1
|
Easley KF, Edenfield RC, Lott MEJ, Reed RC, Das Sarma J, Mehta AJ, Staitieh BS, Lipp EK, Cho IK, Johnson SK, Jones CA, Bebin-Blackwell AG, Levy JM, Tompkins SM, Easley CA, Koval M. Chronic alcohol use primes bronchial cells for altered inflammatory response and barrier dysfunction during SARS-CoV-2 infection. Am J Physiol Lung Cell Mol Physiol 2023; 325:L647-L661. [PMID: 37786945 PMCID: PMC11498272 DOI: 10.1152/ajplung.00381.2022] [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: 11/18/2022] [Revised: 07/26/2023] [Accepted: 09/15/2023] [Indexed: 10/04/2023] Open
Abstract
Alcohol use disorder (AUD) is a significant public health concern and people with AUD are more likely to develop severe acute respiratory distress syndrome (ARDS) in response to respiratory infections. To examine whether AUD was a risk factor for more severe outcome in response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, we examined early responses to infection using cultured differentiated bronchial epithelial cells derived from brushings obtained from people with AUD or without AUD. RNA-seq analysis of uninfected cells determined that AUD cells were enriched for expression of epidermal genes as compared with non-AUD cells. Bronchial epithelial cells from patients with AUD showed a significant decrease in barrier function 72 h postinfection, as determined by transepithelial electrical resistance. In contrast, barrier function of non-AUD cells was enhanced 72 h after SARS-CoV-2 infection. AUD cells showed claudin-7 that did not colocalize with zonula occludens-1 (ZO-1), indicative of disorganized tight junctions. However, both AUD and non-AUD cells showed decreased β-catenin expression following SARS-CoV-2 infection. To determine the impact of AUD on the inflammatory response to SARS-CoV-2 infection, cytokine secretion was measured by multiplex analysis. SARS-CoV-2-infected AUD bronchial cells had enhanced secretion of multiple proinflammatory cytokines including TNFα, IL-1β, and IFNγ as opposed to non-AUD cells. In contrast, secretion of the barrier-protective cytokines epidermal growth factor (EGF) and granulocyte macrophage-colony stimulating factor (GM-CSF) was enhanced for non-AUD bronchial cells. Taken together, these data support the hypothesis that AUD is a risk factor for COVID-19, where alcohol primes airway epithelial cells for increased inflammation and increased barrier dysfunction and increased inflammation in response to infection by SARS-CoV-2.NEW & NOTEWORTHY Alcohol use disorder (AUD) is a significant risk factor for severe acute respiratory distress syndrome. We found that AUD causes a phenotypic shift in gene expression in human bronchial epithelial cells, enhancing expression of epidermal genes. AUD cells infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) had higher levels of proinflammatory cytokine secretion and barrier dysfunction not present in infected non-AUD cells, consistent with increased early COVID-19 severity due to AUD.
Collapse
Affiliation(s)
- Kristen F Easley
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - R Clayton Edenfield
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia, United States
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, United States
| | - Megan E J Lott
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia, United States
| | - Ryan C Reed
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Ashish J Mehta
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
- Atlanta Veterans Affairs Health Care System, Decatur, Georgia, United States
| | - Bashar S Staitieh
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Erin K Lipp
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia, United States
| | - In Ki Cho
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia, United States
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, United States
| | - Scott K Johnson
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, United States
| | - Cheryl A Jones
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, United States
| | | | - Joshua M Levy
- Department of Otolaryngology, Emory University School of Medicine, Atlanta, Georgia, United States
- National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, United States
| | - S Mark Tompkins
- Center for Vaccines and Immunology, University of Georgia, Athens, Georgia, United States
| | - Charles A Easley
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia, United States
- Regenerative Bioscience Center, University of Georgia, Athens, Georgia, United States
| | - Michael Koval
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University School of Medicine, Atlanta, Georgia, United States
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, United States
| |
Collapse
|
2
|
Muralidharan A, Bauer CD, Katafiasz DM, Strah HM, Siddique A, Reid SP, Bailey KL, Wyatt TA. Synergistic Detrimental Effects of Cigarette Smoke, Alcohol, and SARS-CoV-2 in COPD Bronchial Epithelial Cells. Pathogens 2023; 12:498. [PMID: 36986420 PMCID: PMC10056639 DOI: 10.3390/pathogens12030498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/09/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Lung conditions such as COPD, as well as risk factors such as alcohol misuse and cigarette smoking, can exacerbate COVID-19 disease severity. Synergistically, these risk factors can have a significant impact on immunity against pathogens. Here, we studied the effect of a short exposure to alcohol and/or cigarette smoke extract (CSE) in vitro on acute SARS-CoV-2 infection of ciliated human bronchial epithelial cells (HBECs) collected from healthy and COPD donors. We observed an increase in viral titer in CSE- or alcohol-treated COPD HBECs compared to untreated COPD HBECs. Furthermore, we treated healthy HBECs accompanied by enhanced lactate dehydrogenase activity, indicating exacerbated injury. Finally, IL-8 secretion was elevated due to the synergistic damage mediated by alcohol, CSE, and SARS-CoV-2 in COPD HBECs. Together, our data suggest that, with pre-existing COPD, short exposure to alcohol or CSE is sufficient to exacerbate SARS-CoV-2 infection and associated injury, impairing lung defences.
