1
|
Vaillancourt M, Aguilar D, Fernandes SE, Jorth PA. A chronic Pseudomonas aeruginosa mouse lung infection modeling the pathophysiology and inflammation of human cystic fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617039. [PMID: 39416002 PMCID: PMC11482824 DOI: 10.1101/2024.10.07.617039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Investigation of chronic cystic fibrosis (CF) lung infections has been limited by a lack of murine models that reproduce obstructive lung pathology, chronicity of bacterial infections, and complex inflammation in human CF lung pathology. Three different approaches have been used separately to address these limitations, including using transgenic Scnn1b-Tg mice overexpressing a lung epithelial sodium channel to mimic the mucus-rich and hyperinflammatory CF lung environment, using synthetic CF sputum medium (SCFM) in an acute infection to induce bacterial phenotypes consistent with human CF, or using agar beads to promote chronic infections. Here, we combine these three models to establish a chronic Pseudomonas aeruginosa lung infection model using SCFM agar beads and Scnn1b-Tg mice (SCFM-Tg-mice) to recapitulate nutrients, mucus, and inflammation characteristic of the human CF lung environment. Like people with CF, SCFM-Tg-mice failed to clear bacterial infections. Lung function measurements showed that infected SCFM-Tg-mice had decreased inspiratory capacity and compliance, elevated airway resistance, and significantly reduced FVC and FEV0.1. Using spectral flow cytometry and multiplex cytokine arrays we show that, like people with CF, SCFM-Tg-mice developed inflammation characterized by eosinophil infiltration and Th2 lymphocytic cytokine responses. Chronically infected SCFM-Tg-mice developed an exacerbated mix of innate and Th1, Th2, and Th17-mediated inflammation, causing higher lung cellular damage, and elevated numbers of unusual Siglec F+ neutrophils. Thus, SCFM-Tg-mice represents a powerful tool to investigate bacterial pathogenesis and potential treatments for chronic CF lung infections and reveal a potential role for Siglec F+ neutrophils in CF inflammation.
Collapse
Affiliation(s)
- Mylene Vaillancourt
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Diane Aguilar
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sheryl E. Fernandes
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Peter A. Jorth
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
2
|
Han S, Kim B, Hyeon DY, Jeong D, Ryu J, Nam JS, Choi YH, Kim BR, Park SC, Chung YW, Shin SJ, Lee JY, Kim JK, Park J, Lee SW, Kim TB, Cheon JH, Cho HJ, Kim CH, Yoon JH, Hwang D, Ryu JH. Distinctive CD39 +CD9 + lung interstitial macrophages suppress IL-23/Th17-mediated neutrophilic asthma by inhibiting NETosis. Nat Commun 2024; 15:8628. [PMID: 39366998 PMCID: PMC11452667 DOI: 10.1038/s41467-024-53038-2] [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: 06/04/2023] [Accepted: 09/30/2024] [Indexed: 10/06/2024] Open
Abstract
The IL-23-Th17 axis is responsible for neutrophilic inflammation in various inflammatory diseases. Here, we discover a potential pathway to inhibit neutrophilic asthma. In our neutrophil-dominant asthma (NDA) model, single-cell RNA-seq analysis identifies a subpopulation of CD39+CD9+ interstitial macrophages (IMs) suppressed by IL-23 in NDA conditions but increased by an IL-23 inhibitor αIL-23p19. Adoptively transferred CD39+CD9+ IMs suppress neutrophil extracellular trap formation (NETosis), a representative phenotype of NDA, and also Th17 cell activation and neutrophilic inflammation. CD39+CD9+ IMs first attach to neutrophils in a CD9-dependent manner, and then remove ATP near neutrophils that contribute to NETosis in a CD39-dependent manner. Transcriptomic data from asthmatic patients finally show decreased CD39+CD9+ IMs in severe asthma than mild/moderate asthma. Our results suggest that CD39+CD9+ IMs function as a potent negative regulator of neutrophilic inflammation by suppressing NETosis in the IL-23-Th17 axis and can thus serve as a potential therapeutic target for IL-23-Th17-mediated neutrophilic asthma.
Collapse
Affiliation(s)
- Seunghan Han
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Bomin Kim
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Do Young Hyeon
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Daeun Jeong
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Jaechan Ryu
- Institut Pasteur, Microenvironment and Immunity Unit, Paris, France
| | - Jae-Sung Nam
- Department of Otorhinolaryngology and Airway Mucus Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Yoon Ha Choi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea
| | - Bo-Ram Kim
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Korea
| | - Sang Chul Park
- Department of Otorhinolaryngology-Head and Neck Surgery, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Youn Wook Chung
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Jae Shin
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - June-Yong Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Kyoung Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Korea
| | - Jihye Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Sei Won Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Tae-Bum Kim
- Department of Allergy and Clinical Immunology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Hee Cheon
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Hyung-Ju Cho
- Department of Otorhinolaryngology and Airway Mucus Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Chang-Hoon Kim
- Department of Otorhinolaryngology and Airway Mucus Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Joo-Heon Yoon
- Department of Otorhinolaryngology and Airway Mucus Institute, Yonsei University College of Medicine, Seoul, Korea.
| | - Daehee Hwang
- School of Biological Sciences, Seoul National University, Seoul, Korea.
| | - Ji-Hwan Ryu
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Korea.
