2015; 31:1-8. [PMID: 25595138 DOI: 10.1016/j.pupt.2015.01.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 01/05/2015] [Accepted: 01/06/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Statins have pleiotropic immunomodulatory effects that may be beneficial in the treatment of asthma. We previously reported that treatment with atorvastatin improved asthma symptoms in smokers with asthma in the absence of a change in the concentration of a selection of sputum inflammatory mediators. OBJECTIVE To determine the effects of atorvastatin alone and in combination with inhaled corticosteroid on a range of sputum cytokines, chemokines and growth factors implicated in the pathogenesis of asthma, and their association with asthma control questionnaire (ACQ) and/or asthma quality of life questionnaire (AQLQ) scores. METHODS Sputum samples were analysed from a sub-group of 39 smokers with mild to moderate asthma recruited to a randomised controlled trial comparing atorvastatin (40 mg/day) versus placebo for four weeks, followed by inhaled beclometasone (400 μg/day) for a further four weeks. Induced sputum supernatant fluid was analysed (Luminex or biochemical analyses) for concentrations of 35 mediators. RESULTS Sputum mediator concentrations were not reduced by inhaled beclometasone alone. Atorvastatin significantly reduced sputum concentrations of CCL7, IL-12p70, sCD40L, FGF-2, CCL4, TGF-α and MMP-8 compared with placebo and, when combined with inhaled beclometasone, reduced sputum concentrations of MMP-8, IL-1β, IL-10, MMP-9, sCD40L, FGF-2, IL-7, G-CSF and CCL7 compared to ICS alone. Improvements in ACQ and/or AQLQ scores with atorvastatin and ICS were associated with decreases in G-CSF, IL-7, CCL2 and CXCL8. CONCLUSION Short-term treatment with atorvastatin alone or in combination with inhaled beclometasone reduces several sputum cytokines, chemokines and growth factors concentrations unresponsive to inhaled corticosteroids alone, in smokers with asthma.
Collapse
Affiliation(s)
- Neil C Thomson
- Respiratory Medicine, Institute of Infection, Immunity and Inflammation, University of Glasgow, Gartnavel General Hospital, Glasgow, G12 OYN, Scotland, UK.
| | | | - Rekha Chaudhuri
- Respiratory Medicine, Institute of Infection, Immunity and Inflammation, University of Glasgow, Gartnavel General Hospital, Glasgow, G12 OYN, Scotland, UK
| | - Mark Spears
- Respiratory Medicine, Forth Valley Royal Hospital, Larbert, UK
| | - Kazuhiro Ito
- Airway Disease Section, Imperial College, London, UK
| | - Charles McSharry
- Respiratory Medicine, Institute of Infection, Immunity and Inflammation, University of Glasgow, Gartnavel General Hospital, Glasgow, G12 OYN, Scotland, UK
| |
Collapse
|
34
|
Affiliation(s)
- J. E. Gern
- Pediatrics and Medicine; University of Wisconsin-Madison; Madison WI USA
| |
Collapse
|
35
|
Manthei DM, Schwantes EA, Mathur SK, Guadarrama AG, Kelly EA, Gern JE, Jarjour NN, Denlinger LC. Nasal lavage VEGF and TNF-α levels during a natural cold predict asthma exacerbations. Clin Exp Allergy 2014; 44:1484-93. [PMID: 25109477 PMCID: PMC4247169 DOI: 10.1111/cea.12387] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 06/09/2014] [Accepted: 06/20/2014] [Indexed: 01/09/2023]
Abstract