Collapse
Affiliation(s)
- Abenaya Muralidharan
- Department of Pathology and Microbiology, College of Medicine, The University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Christopher D. Bauer
- Pulmonary, Critical Care, and Sleep Medicine Division, Department of Internal Medicine, College of Medicine, the University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Dawn M. Katafiasz
- Pulmonary, Critical Care, and Sleep Medicine Division, Department of Internal Medicine, College of Medicine, the University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Heather M. Strah
- Pulmonary, Critical Care, and Sleep Medicine Division, Department of Internal Medicine, College of Medicine, the University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Aleem Siddique
- Department of Surgery, College of Medicine, the University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - St Patrick Reid
- Department of Pathology and Microbiology, College of Medicine, The University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kristina L. Bailey
- Pulmonary, Critical Care, and Sleep Medicine Division, Department of Internal Medicine, College of Medicine, the University of Nebraska Medical Center, Omaha, NE 68198, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Todd A. Wyatt
- Pulmonary, Critical Care, and Sleep Medicine Division, Department of Internal Medicine, College of Medicine, the University of Nebraska Medical Center, Omaha, NE 68198, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
- Department of Environmental, Agricultural & Occupational Health, College of Public Health, the University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
3
|
Bailey KL, Sayles H, Campbell J, Khalid N, Anglim M, Ponce J, Wyatt TA, McClay JC, Burnham EL, Anzalone A, Hanson C. COVID-19 patients with documented alcohol use disorder or alcohol-related complications are more likely to be hospitalized and have higher all-cause mortality. Alcohol Clin Exp Res 2022; 46:1023-1035. [PMID: 35429004 PMCID: PMC9111368 DOI: 10.1111/acer.14838] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/25/2022] [Accepted: 04/07/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Coronavirus Disease 2019 (COVID-19) has affected every country globally, with hundreds of millions of people infected with the SARS-CoV-2 virus and over 6 million deaths to date. It is unknown how alcohol use disorder (AUD) affects the severity and mortality of COVID-19. AUD is known to increase the severity and mortality of bacterial pneumonia and the risk of developing acute respiratory distress syndrome. Our objective is to determine whether individuals with AUD have increased severity and mortality from COVID-19. METHODS We utilized a retrospective cohort study of inpatients and outpatients from 44 centers participating in the National COVID Cohort Collaborative. All were adult COVID-19 patients with and without documented AUDs. RESULTS We identified 25,583 COVID-19 patients with an AUD and 1,309,445 without. In unadjusted comparisons, those with AUD had higher odds of hospitalization (odds ratio [OR] 2.00, 95% confidence interval [CI] 1.94 to 2.06, p < 0.001). After adjustment for age, sex, race/ethnicity, smoking, body mass index, and comorbidities, individuals with an AUD still had higher odds of requiring hospitalization (adjusted OR [aOR] 1.51, CI 1.46 to 1.56, p < 0.001). In unadjusted comparisons, individuals with AUD had higher odds of all-cause mortality (OR 2.18, CI 2.05 to 2.31, p < 0.001). After adjustment as above, individuals with an AUD still had higher odds of all-cause mortality (aOR 1.55, CI 1.46 to 1.65, p < 0.001). CONCLUSION This work suggests that AUD can increase the severity and mortality of COVID-19 infection. This reinforces the need for clinicians to obtain an accurate alcohol history from patients hospitalized with COVID-19. For this study, our results are limited by an inability to quantify the daily drinking habits of the participants. Studies are needed to determine the mechanisms by which AUD increases the severity and mortality of COVID-19.