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
3
|
Skibba ME, Brasier AR. NF-κB/RelA signaling in secretoglobin progenitors mediates plasticity and MMP-induced barrier disruption in house dust mite-induced allergic asthma. Am J Physiol Lung Cell Mol Physiol 2024; 327:L86-L101. [PMID: 38713619 PMCID: PMC11380976 DOI: 10.1152/ajplung.00066.2024] [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: 02/20/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/09/2024] Open
Abstract
The mechanisms how aeroallergens induce sensitization are incompletely understood. The house dust mite (HDM) Dermatophagoides pteronyssius (Der p) is a ubiquitous aeroallergen that represents a major cause of allergic rhinitis and asthma. Herein, we tested whether HDM-induced aeroallergen exposure sensitivity is caused by the innate-immune response in small airway epithelial cells. HDM exposure is a rapid activator of NF-κB/RelA in the Secretoglobin (Scgb1a1+) lineage associated with upregulation of NF-κB/RelA-dependent markers of epithelial plasticity. To determine the effect of epithelial NF-κB signaling, NF-κB was depleted in a tamoxifen (TMX)-inducible Scgb1a1-CreERTM mouse within a CL57B/L6 background. Corn oil or TMX-treated/RelA-depleted [RelA knockdown (KD)] mice were repetitively exposed to airway HDM challenges to induce airway hyperresponsiveness (AHR). Strikingly, we observed that HDM induces hallmarks of epithelial plasticity through upregulation of the mesenchymal core factors SNAI1 and ZEB1 and production of metalloproteinase (MMP)9 that are RelA-dependent. Downstream, HDM-induced mucous metaplasia, Th2 polarization, allergen sensitivity, and airway hyperreactivity were all reduced in the RelA-depleted mice. Mechanistically, HDM-induced functional and structural barrier disruption was dependent on RelA signaling and associated with active MMP secretion into the bronchoalveolar lavage fluid. To establish the role of MMP2/9 in barrier disruption, we observe that a small-molecule MMP inhibitor (SB-3CT) blocked HDM-induced barrier disruption and activation of plasticity in naïve wild-type (WT) mice. Loss of functional barrier was associated with MMP disruption of zona occludens (ZO)-1 containing adherens junctions. Overall, this data indicates that host innate signaling in the Scgb1a1+ progenitors is directly linked to epithelial plasticity, MMP9 secretion, and enhanced barrier permeability that allows allergen penetration, sensitization producing allergic asthma (AA) in vivo. We propose that maintenance of epithelial integrity may reduce allergic sensitization and AA.NEW & NOTEWORTHY Allergic asthma from house dust mite (HDM) allergy causes substantial morbidity. This study examines the dynamic changes in small airway epithelial cells in a mouse model of HDM exposure. Our findings indicate that NF-κB/RelA signaling mediates matrix metalloproteinase production, disrupting the epithelial barrier resulting in allergic sensitization. Our findings bring new insight into mechanisms for epithelial cell-state change in the allergen response, creating a potential therapeutic pathway for maintaining barrier function in asthma.
Collapse
Affiliation(s)
- Melissa E Skibba
- School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States
| | - Allan R Brasier
- School of Medicine and Public Health, University of Wisconsin Madison, Madison, Wisconsin, United States
- Institute for Clinical and Translational Research, Madison, Wisconsin, United States
| |
Collapse
|
4
|
Zimmermann R, Roeder F, Ruppert C, Smith BJ, Knudsen L. Low-volume ventilation of preinjured lungs degrades lung function via stress concentration and progressive alveolar collapse. Am J Physiol Lung Cell Mol Physiol 2024; 327:L19-L39. [PMID: 38712429 DOI: 10.1152/ajplung.00323.2023] [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: 10/18/2023] [Revised: 03/27/2024] [Accepted: 04/22/2024] [Indexed: 05/08/2024] Open
Abstract
Mechanical ventilation can cause ventilation-induced lung injury (VILI). The concept of stress concentrations suggests that surfactant dysfunction-induced microatelectases might impose injurious stresses on adjacent, open alveoli and function as germinal centers for injury propagation. The aim of the present study was to quantify the histopathological pattern of VILI progression and to test the hypothesis that injury progresses at the interface between microatelectases and ventilated lung parenchyma during low-positive end-expiratory pressure (PEEP) ventilation. Bleomycin was used to induce lung injury with microatelectases in rats. Lungs were then mechanically ventilated for up to 6 h at PEEP = 1 cmH2O and compared with bleomycin-treated group ventilated protectively with PEEP = 5 cmH2O to minimize microatelectases. Lung mechanics were measured during ventilation. Afterward, lungs were fixed at end-inspiration or end-expiration for design-based stereology. Before VILI, bleomycin challenge reduced the number of open alveoli [N(alvair,par)] by 29%. No differences between end-inspiration and end-expiration were observed. Collapsed alveoli clustered in areas with a radius of up to 56 µm. After PEEP = 5 cmH2O ventilation for 6 h, N(alvair,par) remained stable while PEEP = 1 cmH2O ventilation led to an additional loss of aerated alveoli by 26%, mainly due to collapse, with a small fraction partly edema filled. Alveolar loss strongly correlated to worsening of tissue elastance, quasistatic compliance, and inspiratory capacity. The radius of areas of collapsed alveoli increased to 94 µm, suggesting growth of the microatelectases. These data provide evidence that alveoli become unstable in neighborhood of microatelectases, which most likely occurs due to stress concentration-induced local vascular leak and surfactant dysfunction.NEW & NOTEWORTHY Low-volume mechanical ventilation in the presence of high surface tension-induced microatelectases leads to the degradation of lung mechanical function via the progressive loss of alveoli. Microatelectases grow at the interfaces of collapsed and open alveoli. Here, stress concentrations might cause injury and alveolar instability. Accumulation of small amounts of alveolar edema can be found in a fraction of partly collapsed alveoli but, in this model, alveolar flooding is not a major driver for degradation of lung mechanics.