BACKGROUND Asthma exacerbations contribute to significant morbidity, mortality and healthcare utilization. Furthermore, viral infections are associated with asthma exacerbations by mechanisms that are not fully understood. OBJECTIVE The aim of this analysis was to determine whether cytokine patterns in patients with colds could identify risks for subsequent asthma exacerbations. METHODS We analysed cytokine levels in nasal lavage fluid (NLF) in 59 subjects (46 with asthma) with acute upper respiratory symptoms and after symptomatic resolution. Analyte choice was based on potential relevance to asthma exacerbations: antiviral (IFN-α, IFN-β, IFN-γ, IFN-λ1, IP-10, TRAIL), cell recruiting (G-CSF, IL-1β, IL-8, MCP-1, MCP-3, TNF-α), polarizing (CXCL13, IL-10, IL-13, IL-17, TSLP), and injury remodelling (fibronectin, IL-33, MMP-9, VEGF). RESULTS The overall cytokine response induced during viral infections was not different between asthmatic and non-asthmatic individuals for a wide array of cytokines. However, mean levels of VEGF, TNF-α and IL-1β were 1.7-, 5.1- and 4.7-fold higher in samples from asthma subjects who exacerbated in the first 3 weeks of the cold compared with those who did not exacerbate (P = 0.006, 0.01, 0.048, respectively). Using receiver operating characteristic curve-defined thresholds, high VEGF and TNF-α levels predicted a shorter time-to-exacerbation after NLF sampling (25% exacerbation rate: 3 vs. 45 days, and 3 vs. 26 days; P = 0.03, 0.04, respectively). CONCLUSION AND CLINICAL RELEVANCE Although they produce similar cytokine responses to viral infection as non-asthmatics, asthmatics with higher levels of VEGF and TNF-α in NLF obtained during acute cold phases predicted subsequent asthma exacerbations in this cohort of patients with mild-to-moderate disease. In the future, stratifying the risk of an asthma exacerbation by cytokine profile may aid the targeting of personalized treatment and intervention strategies.
Collapse
Affiliation(s)
- D M Manthei
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
van den Kieboom CH, Ahout IML, Zomer A, Brand KH, de Groot R, Ferwerda G, de Jonge MI. Nasopharyngeal gene expression, a novel approach to study the course of respiratory syncytial virus infection. Eur Respir J 2014; 45:718-25. [PMID: 25261323 DOI: 10.1183/09031936.00085614] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Respiratory syncytial virus (RSV) causes mild infections in the vast majority of children. However, in some cases, it causes severe disease, such as bronchiolitis and pneumonia. Development of severe RSV infection is determined by the host response. Therefore, the main aim of this study was to identify biomarkers associated with severe RSV infection. To identify biomarkers, nasopharyngeal gene expression was profiled by microarray studies, resulting in the selection of five genes: ubiquitin D, tetraspanin 8, mucin 13, β-microseminoprotein and chemokine ligand 7. These genes were validated by real-time quantitative PCR in an independent validation cohort, which confirmed significant differences in gene expression between mildly and severely infected and between recovery and acute patients. Nasopharyngeal aspirate samples are regularly taken when a viral respiratory tract infection is suspected. In this article, we describe a method to discriminate between mild and severe RSV infection based on differential host gene expression. The combination of pathogen detection and host gene expression analysis in nasopharyngeal aspirates will significantly improve the diagnosis and prognosis of respiratory tract infections.