Collapse
Affiliation(s)
- Kristina L. Bailey
- Division of PulmonaryCritical Care, and Sleep MedicineDepartment of Internal MedicineUniversity of Nebraska Medical Center, College of MedicineOmahaNebraskaUSA
- VA Nebraska‐Western Iowa Health SystemsOmahaNebraskaUSA
| | - Harlan Sayles
- Department of BiostatisticsUniversity of Nebraska Medical Center, College of Public HealthOmahaNebraskaUSA
| | - James Campbell
- Division of PulmonaryCritical Care, and Sleep MedicineDepartment of Internal MedicineUniversity of Nebraska Medical Center, College of MedicineOmahaNebraskaUSA
| | - Neha Khalid
- Division of PulmonaryCritical Care, and Sleep MedicineDepartment of Internal MedicineUniversity of Nebraska Medical Center, College of MedicineOmahaNebraskaUSA
| | - Madyson Anglim
- Division of PulmonaryCritical Care, and Sleep MedicineDepartment of Internal MedicineUniversity of Nebraska Medical Center, College of MedicineOmahaNebraskaUSA
| | - Jana Ponce
- University of Nebraska Medical Center, College of Allied HealthOmahaNebraskaUSA
| | - Todd A. Wyatt
- Division of PulmonaryCritical Care, and Sleep MedicineDepartment of Internal MedicineUniversity of Nebraska Medical Center, College of MedicineOmahaNebraskaUSA
- VA Nebraska‐Western Iowa Health SystemsOmahaNebraskaUSA
- Department of Environmental, Agricultural and Occupational HealthUniversity of Nebraska Medical Center, College of Public HealthOmahaNebraskaUSA
| | - James C. McClay
- Department of Emergency MedicineUniversity of Nebraska Medical Center, College of MedicineOmahaNebraskaUSA
| | - Ellen L. Burnham
- Division of Pulmonary Sciences and Critical Care MedicineDepartment of MedicineUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Alfred Anzalone
- Department of Neurological SciencesUniversity of Nebraska Medical Center, College of MedicineOmahaNebraskaUSA
- Great Plains IDeA‐CTROmahaNebraskaUSA
| | - Corrine Hanson
- University of Nebraska Medical Center, College of Allied HealthOmahaNebraskaUSA
| |
Collapse
|
4
|
Wladis EJ, Adam AP. Immune signaling in rosacea. Ocul Surf 2021; 22:224-229. [PMID: 34481075 DOI: 10.1016/j.jtos.2021.08.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 08/24/2021] [Accepted: 08/30/2021] [Indexed: 11/29/2022]
Abstract
Rosacea is a common chronic skin disease affecting mostly people aged 40 and above, with currently no cure. When it affects the eyelids and periocular skin, it leads to dry eye and potentially corneal damage. Research performed over the last decade shed light into the potential mechanisms leading to skin hypersensitivity and provided promising avenues for development of novel, rational therapeutics aimed at reducing the skin inflammatory state. In this review, we discuss the current knowledge on the mechanisms of rosacea in general and of periocular skin-affecting disease in particular, identify key questions that remain to be answered in future research, and offer a disease model that can explain the key characteristics of this disease, with particular emphasis on a potential positive feedback loop that could explain both the acute and chronic features of rosacea.
Collapse
Affiliation(s)
- Edward J Wladis
- Lions Eye Institute, Department of Ophthalmology, Albany Medical College, 1220 New Scotland Rd, Suite 302, Slingerlands, NY, 12159, United States.
| | - Alejandro P Adam
- Department of Molecular and Cellular Physiology and Department of Ophthalmology, Albany Medical College, United States
| |
Collapse
|
5
|
Cao QT, Aguiar JA, Tremblay BJM, Abbas N, Tiessen N, Revill S, Makhdami N, Ayoub A, Cox G, Ask K, Doxey AC, Hirota JA. ABCF1 Regulates dsDNA-induced Immune Responses in Human Airway Epithelial Cells. Front Cell Infect Microbiol 2020; 10:487. [PMID: 33042865 PMCID: PMC7525020 DOI: 10.3389/fcimb.2020.00487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 08/05/2020] [Indexed: 11/24/2022] Open
Abstract
Background: The airway epithelium represents a critical component of the human lung that helps orchestrate defenses against respiratory tract viral infections, which are responsible for more than 2.5 million deaths/year globally. Innate immune activities of the airway epithelium rely on Toll-like receptors (TLRs), nucleotide binding and leucine-rich-repeat pyrin domain containing (NLRP) receptors, and cytosolic nucleic acid sensors. ATP Binding Cassette (ABC) transporters are ubiquitous across all three domains of life—Archaea, Bacteria, and Eukarya—and expressed in the human airway epithelium. ABCF1, a unique ABC family member that lacks a transmembrane domain, has been defined as a cytosolic nucleic acid sensor that regulates CXCL10, interferon-β expression, and downstream type I interferon responses. We tested the hypothesis that ABCF1 functions as a dsDNA nucleic acid sensor in human airway epithelial cells important in regulating antiviral responses. Methods: Expression and localization experiments were performed using in situ hybridization and immunohistochemistry in human lung tissue, while confirmatory transcript and protein expression was performed in human airway epithelial cells. Functional experiments were performed with siRNA methods in a human airway epithelial cell line. Complementary transcriptomic analyses were performed to explore the contributions of ABCF1 to gene expression patterns. Results: Using archived human lung and human airway epithelial cells, we confirm expression of ABCF1 gene and protein expression in these tissue samples, with a role for mediating CXCL10 production in response to dsDNA viral mimic challenge. Although, ABCF1 knockdown was associated with an attenuation of select genes involved in the antiviral responses, Gene Ontology analyses revealed a greater interaction of ABCF1 with TLR signaling suggesting a multifactorial role for ABCF1 in innate immunity in human airway epithelial cells. Conclusion: ABCF1 is a candidate cytosolic nucleic acid sensor and modulator of TLR signaling that is expressed at gene and protein levels in human airway epithelial cells. The precise level where ABCF1 protein functions to modulate immune responses to pathogens remains to be determined but is anticipated to involve IRF-3 and CXCL10 production.