Collapse
Affiliation(s)
- Richard Zimmermann
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Franziska Roeder
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Clemens Ruppert
- Department of Internal Medicine, Justus-Liebig-University Giessen, Giessen, Germany
- University of Giessen and Marburg Lung Center (UGMLC), member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Bradford J Smith
- Department of Bioengineering, College of Engineering, Design & Computing, University of Colorado Denver | Anschutz Medical Campus, Aurora, Colorado, United States
- Section of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, United States
| | - Lars Knudsen
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| |
Collapse
|
5
|
Seasock MJ, Shafiquzzaman M, Ruiz-Echartea ME, Kanchi RS, Tran BT, Simon LM, Meyer MD, Erice PA, Lotlikar SL, Wenlock SC, Ochsner SA, Enright A, Carisey AF, Romero F, Rosas IO, King KY, McKenna NJ, Coarfa C, Rodriguez A. Let-7 restrains an oncogenic epigenetic circuit in AT2 cells to prevent ectopic formation of fibrogenic transitional cell intermediates and pulmonary fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595205. [PMID: 38826218 PMCID: PMC11142151 DOI: 10.1101/2024.05.22.595205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Analysis of lung alveolar type 2 (AT2) progenitor stem cells has highlighted fundamental mechanisms that direct their differentiation into alveolar type 1 cells (AT1s) in lung repair and disease. However, microRNA (miRNA) mediated post-transcriptional mechanisms which govern this nexus remain understudied. We show here that the let-7 miRNA family serves a homeostatic role in governance of AT2 quiescence, specifically by preventing the uncontrolled accumulation of AT2 transitional cells and by promoting AT1 differentiation to safeguard the lung from spontaneous alveolar destruction and fibrosis. Using mice and organoid models with genetic ablation of let-7a1/let-7f1/let-7d cluster (let-7afd) in AT2 cells, we demonstrate prevents AT1 differentiation and results in aberrant accumulation of AT2 transitional cells in progressive pulmonary fibrosis. Integration of enhanced AGO2 UV-crosslinking and immunoprecipitation sequencing (AGO2-eCLIP) with RNA-sequencing from AT2 cells uncovered the induction of direct targets of let-7 in an oncogene feed-forward regulatory network including BACH1/EZH2 which drives an aberrant fibrotic cascade. Additional analyses by CUT&RUN-sequencing revealed loss of let-7afd hampers AT1 differentiation by eliciting aberrant histone EZH2 methylation which prevents the exit of AT2 transitional cells into terminal AT1s. This study identifies let-7 as a key gatekeeper of post-transcriptional and epigenetic chromatin signals to prevent AT2-driven pulmonary fibrosis.
Collapse
Affiliation(s)
- Matthew J. Seasock
- Immunology & Microbiology Graduate Program, Baylor College of Medicine, Houston, TX, 77030
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of Medicine Houston TX, 77030
| | - Md Shafiquzzaman
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of Medicine Houston TX, 77030
| | - Maria E. Ruiz-Echartea
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030
| | - Rupa S. Kanchi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine Houston, TX, 77030
| | - Brandon T. Tran
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, 77030
- Department of Pediatrics, Division of Infectious Diseases, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, 77030
| | - Lukas M. Simon
- Therapeutic Innovation Center, Baylor College of Medicine, Houston, TX, 77030
| | | | - Phillip A. Erice
- Immunology & Microbiology Graduate Program, Baylor College of Medicine, Houston, TX, 77030
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of Medicine Houston TX, 77030
| | - Shivani L. Lotlikar
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of Medicine Houston TX, 77030
| | | | - Scott A. Ochsner
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030
| | - Anton Enright
- Department of Pathology, University of Cambridge, Cambridge, CB2 1TN, UK
| | - Alex F. Carisey
- William T. Shearer Center for Immunobiology, Texas Children’s Hospital, Houston, TX, 77030
- Current Address: Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Freddy Romero
- Department of Medicine, Section of Pulmonary and Critical Care, Baylor College of Medicine. Houston, TX, 77030
- Current Address: Vertex Pharmaceuticals, 3215 Merryfield Row, San Diego, CA, 92121
| | - Ivan O. Rosas
- Department of Medicine, Section of Pulmonary and Critical Care, Baylor College of Medicine. Houston, TX, 77030
| | - Katherine Y. King
- Department of Pediatrics, Division of Infectious Diseases, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, 77030
| | - Neil J. McKenna
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine Houston, TX, 77030
| | - Antony Rodriguez
- Department of Medicine, Immunology & Allergy Rheumatology, Baylor College of Medicine Houston TX, 77030
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine Houston, TX, 77030
- Center for Translational Research on Inflammatory Diseases, Michael E. Debakey, Baylor College of Medicine, Houston, TX, 77030
| |
Collapse
|
6
|
Chou H, Godbeer L, Ball ML. Establishing breath as a biomarker platform-take home messages from the Breath Biopsy Conference 2023. J Breath Res 2024; 18:030401. [PMID: 38631337 DOI: 10.1088/1752-7163/ad3fdf] [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: 12/08/2023] [Accepted: 04/17/2024] [Indexed: 04/19/2024]
Abstract
The annual Breath Biopsy Conference hosted by Owlstone Medical gathers together the leading experts, early career researchers, and physicians working with breath as a biomarker platform for clinical purposes. The current topics in breath research are discussed and presented, and an overarching topical theme is identified and discussed as part of an expert panel to close the conference. The profiling of normal breath composition and the establishment of standards for analyzing breath compared to background signal were two important topics that were major focuses of this conference, as well as important innovative progress that has been made since last year, including the development of a non-invasive breath test for lung cancer and liver disease. This meeting report offers an overview of the key take-home messages from the various presentations, posters, and discussions from the conference.