Collapse
Affiliation(s)
- Corné H van den Kieboom
- Laboratory of Pediatric Infectious Diseases, Dept of Pediatrics, Radboud University Medical Center, Nijmegen, The Netherlands Laboratory of Medical Immunology, Dept of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Inge M L Ahout
- Laboratory of Pediatric Infectious Diseases, Dept of Pediatrics, Radboud University Medical Center, Nijmegen, The Netherlands Laboratory of Medical Immunology, Dept of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Aldert Zomer
- Laboratory of Pediatric Infectious Diseases, Dept of Pediatrics, Radboud University Medical Center, Nijmegen, The Netherlands Laboratory of Medical Immunology, Dept of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kim H Brand
- Laboratory of Pediatric Infectious Diseases, Dept of Pediatrics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ronald de Groot
- Laboratory of Pediatric Infectious Diseases, Dept of Pediatrics, Radboud University Medical Center, Nijmegen, The Netherlands Laboratory of Medical Immunology, Dept of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gerben Ferwerda
- Laboratory of Pediatric Infectious Diseases, Dept of Pediatrics, Radboud University Medical Center, Nijmegen, The Netherlands Laboratory of Medical Immunology, Dept of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marien I de Jonge
- Laboratory of Pediatric Infectious Diseases, Dept of Pediatrics, Radboud University Medical Center, Nijmegen, The Netherlands Laboratory of Medical Immunology, Dept of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
37
|
Wen D, Du X, Nie SP, Dong JZ, Ma CS. Association between RANTES gene polymorphisms and asthma: a meta-analysis. PLoS One 2014; 9:e90460. [PMID: 24963658 PMCID: PMC4070887 DOI: 10.1371/journal.pone.0090460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 01/30/2014] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND A few recent studies have suggested that regulated on activation, normal T cell expressed and secreted (RANTES) polymorphisms (-403 G/A, -28C/G) are associated with asthma. However, there still existed studies which did not confirm these correlations. OBJECTIVE The objective of this study was to evaluate the relationship of RANTES and asthma using a meta-analysis. METHODS Pubmed, Embase, and Cochrane library databases were systemically searched. Data were extracted by two independent reviewers and pooled odds ratio (OR) with 95% confidence interval (CI) were calculated. RESULTS Eighteen studies were enrolled, including a total of 2558 cases and 2630 controls of -403 G/A, as well as 3311 cases and 4031 controls of -28C/G in this meta-analysis. The overall ORs and 95% CIs of -403 G/A were 1.19, 1.06-1.33 (P<0.001) and 1.25, 1.03-1.51 (P = 0.933) in dominant and recessive models, respectively. The overall ORs and 95% CIs of -28G were 1.23, 1.09-1.39 (P = 0.221) and 1.76, 1.32-2.34 (P = 0.356) in dominant and recessive models, respectively. No publication bias among studies was showed. CONCLUSIONS This meta-analysis showed that RANTES -403 G/A polymorphism was a risk factor for asthma, while -28C/G polymorphism were not associated with asthma.
Collapse
Affiliation(s)
- Dan Wen
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xin Du
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Shao-Ping Nie
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jian-Zeng Dong
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chang-Sheng Ma
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
38
|
The Editor takes a closer look at some of this month's articles. Clin Exp Allergy 2014. [DOI: 10.1111/cea.12342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
39
|
Ramirez IA, Caverly LL, Kalikin LM, Goldsmith AM, Lewis TC, Burke DT, LiPuma JJ, Sajjan US, Hershenson MB. Differential responses to rhinovirus- and influenza-associated pulmonary exacerbations in patients with cystic fibrosis. Ann Am Thorac Soc 2014; 11:554-61. [PMID: 24641803 PMCID: PMC4225796 DOI: 10.1513/annalsats.201310-346oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 01/26/2014] [Indexed: 12/25/2022] Open
Abstract