Collapse
Affiliation(s)
- Quynh T Cao
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | | | | | - Nadin Abbas
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | - Nicholas Tiessen
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | - Spencer Revill
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | - Nima Makhdami
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | - Anmar Ayoub
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | - Gerard Cox
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | - Kjetil Ask
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada.,McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Andrew C Doxey
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada.,Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Jeremy A Hirota
- Division of Respirology, Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada.,Department of Biology, University of Waterloo, Waterloo, ON, Canada.,McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada.,Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
6
|
Bailey KL, Smith H, Mathai SK, Huber J, Yacoub M, Yang IV, Wyatt TA, Kechris K, Burnham EL. Alcohol Use Disorders Are Associated With a Unique Impact on Airway Epithelial Cell Gene Expression. Alcohol Clin Exp Res 2020; 44:1571-1584. [PMID: 32524622 PMCID: PMC7484391 DOI: 10.1111/acer.14395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 05/28/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Alcohol use disorders (AUDs) and cigarette smoking both increase risk for the development of community-acquired pneumonia (CAP), likely through adverse effects on proximal airway mucociliary clearance and pathogen recognition. Smoking-related alterations on airway gene expression are well described, but little is known about the impact of AUDs. We measured gene expression in human airway epithelial cells (AECs), hypothesizing that AUDs would be associated with novel differences in gene expression that could alter risk for CAP. METHODS Bronchoscopy with airway brushings was performed in participants with AUDs and controls to obtain AECs. An AUD Identification Test was used to define AUD. RNA was extracted from AECs, and mRNA expression data were collected on an Agilent micro-array. Differential expression analyses were performed on the filtered and normalized data with correction for multiple testing. Enrichment analyses were performed using clusterProfiler. RESULTS Expression data from 19 control and 18 AUD participants were evaluated. After adjustment for smoking, AUDs were associated with significant differential expression of 520 AEC genes, including genes for ribosomal proteins and genes involved in protein folding. Enrichment analyses indicated significant differential expression of 24 pathways in AUDs, including those implicated in protein targeting to membrane and viral gene expression. Smoking-associated AEC gene expression differences mirrored previous reports, but differed from those associated with AUDs. CONCLUSIONS AUDs have a distinct impact on AEC gene expression that may influence proximal airway function independent of smoking. Alcohol-associated alterations may influence risk for CAP through modifying key mechanisms important in protecting proximal airway integrity.
Collapse
Affiliation(s)
- Kristina L. Bailey
- University of Nebraska Medical Center, Department of Internal Medicine. Division of Pulmonary, Critical Care, Sleep and Allergy
- VA Nebraska-Western Iowa Health Care System
| | - Harry Smith
- University of Colorado Anschutz Medical Campus, Department of Biostatistics and Informatics, Colorado School of Public Health
| | - Susan K. Mathai
- Baylor University Medical Center, Center for Advanced Heart & Lung Disease
| | - Jonathan Huber
- University of Colorado Anschutz Medical Campus, Department of Medicine, Division of Allergy & Clinical Immunology
| | - Mark Yacoub
- University of Colorado Anschutz Medical Campus, Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine
| | - Ivana V. Yang
- University of Colorado Anschutz Medical Campus, Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine
- University of Colorado Anschutz Medical Campus, Department of Medicine, Division of Biomedical Informatics and Personalized Medicine
| | - Todd A. Wyatt
- VA Nebraska-Western Iowa Health Care System
- University of Nebraska Medical Center, Department of Environmental, Agricultural, & Occupational Health
| | - Katerina Kechris
- University of Colorado Anschutz Medical Campus, Department of Biostatistics and Informatics, Colorado School of Public Health
- University of Colorado Anschutz Medical Campus, Department of Medicine, Division of Biomedical Informatics and Personalized Medicine
| | - Ellen L. Burnham
- University of Colorado Anschutz Medical Campus, Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine
| |
Collapse
|
7
|
Gaydos J, McNally A, Burnham EL. The impact of alcohol use disorders on pulmonary immune cell inflammatory responses to Streptococcus pneumoniae. Alcohol 2019; 80:119-130. [PMID: 30195043 DOI: 10.1016/j.alcohol.2018.08.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/29/2018] [Accepted: 08/31/2018] [Indexed: 12/11/2022]
Abstract
Community-acquired pneumonia due to Streptococcus pneumoniae occurs commonly in alcohol use disorders (AUDs). Pneumonia in the AUD patient is associated with poorer outcomes, and specific therapies to mitigate disease severity in these patients do not exist. Numerous investigations have attributed increased severity of pneumonia in AUDs to aberrant function of the alveolar macrophage (AM), a lung immune cell critical in host defense initiation. No studies have examined the response of human AMs to S. pneumoniae in AUDs. We hypothesized that the inflammatory mediators released by AMs after S. pneumoniae stimulation would differ quantitatively in individuals with AUDs compared to non-AUD participants. We further postulated that AM inflammatory mediators would be diminished after exposure to the antioxidant, N-acetylcysteine (NAC). For comparison, responses of peripheral blood mononuclear cells (PBMCs) to pneumococcal protein were also examined. Otherwise healthy participants with AUDs and smoking-matched controls underwent bronchoalveolar lavage and peripheral blood sampling to obtain AMs and PBMCs, respectively. Freshly collected cells were cultured with increasing doses of heat-killed S. pneumoniae protein, with and without exposure to N-acetylcysteine. Cell culture supernatants were collected, and inflammatory mediators were measured, including interferon (IFN)-γ, interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α. IFN-γ and IL-6 were significantly higher in unstimulated AM cell culture supernatants from subjects with AUDs. After stimulation with pneumococcal protein, a dose-response and time-dependent increase in pro-inflammatory cytokine production by both AMs and PBMCs was also observed; differences were not observed between AUD and non-AUD subjects. Addition of NAC to pneumococcal-stimulated AMs and PBMCs was generally associated with diminished cytokine production, with the exception of IL-1β that was elevated in AM culture supernatants from subjects with AUDs. Our observations suggest that AUDs contribute to basal alterations in AM pro-inflammatory cytokine elaboration, but did not support consistent differences in pneumococcal-stimulated AM or PBMC inflammatory mediator secretion that were referable to AUDs.