Collapse
Affiliation(s)
- Hsuan Chou
- Owlstone Medical Ltd, 183 Cambridge Science Park, Milton Road, Cambridge, CB4 0GJ, United Kingdom
| | - Lucy Godbeer
- Owlstone Medical Ltd, 183 Cambridge Science Park, Milton Road, Cambridge, CB4 0GJ, United Kingdom
| | - Madeleine L Ball
- Owlstone Medical Ltd, 183 Cambridge Science Park, Milton Road, Cambridge, CB4 0GJ, United Kingdom
| |
Collapse
|
7
|
Achanta S, Gentile MA, Albert CJ, Schulte KA, Pantazides BG, Crow BS, Quiñones-González J, Perez JW, Ford DA, Patel RP, Blake TA, Gunn MD, Jordt SE. Recapitulation of human pathophysiology and identification of forensic biomarkers in a translational model of chlorine inhalation injury. Am J Physiol Lung Cell Mol Physiol 2024; 326:L482-L495. [PMID: 38318664 PMCID: PMC11281795 DOI: 10.1152/ajplung.00162.2023] [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: 05/18/2023] [Revised: 01/16/2024] [Accepted: 01/27/2024] [Indexed: 02/07/2024] Open
Abstract
Chlorine gas (Cl2) has been repeatedly used as a chemical weapon, first in World War I and most recently in Syria. Life-threatening Cl2 exposures frequently occur in domestic and occupational environments, and in transportation accidents. Modeling the human etiology of Cl2-induced acute lung injury (ALI), forensic biomarkers, and targeted countermeasures development have been hampered by inadequate large animal models. The objective of this study was to develop a translational model of Cl2-induced ALI in swine to understand toxico-pathophysiology and evaluate whether it is suitable for screening potential medical countermeasures and to identify biomarkers useful for forensic analysis. Specific pathogen-free Yorkshire swine (30-40 kg) of either sex were exposed to Cl2 (≤240 ppm for 1 h) or filtered air under anesthesia and controlled mechanical ventilation. Exposure to Cl2 resulted in severe hypoxia and hypoxemia, increased airway resistance and peak inspiratory pressure, and decreased dynamic lung compliance. Cl2 exposure resulted in increased total leucocyte and neutrophil counts in bronchoalveolar lavage fluid, vascular leakage, and pulmonary edema compared with the air-exposed group. The model recapitulated all three key histopathological features of human ALI, such as neutrophilic alveolitis, deposition of hyaline membranes, and formation of microthrombi. Free and lipid-bound 2-chlorofatty acids and chlorotyrosine-modified proteins (3-chloro-l-tyrosine and 3,5-dichloro-l-tyrosine) were detected in plasma and lung tissue after Cl2 exposure. In this study, we developed a translational swine model that recapitulates key features of human Cl2 inhalation injury and is suitable for testing medical countermeasures, and validated chlorinated fatty acids and protein adducts as biomarkers of Cl2 inhalation.NEW & NOTEWORTHY We established a swine model of chlorine gas-induced acute lung injury that exhibits several features of human acute lung injury and is suitable for screening potential medical countermeasures. We validated chlorinated fatty acids and protein adducts in plasma and lung samples as forensic biomarkers of chlorine inhalation.
Collapse
Affiliation(s)
- Satyanarayana Achanta
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Michael A Gentile
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Carolyn J Albert
- Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, Missouri, United States
| | - Kevin A Schulte
- Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, Missouri, United States
| | - Brooke G Pantazides
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia, United States
| | - Brian S Crow
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia, United States
| | - Jennifer Quiñones-González
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia, United States
| | - Jonas W Perez
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia, United States
| | - David A Ford
- Department of Biochemistry and Molecular Biology, Saint Louis University, St. Louis, Missouri, United States
| | - Rakesh P Patel
- Center for Free Radical Biology and Lung Injury and Repair Center, The University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Thomas A Blake
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia, United States
| | - Michael D Gunn
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States
| | - Sven E Jordt
- Department of Anesthesiology, Duke University School of Medicine, Durham, North Carolina, United States
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, United States
- Integrated Toxicology & Environmental Health Program, Duke University, Durham, North Carolina, United States
| |
Collapse
|
8
|
Roberts JD. LungElast-an open-source, flexible, low-cost, microprocessor-controlled mouse lung elastometer. Sci Rep 2023; 13:11246. [PMID: 37438462 PMCID: PMC10338507 DOI: 10.1038/s41598-023-38310-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 07/06/2023] [Indexed: 07/14/2023] Open
Abstract
The study of mouse lung mechanics provides essential insights into the physiological mechanisms of pulmonary disease. Consequently, investigators assemble custom systems comprising infusion-withdrawal syringe pumps and analog pressure sensors to investigate the lung function of these animals. But these systems are expensive and require ongoing regulation, making them challenging to use. Here I introduce LungElast, an open-source, inexpensive, and self-contained instrument that can experimentally determine lung elasticity and volumes even in immature mice. It is assembled using custom 3D printed parts and readily available or easily constructed components. In this device, a microprocessor-controlled stepper motor automatically regulates lung volume by precisely driving a syringe piston whose position is determined using time-of-flight LIDAR technology. The airway pressures associated with the lung volumes are determined using compact sensor-on-chip technology, retrieved in a digital format, and stored by the microcontroller. The instrument software is modular, which eases device testing, calibration, and use. Data are also provided here that specify the accuracy and precision of the elastometer's sensors and volume delivery and demonstrate its use with lung models and mouse pups. This instrument has excellent potential for research and educational work.