RATIONALE The mechanism by which viruses cause exacerbations of chronic airway disease and the capacity of patients with cystic fibrosis (CF) to respond to viral infection are not precisely known. OBJECTIVES To determine the antiviral response to infection in patients with CF. METHODS Sputum was collected from patients with CF with respiratory exacerbation. Viruses were detected in multiplex polymerase chain reaction (PCR)-based assays. Gene expression of 84 antiviral response genes was measured, using a focused quantitative PCR gene array. MEASUREMENTS AND MAIN RESULTS We examined 36 samples from 23 patients with respiratory exacerbation. Fourteen samples tested virus-positive and 22 virus-negative. When we compared exacerbations associated with rhinovirus (RV, n = 9) and influenza (n = 5) with virus-negative specimens, we found distinct patterns of antiviral gene expression. RV was associated with greater than twofold induction of five genes, including those encoding the monocyte-attracting chemokines CXCL10, CXCL11, and CXCL9. Influenza was associated with overexpression of 20 genes, including those encoding the cytokines tumor necrosis factor and IL-12; the kinases MEK, TBK-1, and STAT-1; the apoptosis proteins caspase-8 and caspase-10; the influenza double-stranded RNA receptor RIG-I and its downstream effector MAVS; and pyrin, an IFN-stimulated protein involved in influenza resistance. CONCLUSIONS We conclude that virus-induced exacerbations of CF are associated with immune responses tailored to specific infections. Influenza induced a more potent response consisting of inflammation, whereas RV infection had a pronounced effect on chemokine expression. As far as we are aware, this study is the first to compare specific responses to different viruses in live patients with chronic airway disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Marc B. Hershenson
- Department of Pediatrics and Communicable Diseases
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
40
|
Schuler BA, Schreiber MT, Li L, Mokry M, Kingdon ML, Raugi DN, Smith C, Hameister C, Racaniello VR, Hall DJ. Major and minor group rhinoviruses elicit differential signaling and cytokine responses as a function of receptor-mediated signal transduction. PLoS One 2014; 9:e93897. [PMID: 24736642 PMCID: PMC3988043 DOI: 10.1371/journal.pone.0093897] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 03/08/2014] [Indexed: 11/18/2022] Open
Abstract
Major- and minor-group human rhinoviruses (HRV) enter their host by binding to the cell surface molecules ICAM-1 and LDL-R, respectively, which are present on both macrophages and epithelial cells. Although epithelial cells are the primary site of productive HRV infection, previous studies have implicated macrophages in establishing the cytokine dysregulation that occurs during rhinovirus-induced asthma exacerbations. Analysis of the transcriptome of primary human macrophages exposed to major- and minor-group HRV demonstrated differential gene expression. Alterations in gene expression were traced to differential mitochondrial activity and signaling pathway activation between two rhinovirus serotypes, HRV16 (major-group) and HRV1A (minor-group), upon initial HRV binding. Variances in phosphorylation of kinases (p38, JNK, ERK5) and transcription factors (ATF-2, CREB, CEBP-alpha) were observed between the major- and minor-group HRV treatments. Differential activation of signaling pathways led to changes in the production of the asthma-relevant cytokines CCL20, CCL2, and IL-10. This is the first report of genetically similar viruses eliciting dissimilar cytokine release, transcription factor phosphorylation, and MAPK activation from macrophages, suggesting that receptor use is a mechanism for establishing the inflammatory microenvironment in the human airway upon exposure to rhinovirus.
Collapse
Affiliation(s)
- Bryce A. Schuler
- Department of Chemistry, Lawrence University, Appleton, Wisconsin, United States of America
| | - Michael T. Schreiber
- Department of Chemistry, Lawrence University, Appleton, Wisconsin, United States of America
- Department of Microbiology & Immunology, Columbia University College of Physicians and Surgeons, New York, New York, United States of America
| | - LuYuan Li
- Department of Chemistry, Lawrence University, Appleton, Wisconsin, United States of America
| | - Michal Mokry
- Division of Pediatrics, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Megan L. Kingdon
- Department of Chemistry, Lawrence University, Appleton, Wisconsin, United States of America
| | - Dana N. Raugi
- Department of Chemistry, Lawrence University, Appleton, Wisconsin, United States of America
| | - Cosonya Smith
- Department of Chemistry, Lawrence University, Appleton, Wisconsin, United States of America
| | - Chelsea Hameister
- Department of Chemistry, Lawrence University, Appleton, Wisconsin, United States of America
| | - Vincent R. Racaniello