Collapse
|
8
|
Bailey KL, Wyatt TA, Katafiasz DM, Taylor KW, Heires AJ, Sisson JH, Romberger DJ, Burnham EL. Alcohol and cannabis use alter pulmonary innate immunity. Alcohol 2019; 80:131-138. [PMID: 30419300 DOI: 10.1016/j.alcohol.2018.11.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 10/19/2018] [Accepted: 11/03/2018] [Indexed: 11/30/2022]
Abstract
PURPOSE Cannabis use is increasing due to recent legislative changes. In addition, cannabis is often used in conjunction with alcohol. The airway epithelium is the first line of defense against infectious microbes. Toll-like receptors (TLR) recognize airborne microbes and initiate the inflammatory cytokine response. The mechanism by which cannabis use in conjunction with alcohol affects pulmonary innate immunity mediated by TLRs is unknown. METHODS Samples and data from an existing cohort of individuals with alcohol use disorders (AUDs), along with samples from additional participants with cannabis use alone and with AUD were utilized. Subjects were categorized into the following groups: no alcohol use disorder (AUD) or cannabis use (control) (n = 46), AUD only (n = 29), cannabis use-only (n = 39), and AUD and cannabis use (n = 29). The participants underwent bronchoscopy with bronchoalveolar lavage (BAL) and airway epithelial brushings. We measured IL-6, IL-8, TNF⍺, and IL-10 levels in BAL fluid, and performed real-time PCR for TLR1-9 on the airway epithelial brushings. RESULTS We found significant increases in TLR2 with AUD alone, cannabis use alone, and cannabis use with AUD, compared to control. TLR5 was increased in cannabis users compared to control, TLR6 was increased in cannabis users and cannabis users with AUD compared to control, TLR7 was increased in cannabis users compared to control, and TLR9 was increased in cannabis users compared to control. In terms of cytokine production, IL-6 was increased in cannabis users compared to control. IL-8 and IL-10 were increased in AUD only. CONCLUSIONS AUD and cannabis use have complex effects on pulmonary innate immunity that promote airway inflammation.
Collapse
Affiliation(s)
- Kristina L Bailey
- University of Nebraska Medical Center, Internal Medicine/Pulmonary, Critical Care Sleep and Allergy, Omaha, NE 68198, United States; Veterans Affairs, Nebraska Western-Iowa Healthcare System, Omaha, NE 68105, United States.