Collapse
Affiliation(s)
- Jesse D Roberts
- Cardiovascular Research Center of the General Medical Services and the Departments of Anesthesia, Critical Care and Pain Medicine, Pediatrics, and Medicine, Massachusetts General Hospital - East, 149 13th St, Boston, MA, USA.
- Harvard Medical School, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
9
|
Short-term PM exposure and social stress cause pulmonary and cardiac dysfunction. Toxicol Lett 2022; 370:66-73. [PMID: 36122649 DOI: 10.1016/j.toxlet.2022.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/25/2022] [Accepted: 09/12/2022] [Indexed: 11/20/2022]
Abstract
Ambient particulate matter (PM) exposure increases risk for cardiopulmonary health problems which may be exacerbated in a stressful environment. Co-exposure to PM and stress characterizes the experience of many deployed military personnel and first responders but has not been thoroughly investigated. This is especially relevant to military personnel who have been exposed to high PM levels in conjunction with stressful military conflict situations. To understand the mechanisms and time-course of the health consequences following burn pit exposure, we exposed mice to moderate levels of ambient PM less than 2.5 μM in diameter (PM2.5) alone or in combination with psychological stress. We found male mice exposed to PM2.5 alone or in combination with stress had significantly reduced pulmonary function when subjected to methacholine, indicating increased airway hyperreactivity. These mice experienced increased goblet cell hyperplasia in their lungs, with no change in alveolar density. Mice exposed to PM2.5 and/or stress also exhibited reduced cardiac contractility, right ventricular (RV) output, and changes in RV capillary density and cardiac inflammatory markers. Taken together, these data indicate that short-term exposure to PM2.5 with or without stress causes a clear reduction in pulmonary and cardiac function. We believe that this model is well-suited for the study of military and other occupational exposures, and future work will identify potential mechanisms, including the inflammatory progression of these co-exposures.
Collapse
|
10
|
Sallé-Lefort S, Miard S, Henry C, Arias-Reyes C, Marcouiller F, Beaulieu MJ, Aubin S, Lechasseur A, Jubinville É, Marsolais D, Morissette MC, Joseph V, Soliz J, Bossé Y, Picard F. Malat1 deficiency prevents hypoxia-induced lung dysfunction by protecting the access to alveoli. Front Physiol 2022; 13:949378. [PMID: 36105289 PMCID: PMC9464821 DOI: 10.3389/fphys.2022.949378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/02/2022] [Indexed: 12/02/2022] Open
Abstract
Hypoxia is common in lung diseases and a potent stimulator of the long non-coding RNA Metastasis-Associated Lung Adenocarcinoma Transcript 1 (MALAT1). Herein, we investigated the impact of Malat1 on hypoxia-induced lung dysfunction in mice. Malat1-deficient mice and their wild-type littermates were tested after 8 days of normoxia or hypoxia (10% oxygen). Hypoxia decreased elastance of the lung by increasing lung volume and caused in vivo hyperresponsiveness to methacholine without altering the contraction of airway smooth muscle. Malat1 deficiency also modestly decreased lung elastance but only when tested at low lung volumes and without altering lung volume and airway smooth muscle contraction. The in vivo responsiveness to methacholine was also attenuated by Malat1 deficiency, at least when elastance, a readout sensitive to small airway closure, was used to assess the response. More impressively, in vivo hyperresponsiveness to methacholine caused by hypoxia was virtually absent in Malat1-deficient mice, especially when hysteresivity, a readout sensitive to small airway narrowing heterogeneity, was used to assess the response. Malat1 deficiency also increased the coefficient of oxygen extraction and decreased ventilation in conscious mice, suggesting improvements in gas exchange and in clinical signs of respiratory distress during natural breathing. Combined with a lower elastance at low lung volumes at baseline, as well as a decreased propensity for small airway closure and narrowing heterogeneity during a methacholine challenge, these findings represent compelling evidence suggesting that the lack of Malat1 protects the access to alveoli for air entering the lung.