- Department of Microbiology & Immunology, Columbia University College of Physicians and Surgeons, New York, New York, United States of America
| | - David J. Hall
- Department of Chemistry, Lawrence University, Appleton, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
41
|
Nasal epithelial repair and remodeling in physical injury, infection, and inflammatory diseases. Curr Opin Otolaryngol Head Neck Surg 2013; 21:263-70. [PMID: 23449287 DOI: 10.1097/moo.0b013e32835f80a0] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW To summarize the current knowledge of cellular and molecular mechanisms of nasal epithelial repair and remodeling during physical and pathophysiological conditions. RECENT FINDINGS Nasal epithelial repair and remodeling is a highly organized and well coordinated process, involving inflammation, proliferation, differentiation, matrix deposition, and remodeling, and is regulated by a wide variety of growth factors and cytokines. From the in-vivo and in-vitro studies conducted in both human and animal models, undifferentiated basal cells (progenitors) are able to migrate from adjacent epithelium, spread over the denuded basement membrane, and proliferate in injured regions (self-renewal) in necessary (homeostasis) or excessive (hyperplasia) degree. Progenitor cells reorient to an apical-basal polarity, and progressively differentiate into ciliated and nonciliated columnar cells and goblet cells, reconstituting a functional respiratory epithelium after several weeks. This recovery process can be observed during various types and severity of injury, and also in common nasal diseases, including acute viral, allergic, and nonallergic rhinitis, as well as chronic rhinosinusitis with and without nasal polyps. SUMMARY Although nearly 10 000 articles about nasal epithelium have been published in the last decade, the mechanisms underlying the nasal epithelial repair are still understood at only a superficial descriptive level. In order to advance rhinology to the next level of a comprehensive knowledge of the orchestrated genetic and molecular processes acting during epithelial repair, combined clinical and experimental studies using sophisticated investigational plans to elucidate the functions of both the protein-coding and regulatory portions of the human genome are required.
Collapse
|
42
|
Kimura H, Yoshizumi M, Ishii H, Oishi K, Ryo A. Cytokine production and signaling pathways in respiratory virus infection. Front Microbiol 2013; 4:276. [PMID: 24062733 PMCID: PMC3774987 DOI: 10.3389/fmicb.2013.00276] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 08/26/2013] [Indexed: 12/13/2022] Open
Abstract
It has been confirmed that respiratory virus infections can induce abberant cytokine production in the host. These cytokines may be associated with both elimination of the virus and complications in the host, such as virus-induced asthma. Representative host defense mechanisms against pathogens, including bacteria and viruses, are mediated by the innate immune system. Cells of the innate immune system express essential molecules, namely pattern recognition receptors (PRRs), such as Toll-like receptors, nucleotide-binding oligomerization domain-like receptors, and retinoic acid-inducible gene-I-like receptors. These PRRs can recognize components of pathogens such as bacterial lipopolysaccharide, viral antigens, and their genomes (DNA and RNA). Furthermore, PRRs activate various signaling pathways resulting in cytokine production against pathogen infection. However, the exact mechanisms remain unknown. In this review, we mainly focus on the representative mechanisms of cytokine production through PRRs and signaling pathways due to virus infections, including respiratory virus infections. In addition, we describe the relationships between respiratory infections and virus-induced asthma.
Collapse
Affiliation(s)
- Hirokazu Kimura
- Infectious Disease Surveillance Center, National Institute of Infectious Diseases Tokyo, Japan ; Gunma Prefectural Institute of Public Health and Environmental Sciences Gunma, Japan ; Department of Molecular Biodefence Research, Graduate School of Medicine, Yokohama City University Kanagawa, Japan
| | | | | | | | | |
Collapse
|
43
|