| | - Todd A Wyatt
- University of Nebraska Medical Center, Internal Medicine/Pulmonary, Critical Care Sleep and Allergy, Omaha, NE 68198, United States; Veterans Affairs, Nebraska Western-Iowa Healthcare System, Omaha, NE 68105, United States; Department of Environmental, Agricultural and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Dawn M Katafiasz
- University of Nebraska Medical Center, Internal Medicine/Pulmonary, Critical Care Sleep and Allergy, Omaha, NE 68198, United States
| | - Keenan W Taylor
- University of Nebraska Medical Center, Internal Medicine/Pulmonary, Critical Care Sleep and Allergy, Omaha, NE 68198, United States
| | - Art J Heires
- University of Nebraska Medical Center, Internal Medicine/Pulmonary, Critical Care Sleep and Allergy, Omaha, NE 68198, United States
| | - Joseph H Sisson
- University of Nebraska Medical Center, Internal Medicine/Pulmonary, Critical Care Sleep and Allergy, Omaha, NE 68198, United States
| | - Debra J Romberger
- University of Nebraska Medical Center, Internal Medicine/Pulmonary, Critical Care Sleep and Allergy, Omaha, NE 68198, United States; Veterans Affairs, Nebraska Western-Iowa Healthcare System, Omaha, NE 68105, United States
| | - Ellen L Burnham
- University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, United States
| |
Collapse
|
9
|
Warren KJ, Poole JA, Sweeter JM, DeVasure JM, Wyatt TA. An association between MMP-9 and impaired T cell migration in ethanol-fed BALB/c mice infected with respiratory syncytial virus-2A. Alcohol 2019; 80:25-32. [PMID: 30291948 DOI: 10.1016/j.alcohol.2018.09.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 09/14/2018] [Accepted: 09/26/2018] [Indexed: 12/20/2022]
Abstract
Matrix metalloproteinases are important for proper airway matrix structure and wound healing. These enzymes are also implicated in many airway diseases. Previously, chronic ethanol consumption was shown to prolong inflammation and delay viral clearance in respiratory syncytial virus (RSV)-infected mice. We hypothesize that alcohol alters anti-viral immunity by disrupting immune cell chemotaxis in the lung. BALB/c mice were randomly selected to consume 18% alcohol ad libitum for 8 weeks prior to infection with RSV-2A. Bronchoalveolar lavage (BAL) cell populations were measured by flow cytometry, and chemokines were detected by Western blot or ELISA. MMP-9 levels were determined by polymerase chain reaction (PCR) in mouse lungs and in BAL fluid by ELISA. T cells were acquired from the spleens of water-fed, non-infected control mice (CTRL); alcohol-fed, non-infected (ETOH); water-fed, RSV-infected (RSV); or ethanol-fed, RSV-infected (ETOH-RSV) 4 days after RSV infection. T cells were placed in a transmigration system where chemokines had been treated with and without activated MMP-9. Lymphocyte recruitment was significantly reduced in the BAL 4 days after RSV infection in ETOH-RSV mice, whereas chemokine levels were the highest in this group at all experimental time points examined in comparison to RSV (p < 0.05). MMP-9 mRNA and protein were detected at high levels in ETOH-RSV mice compared to RSV. Using ex vivo transmigration to CCL2 and CXCL10, T cell migration was not impaired between any of the treatment groups, yet when CCL2 and CXCL10 were treated with activated MMP-9, significantly fewer T cells migrated across collagen-coated 5-μm membranes (p < 0.05). Immune cell recruitment is necessary for viral clearance. We show that immune cells are decreased in the lungs of ETOH-RSV mice. In contrast to decreased cell recruitment, key inflammatory chemokines were elevated in the lungs of ETOH-RSV mice. These proteins may be prematurely degraded by MMP-9 in the lung, leading to defective immunity and reduced viral clearance.
Collapse
Affiliation(s)
- Kristi J Warren
- University of Nebraska Medical Center, Pulmonary, Critical Care, Sleep & Allergy, 985910 Nebraska Medical Center, Omaha, NE, 68198-5910, United States.
| | - Jill A Poole
- University of Nebraska Medical Center, Pulmonary, Critical Care, Sleep & Allergy, 985910 Nebraska Medical Center, Omaha, NE, 68198-5910, United States
| | - Jenea M Sweeter
- University of Nebraska Medical Center, Pulmonary, Critical Care, Sleep & Allergy, 985910 Nebraska Medical Center, Omaha, NE, 68198-5910, United States.
| | - Jane M DeVasure
- University of Nebraska Medical Center, Pulmonary, Critical Care, Sleep & Allergy, 985910 Nebraska Medical Center, Omaha, NE, 68198-5910, United States.
| | - Todd A Wyatt
- University of Nebraska Medical Center, Pulmonary, Critical Care, Sleep & Allergy, 985910 Nebraska Medical Center, Omaha, NE, 68198-5910, United States; Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, United States; University of Nebraska Medical Center, Department of Environmental, Agricultural, & Occupational Health, Omaha, NE, 68198, United States.
| |
Collapse
|
10
|
Simou E, Leonardi-Bee J, Britton J. The Effect of Alcohol Consumption on the Risk of ARDS: A Systematic Review and Meta-Analysis. Chest 2018; 154:58-68. [PMID: 29288645 PMCID: PMC6045784 DOI: 10.1016/j.chest.2017.11.041] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/13/2017] [Accepted: 11/29/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND To conduct a systematic review and meta-analysis evaluating the association between alcohol consumption and the risk of ARDS in adults. METHODS Medline, EMBASE and Web of Science were searched to identify observational studies evaluating the association between prior alcohol intake and the occurrence of ARDS among adults, published between 1985 and 2015 and with no language restriction. Reference lists were also screened. Demographic baseline data were extracted independently by two reviewers and random-effects meta-analyses were used to estimate pooled effect sizes with 95% confidence intervals. Subgroup analyses were used to explore heterogeneity. RESULTS Seventeen observational studies (177,674 people) met the inclusion criteria. Meta-analysis of 13 studies showed that any measure of high relative to low alcohol consumption was associated with a significantly increased risk of ARDS (OR, 1.89; 95% CI, 1.45-2.48; I2 = 48%; 13 studies); no evidence of publication bias was seen (P = .150). Sensitivity analyses indicated that this association was attributable primarily to an effect of a history of alcohol abuse (OR, 1.90; 95% CI, 1.40-2.60; 10 studies). Also, subgroup analyses identified that heterogeneity was explained by predisposing condition (trauma, sepsis/septic shock, pneumonia; P = .003). CONCLUSIONS Chronic high alcohol consumption significantly increases the risk of ARDS. This finding suggests that patients admitted to hospital should be screened for chronic alcohol use.