Collapse
Affiliation(s)
- Sandrine Sallé-Lefort
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, Canada
- Faculty of Pharmacy, Université Laval, Quebec, QC, Canada
| | - Stéphanie Miard
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, Canada
| | - Cyndi Henry
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, Canada
| | - Christian Arias-Reyes
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, Canada
- Faculty of Medicine, Université Laval, Quebec, QC, Canada
| | - François Marcouiller
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, Canada
| | - Marie-Josée Beaulieu
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, Canada
| | - Sophie Aubin
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, Canada
| | - Ariane Lechasseur
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, Canada
- Faculty of Medicine, Université Laval, Quebec, QC, Canada
| | - Éric Jubinville
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, Canada
- Faculty of Medicine, Université Laval, Quebec, QC, Canada
| | - David Marsolais
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, Canada
- Faculty of Medicine, Université Laval, Quebec, QC, Canada
| | - Mathieu C. Morissette
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, Canada
- Faculty of Medicine, Université Laval, Quebec, QC, Canada
| | - Vincent Joseph
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, Canada
- Faculty of Medicine, Université Laval, Quebec, QC, Canada
| | - Jorge Soliz
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, Canada
- Faculty of Medicine, Université Laval, Quebec, QC, Canada
| | - Ynuk Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, Canada
- Faculty of Medicine, Université Laval, Quebec, QC, Canada
- *Correspondence: Ynuk Bossé, ; Frédéric Picard,
| | - Frédéric Picard
- Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC, Canada
- Faculty of Pharmacy, Université Laval, Quebec, QC, Canada
- *Correspondence: Ynuk Bossé, ; Frédéric Picard,
| |
Collapse
|
11
|
Francisco D, Wang Y, Marshall C, Conway M, Addison KJ, Billheimer D, Kimura H, Numata M, Chu HW, Voelker DR, Kraft M, Ledford JG. Small Peptide Derivatives Within the Carbohydrate Recognition Domain of SP-A2 Modulate Asthma Outcomes in Mouse Models and Human Cells. Front Immunol 2022; 13:900022. [PMID: 35874703 PMCID: PMC9304716 DOI: 10.3389/fimmu.2022.900022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
Surfactant Protein-A (SP-A) is an innate immune modulator that regulates a variety of pulmonary host defense functions. We have shown that SP-A is dysfunctional in asthma, which could be partly due to genetic heterogeneity. In mouse models and primary bronchial epithelial cells from asthmatic participants, we evaluated the functional significance of a particular single nucleotide polymorphism of SP-A2, which results in an amino acid substitution at position 223 from glutamine (Q) to lysine (K) within the carbohydrate recognition domain (CRD). We found that SP-A 223Q humanized mice had greater protection from inflammation and mucin production after IL-13 exposure as compared to SP-A-2 223K mice. Likewise, asthmatic participants with two copies the major 223Q allele demonstrated better lung function and asthma control as compared to asthmatic participants with two copies of the minor SP-A 223K allele. In primary bronchial epithelial cells from asthmatic participants, full-length recombinant SP-A 223Q was more effective at reducing IL-13-induced MUC5AC gene expression compared to SP-A 223K. Given this activity, we developed 10 and 20 amino acid peptides of SP-A2 spanning position 223Q. We show that the SP-A 223Q peptides reduce eosinophilic inflammation, mucin production and airways hyperresponsiveness in a house dust mite model of asthma, protect from lung function decline during an IL-13 challenge model in mice, and decrease IL-13-induced MUC5AC gene expression in primary airway epithelial cells from asthmatic participants. These results suggest that position 223 within the CRD of SP-A2 may modulate several outcomes relevant to asthma, and that short peptides of SP-A2 retain anti-inflammatory properties similar to that of the endogenous protein.
Collapse
Affiliation(s)
- Dave Francisco
- Department of Medicine, University of Arizona, Tucson, AZ, United States
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Ying Wang
- Department of Medicine, University of Arizona, Tucson, AZ, United States
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Craig Marshall
- Department of Medicine, University of Arizona, Tucson, AZ, United States
| | - Michelle Conway
- Department of Medicine, University of Arizona, Tucson, AZ, United States
| | - Kenneth J. Addison
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Dean Billheimer
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Hiroki Kimura
- Department of Medicine, University of Arizona, Tucson, AZ, United States
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Mari Numata
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Hong W. Chu
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Dennis R. Voelker
- Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Monica Kraft
- Department of Medicine, University of Arizona, Tucson, AZ, United States
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Julie G. Ledford
- Asthma and Airway Disease Research Center, University of Arizona Health Sciences, Tucson, AZ, United States
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
12
|
Zieger M, Borel F, Greer C, Gernoux G, Blackwood M, Flotte TR, Mueller C. Liver-directed SERPINA1 gene therapy attenuates progression of spontaneous and tobacco smoke-induced emphysema in α1-antitrypsin null mice. Mol Ther Methods Clin Dev 2022; 25:425-438. [PMID: 35592360 PMCID: PMC9097330 DOI: 10.1016/j.omtm.2022.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 04/10/2022] [Indexed: 12/31/2022]
Abstract
α1-antitrypsin deficiency is a rare genetic condition that can cause liver and/or lung disease. There is currently no cure for this disorder, although repeated infusions of plasma-purified protein may slow down emphysema progression. Gene therapy in which a single recombinant adeno-associated viral vector (rAAV) administration would lead to sustained protein expression could therefore similarly affect disease progression, and provide the added benefits of reducing treatment burden and thereby improving the patient’s quality of life. The study presented here tests whether treating the Serpina1a-e knockout mouse model of α1-antitrypsin-deficiency lung disease with gene therapy would have an impact on the disease course, either on spontaneous disease caused by aging or on accelerated disease caused by exposure to cigarette smoke. Liver-directed gene therapy led to dose-dependent levels of biologically active human α1-antitrypsin protein. Furthermore, decreased lung compliance and increased elastic recoil indicate that treated mice had largely preserved lung tissue elasticity and alveolar wall integrity compared with untreated mice. rAAV-mediated gene augmentation is therefore able to compensate for the loss of function and restore a beneficial lung protease-antiprotease balance. This work constitutes a preclinical study report of a disease-modifying treatment in the Serpina1a-e knockout mouse model using a liver-specific rAAV serotype 8 capsid.