Lewis TC, Henderson TA, Carpenter AR, Ramirez IA, McHenry CL, Goldsmith AM, Ren X, Mentz GB, Mukherjee B, Robins TG, Joiner TA, Mohammad LS, Nguyen ER, Burns MA, Burke DT, Hershenson MB. Nasal cytokine responses to natural colds in asthmatic children. Clin Exp Allergy 2013. [PMID: 23181789 PMCID: PMC4219353 DOI: 10.1111/cea.12005] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background The mechanisms by which viruses induce asthma exacerbations are not well understood. Objective We characterized fluctuations in nasal aspirate cytokines during naturally occurring respiratory viral infections in children with asthma. Methods Sixteen children underwent home collections of nasal aspirates when they were without cold symptoms and again during self‐reported respiratory illnesses. The presence of viral infection was ascertained by multiplex PCR. Cytokines were measured using multiplex immune assay. mRNA expression for selected markers of viral infection was measured using RT‐PCR. A cumulative respiratory symptom score was calculated for each day of measurement. Generalized estimated equations were used to evaluate associations between viral infection and marker elevation, and between marker elevation and symptom score. Results The 16 patients completed a total of 37 weeks of assessment (15 ‘well’ weeks; 22 self‐assessed ‘sick’ weeks). Viral infections were detected in 3 of the ‘well’ weeks and 17 of the ‘sick’ weeks (10 rhinovirus, three coronavirus, two influenza A, two influenza B, two respiratory syncytial virus, one parainfluenza). Compared to virus‐negative well weeks, nasal aspirate IFN‐γ, CXCL8/IL‐8, CXCL10/IP‐10, CCL5/RANTES, CCL11/eotaxin‐1, CCL2/MCP‐1, CCL4/MIP‐1β, CCL7/MCP‐3, and CCL20/MIP3α protein levels increased during virus‐positive sick weeks. Only a subset of cytokines (IFN‐γ, CXCL8, CCL2, CCL4, CCL5, and CCL20) correlated with self‐reported respiratory tract symptoms. While many aspirates were dilute and showed no mRNA signal, viral infection significantly increased the number of samples that were positive for IFN‐λ1, IFN‐λ2/3, TLR3, RIG‐I, and IRF7 mRNA. Conclusions and clinical relevance We conclude that in children with asthma, naturally occurring viral infections apparently induce a robust innate immune response including expression of specific chemokines, IFNs, and IFN‐responsive genes.
Collapse
Affiliation(s)
- T C Lewis
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Schneider D, Hong JY, Bowman ER, Chung Y, Nagarkar DR, McHenry CL, Goldsmith AM, Bentley JK, Lewis TC, Hershenson MB. Macrophage/epithelial cell CCL2 contributes to rhinovirus-induced hyperresponsiveness and inflammation in a mouse model of allergic airways disease. Am J Physiol Lung Cell Mol Physiol 2012. [PMID: 23204071 DOI: 10.1152/ajplung.00182.2012] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Human rhinovirus (HRV) infections lead to exacerbations of lower airways disease in asthmatic patients but not in healthy individuals. However, underlying mechanisms remain to be completely elucidated. We hypothesized that the Th2-driven allergic environment enhances HRV-induced CC chemokine production, leading to asthma exacerbations. Ovalbumin (OVA)-sensitized and -challenged mice inoculated with HRV showed significant increases in the expression of lung CC chemokine ligand (CCL)-2/monocyte chemotactic protein (MCP)-1, CCL4/macrophage inflammatory protein (MIP)-1β, CCL7/MCP-3, CCL19/MIP-3β, and CCL20/MIP3α compared with mice treated with OVA alone. Inhibition of CCL2 with neutralizing antibody significantly attenuated HRV-induced airways inflammation and hyperresponsiveness in OVA-treated mice. Immunohistochemical stains showed colocalization of CCL2 with HRV in epithelial cells and CD68-positive macrophages, and flow cytometry showed increased CCL2(+), CD11b(+) cells in the lungs of OVA-treated, HRV-infected mice. Compared with lung macrophages from naïve mice, macrophages from OVA-exposed mice expressed significantly more CCL2 in response to HRV infection ex vivo. Pretreatment of mouse lung macrophages and BEAS-2B human bronchial epithelial cells with interleukin (IL)-4 and IL-13 increased HRV-induced CCL2 expression, and mouse lung macrophages from IL-4 receptor knockout mice showed reduced CCL2 expression in response to HRV, suggesting that exposure to these Th2 cytokines plays a role in the altered HRV response. Finally, bronchoalveolar macrophages from children with asthma elaborated more CCL2 upon ex vivo exposure to HRV than cells from nonasthmatic patients. We conclude that CCL2 production by epithelial cells and macrophages contributes to HRV-induced airway hyperresponsiveness and inflammation in a mouse model of allergic airways disease and may play a role in HRV-induced asthma exacerbations.
Collapse
Affiliation(s)
- Dina Schneider
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|