Collapse
Affiliation(s)
- Evangelia Simou
- UK Centre for Tobacco and Alcohol Studies, Division of Epidemiology and Public Health, University of Nottingham, Nottingham, UK.
| | - Jo Leonardi-Bee
- UK Centre for Tobacco and Alcohol Studies, Division of Epidemiology and Public Health, University of Nottingham, Nottingham, UK
| | - John Britton
- UK Centre for Tobacco and Alcohol Studies, Division of Epidemiology and Public Health, University of Nottingham, Nottingham, UK
| |
Collapse
|
11
|
Sapkota M, Burnham EL, DeVasure JM, Sweeter JM, Hunter C, Duryee MJ, Klassen LW, Kharbanda KK, Sisson JH, Thiele GM, Wyatt TA. Malondialdehyde-Acetaldehyde (MAA) Protein Adducts Are Found Exclusively in the Lungs of Smokers with Alcohol Use Disorders and Are Associated with Systemic Anti-MAA Antibodies. Alcohol Clin Exp Res 2017; 41:2093-2099. [PMID: 28941289 PMCID: PMC5711572 DOI: 10.1111/acer.13509] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 09/18/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Malondialdehyde (MDA) and acetaldehyde (AA) exist following ethanol metabolism and tobacco pyrolysis. As such, lungs of individuals with alcohol use disorders (AUDs) are a target for the effects of combined alcohol and cigarette smoke metabolites. MDA and AA form a stable protein adduct, malondialdehyde-acetaldehyde (MAA) adduct, known to be immunogenic, profibrotic, and proinflammatory. MAA adduct is the dominant epitope in anti-MAA antibody formation. We hypothesized that MAA-adducted protein forms in lungs of those who both abuse alcohol and smoke cigarettes, and that this would be associated with systemically elevated anti-MAA antibodies. METHODS Four groups were established: AUD subjects who smoked cigarettes (+AUD/+smoke), smokers without AUD (-AUD/+smoke), AUD without smoke (+AUD/-smoke), and non-AUD/nonsmokers (-AUD/-smoke). RESULTS We observed a significant increase in MAA adducts in lung cells of +AUD/+smoke versus -AUD/-smoke. No significant increase in MAA adducts was observed in -AUD/+smoke or in +AUD/-smoke compared to -AUD/-smoke. Serum from +AUD/+smoke had significantly increased levels of circulating anti-MAA IgA antibodies. After 1 week of alcohol that MAA-adducted protein is formed in the lungs of those who smoke cigarettes and abuse alcohol, leading to a subsequent increase in serum IgA antibodies. CONCLUSIONS MAA-adducted proteins could play a role in pneumonia and other diseases of the lung in the setting of AUD and smoking.
Collapse
Affiliation(s)
- Muna Sapkota
- Department of Environmental, Agricultural and Occupational Health, University of Nebraska Medical Center, Omaha, NE
| | - Ellen L. Burnham
- University of Colorado School of Medicine, Anschutz Medical Campus, Denver, CO
| | - Jane M. DeVasure
- Department of Internal Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Nebraska Medical Center
| | - Jenea M. Sweeter
- Department of Internal Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Nebraska Medical Center
| | - C.D. Hunter
- Department of Internal Medicine, Division of Rheumatology and Immunology, University of Nebraska Medical Center
| | - Michael J. Duryee
- Department of Internal Medicine, Division of Rheumatology and Immunology, University of Nebraska Medical Center
| | - Lynell W. Klassen
- Department of Internal Medicine, Division of Rheumatology and Immunology, University of Nebraska Medical Center
| | | | - Joseph H. Sisson
- Department of Internal Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Nebraska Medical Center
| | - Geoffrey M. Thiele
- Department of Internal Medicine, Division of Rheumatology and Immunology, University of Nebraska Medical Center
| | - Todd A. Wyatt
- Department of Environmental, Agricultural and Occupational Health, University of Nebraska Medical Center, Omaha, NE
- Department of Internal Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Nebraska Medical Center
- VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| |
Collapse
|
12
|
Jose SS, Bendickova K, Kepak T, Krenova Z, Fric J. Chronic Inflammation in Immune Aging: Role of Pattern Recognition Receptor Crosstalk with the Telomere Complex? Front Immunol 2017; 8:1078. [PMID: 28928745 PMCID: PMC5591428 DOI: 10.3389/fimmu.2017.01078] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 08/18/2017] [Indexed: 12/22/2022] Open
Abstract
Age-related decline in immunity is characterized by stem cell exhaustion, telomere shortening, and disruption of cell-to-cell communication, leading to increased patient risk of disease. Recent data have demonstrated that chronic inflammation exerts a strong influence on immune aging and is closely correlated with telomere length in a range of major pathologies. The current review discusses the impact of inflammation on immune aging, the likely molecular mediators of this process, and the various disease states that have been linked with immunosenescence. Emerging findings implicate NF-κB, the major driver of inflammatory signaling, in several processes that regulate telomere maintenance and/or telomerase activity. While prolonged triggering of pattern recognition receptors is now known to promote immunosenescence, it remains unclear how this process is linked with the telomere complex or telomerase activity. Indeed, enzymatic control of telomere length has been studied for many decades, but alternative roles of telomerase and potential influences on inflammatory responses are only now beginning to emerge. Crosstalk between these pathways may prove to be a key molecular mechanism of immunosenescence. Understanding how components of immune aging interact and modify host protection against pathogens and tumors will be essential for the design of new vaccines and therapies for a wide range of clinical scenarios.