Collapse
Affiliation(s)
- Marina Zieger
- The Li Weibo Institute for Rare Diseases Research, Horae Gene Therapy Center, 368 Plantation Street, Worcester, MA 01605, USA
| | - Florie Borel
- The Li Weibo Institute for Rare Diseases Research, Horae Gene Therapy Center, 368 Plantation Street, Worcester, MA 01605, USA
| | - Cynthia Greer
- The Li Weibo Institute for Rare Diseases Research, Horae Gene Therapy Center, 368 Plantation Street, Worcester, MA 01605, USA
| | - Gwladys Gernoux
- The Li Weibo Institute for Rare Diseases Research, Horae Gene Therapy Center, 368 Plantation Street, Worcester, MA 01605, USA.,Department of Pediatrics, University of Massachusetts Chan Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Meghan Blackwood
- The Li Weibo Institute for Rare Diseases Research, Horae Gene Therapy Center, 368 Plantation Street, Worcester, MA 01605, USA
| | - Terence R Flotte
- The Li Weibo Institute for Rare Diseases Research, Horae Gene Therapy Center, 368 Plantation Street, Worcester, MA 01605, USA.,Department of Pediatrics, University of Massachusetts Chan Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Christian Mueller
- The Li Weibo Institute for Rare Diseases Research, Horae Gene Therapy Center, 368 Plantation Street, Worcester, MA 01605, USA.,Department of Pediatrics, University of Massachusetts Chan Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| |
Collapse
|
13
|
Bruggink S, Kentch K, Kronenfeld J, Renquist BJ. A Leak-Free Head-Out Plethysmography System to Accurately Assess Lung Function in Mice. J Appl Physiol (1985) 2022; 133:104-118. [DOI: 10.1152/japplphysiol.00835.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mice are a valuable model for elegant studies of complex, systems-dependent diseases, including pulmonary diseases. Current tools to assess lung function in mice are either terminal or lack accuracy. We set out to develop a low-cost, accurate, head-out variable-pressure plethysmography system to allow for repeated, non-terminal measurements of lung function in mice. Current head-out plethysmography systems are limited by air leaks that prevent accurate measures of volume and flow. We designed an inflatable cuff that encompasses the mouse's neck preventing air leak. We wrote corresponding software to collect and analyze the data, remove movement artifacts, and automatically calibrate each dataset. This software calculates inspiratory/expiratory volume, inspiratory/expiratory time, breaths per minute, mid-expiratory flow, and end-inspiratory pause. To validate the use, we established that our plethysmography system accurately measured tidal breathing, the bronchoconstrictive response to methacholine, sex and age associated changes in breathing, and breathing changes associated with house dust mite sensitization. Our estimates of volume, flow, and timing of breaths are in line with published estimates, we observed dose-dependent decreases in volume and flow in response to methacholine (P < 0.05), increased lung volume and decreased breathing rate with aging (P < 0.05), and that house dust mite sensitization decreased volume and flow (P <0.05) while exacerbating the methacholine induced increases in inspiratory and expiratory time (P < 0.05). We describe an accurate, sensitive, low-cost, head-out plethysmography system that allows for longitudinal studies of pulmonary disease in mice.
Collapse
Affiliation(s)
- Stephanie Bruggink
- Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, United States
- Physiological Sciences GIDP, University of Arizona, Tucson, AZ, United States
| | - Kyle Kentch
- Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, United States
| | - Jason Kronenfeld
- Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, United States
| | - Benjamin Jennings Renquist
- Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, United States
- Physiological Sciences GIDP, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
14
|
Borbet TC, Pawline MB, Zhang X, Wipperman MF, Reuter S, Maher T, Li J, Iizumi T, Gao Z, Daniele M, Taube C, Koralov SB, Müller A, Blaser MJ. Influence of the early-life gut microbiota on the immune responses to an inhaled allergen. Mucosal Immunol 2022; 15:1000-1011. [PMID: 35842561 PMCID: PMC9835105 DOI: 10.1038/s41385-022-00544-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 06/04/2022] [Accepted: 06/15/2022] [Indexed: 02/04/2023]
Abstract
Antibiotics, among the most used medications in children, affect gut microbiome communities and metabolic functions. These changes in microbiota structure can impact host immunity. We hypothesized that early-life microbiome alterations would lead to increased susceptibility to allergy and asthma. To test this, mouse pups between postnatal days 5-9 were orally exposed to water (control) or to therapeutic doses of azithromycin or amoxicillin. Later in life, these mice were sensitized and challenged with a model allergen, house dust mite (HDM), or saline. Mice with early-life azithromycin exposure that were challenged with HDM had increased IgE and IL-13 production by CD4+ T cells compared to unexposed mice; early-life amoxicillin exposure led to fewer abnormalities. To test that the microbiota contained the immunological cues to alter IgE and cytokine production after HDM challenge, germ-free mice were gavaged with fecal samples of the antibiotic-perturbed microbiota. Gavage of adult germ-free mice did not result in altered HDM responses, however, their offspring, which acquired the antibiotic-perturbed microbiota at birth showed elevated IgE levels and CD4+ cytokines in response to HDM, and altered airway reactivity. These studies indicate that early-life microbiota composition can heighten allergen-driven Th2/Th17 immune pathways and airway responses in an age-dependent manner.