Collapse
Affiliation(s)
- Shyam Sushama Jose
- Cellular and Molecular Immunoregulation Group (CMI), Center for Translational Medicine (CTM), International Clinical Research Center (ICRC), St. Anne’s University Hospital Brno, Brno, Czechia
- Department of Biology, Faculty of Medicine, Masaryk University, Czechia
| | - Kamila Bendickova
- Cellular and Molecular Immunoregulation Group (CMI), Center for Translational Medicine (CTM), International Clinical Research Center (ICRC), St. Anne’s University Hospital Brno, Brno, Czechia
| | - Tomas Kepak
- Pediatric Oncology Translational Research (POTR), International Clinical Research Center (ICRC), St. Anne’s University Hospital Brno, Brno, Czechia
- Pediatric Hematology and Oncology, University Hospital Brno, Brno, Czechia
| | - Zdenka Krenova
- Pediatric Oncology Translational Research (POTR), International Clinical Research Center (ICRC), St. Anne’s University Hospital Brno, Brno, Czechia
- Pediatric Hematology and Oncology, University Hospital Brno, Brno, Czechia
| | - Jan Fric
- Cellular and Molecular Immunoregulation Group (CMI), Center for Translational Medicine (CTM), International Clinical Research Center (ICRC), St. Anne’s University Hospital Brno, Brno, Czechia
| |
Collapse
|
13
|
Burnham EL, McNally A, Gaydos J, Brown LAS. The Relationship Between Airway Antioxidant Levels, Alcohol Use Disorders, and Cigarette Smoking. Alcohol Clin Exp Res 2016; 40:2147-2160. [PMID: 27627733 DOI: 10.1111/acer.13201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 07/27/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Alcohol use disorders (AUDs) and cigarette smoking are associated with pulmonary oxidative stress, likely related to antioxidant depletion. Pulmonary oxidative stress may adversely affect innate immunity, leading to increased pneumonia susceptibility and severity, including development of the acute respiratory distress syndrome. In people with AUDs, most of whom smoke, antioxidant therapy can potentially restore immune cell function and attenuate pneumonia development. Challenges to human investigations of antioxidant therapies include an inability to identify pulmonary oxidative stress noninvasively and the optimal route to deliver pulmonary antioxidants. We sought to determine whether bronchoalveolar lavage (BAL) measures of thiol antioxidants from a 50-ml upper airway aliquot approximated those in the alveolar space and to determine whether AUDs and/or smoking affected these relationships. METHODS Healthy human subjects with and without AUDs, including smokers and nonsmokers, underwent BAL. Samples obtained after the first 50-ml normal saline aliquot were analyzed as representing bronchial airways; subsequent 50-ml aliquots were analyzed as representative of the alveolar space. Reduced and oxidized (GSSG) glutathione, cysteine (Cys), and its oxidized species, cystine, along with mixed disulfides (MDs) were quantified using high-performance liquid chromatography. The percent of total thiols present in their oxidized forms, and thiol redox potentials, were calculated. RESULTS Positive correlations between upper and lower BAL fluid thiol species were observed that were most robust for GSSG (ρ = 0.85), Cys (ρ = 0.83), and MDs (ρ = 0.69), but poor for thiol redox potential measures. In contrast to nonsmokers (either with or without AUDs), in subjects with AUDs who smoked, upper BAL fluid %GSSG, Cys, and MD measures were relatively increased compared to lower. CONCLUSIONS A small volume BAL procedure may be suitable to assess intrapulmonary oxidative stress related to thiol depletion. Factors including AUDs and smoking may disproportionately increase upper airways oxidative stress that could be relevant for therapeutic interventions.
Collapse
Affiliation(s)
- Ellen L Burnham
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.
| | - Alicia McNally
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Jeanette Gaydos
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Lou Ann S Brown
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|