Collapse
Affiliation(s)
- Timothy C. Borbet
- Department of Pathology, New York University School of Medicine, New York, NY USA
| | - Miranda B. Pawline
- Department of Pathology, New York University School of Medicine, New York, NY USA
| | - Xiaozhou Zhang
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Matthew F. Wipperman
- Immunology Program, Sloan Kettering Institute, New York, USA,Clinical and Translational Science Center, Weill Cornell Medicine, New York, New York, USA
| | - Sebastian Reuter
- Department of Pulmonary Medicine, University Hospital Essen – Ruhrlandklinik, Essen, Germany
| | - Timothy Maher
- Department of Pathology, New York University School of Medicine, New York, NY USA
| | - Jackie Li
- Department of Pathology, New York University School of Medicine, New York, NY USA
| | - Tadasu Iizumi
- Department of Pathology, New York University School of Medicine, New York, NY USA,Center for Advanced Biotechnology and Medicine, Rutgers University, New Brunswick, NJ, USA
| | - Zhan Gao
- Center for Advanced Biotechnology and Medicine, Rutgers University, New Brunswick, NJ, USA
| | - Megan Daniele
- Department of Pathology, New York University School of Medicine, New York, NY USA,Department of Pediatrics, New York Presbyterian/Morgan Stanley Children’s Hospital and Columbia University Irving Medical Center, New York, NY USA
| | - Christian Taube
- Immunology Program, Sloan Kettering Institute, New York, USA
| | - Sergei B. Koralov
- Department of Pathology, New York University School of Medicine, New York, NY USA
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland,Corresponding Authors: Martin J. Blaser, , Center for Advanced Biotechnology and Medicine, Rutgers University, 679 Hoes Lane West, Room 106A, Piscataway, NJ 08854, Tel: 848-445-9834, Fax: 732-235-5318, Anne Müller, , Universität Zürich, Institut für Molekulare Krebsforschung, Winterthurerstrasse 190, CH 8057 Zürich, Tel: +41 44 635 34 74, Fax: +41 44 635 3484
| | - Martin J. Blaser
- Center for Advanced Biotechnology and Medicine, Rutgers University, New Brunswick, NJ, USA,Corresponding Authors: Martin J. Blaser, , Center for Advanced Biotechnology and Medicine, Rutgers University, 679 Hoes Lane West, Room 106A, Piscataway, NJ 08854, Tel: 848-445-9834, Fax: 732-235-5318, Anne Müller, , Universität Zürich, Institut für Molekulare Krebsforschung, Winterthurerstrasse 190, CH 8057 Zürich, Tel: +41 44 635 34 74, Fax: +41 44 635 3484
| |
Collapse
|
15
|
Nitta NA, Sato T, Komura M, Yoshikawa H, Suzuki Y, Mitsui A, Kuwasaki E, Takahashi F, Kodama Y, Seyama K, Takahashi K. Exposure to the heated tobacco product IQOS generates apoptosis-mediated pulmonary emphysema in murine lungs. Am J Physiol Lung Cell Mol Physiol 2022; 322:L699-L711. [PMID: 35380471 DOI: 10.1152/ajplung.00215.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 03/21/2022] [Accepted: 03/31/2022] [Indexed: 11/22/2022] Open
Abstract
Pulmonary emphysema is predominantly caused by chronic exposure to cigarette smoke (CS). Novel tobacco substitutes, such as heated tobacco products (HTPs), have emerged as healthier alternatives to cigarettes. IQOS, the most popular HTP in Japan, is advertised as harmless compared with conventional cigarettes. Although some studies have reported its toxicity, few in vivo studies have been conducted. Here, 12-wk-old C57BL6/J male mice were divided into three groups and exposed to air (as control), IQOS aerosol, or CS for 6 mo. After exposure, the weight gain was significantly suppressed in the IQOS and CS groups compared with the control (-4.93 g; IQOS vs. air and -5.504 g; CS vs. air). The serum cotinine level was significantly higher in the IQOS group than in the control group. The neutrophils and lymphocyte count increased in the bronchoalveolar lavage fluid of the IQOS and CS groups compared with those in the control group. Chronic IQOS exposure induced pulmonary emphysema similar to that observed in the CS group. Furthermore, expression levels of the genes involved in the apoptosis-related pathways were significantly upregulated in the lungs of the IQOS-exposed mice. Cytochrome c, cleaved caspase-3, and cleaved poly (ADP-ribose) polymerase-1 were overexpressed in the IQOS group compared with the control. Single-stranded DNA and TdT-mediated dUTP nick-end labeling-positive alveolar septal cell count significantly increased in the IQOS group compared with the control. In conclusion, chronic exposure to IQOS aerosol induces pulmonary emphysema predominantly via apoptosis-related pathways. This suggests that HTPs are not completely safe tobacco products.
Collapse
Affiliation(s)
- Naoko Arano Nitta
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tadashi Sato
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Moegi Komura
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hitomi Yoshikawa
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yohei Suzuki
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Aki Mitsui
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Eriko Kuwasaki
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Fumiyuki Takahashi
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuzo Kodama
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kuniaki Seyama
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kazuhisa Takahashi
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
16
|
High-Fat High-Sugar Diet-Induced Changes in the Lipid Metabolism Are Associated with Mildly Increased COVID-19 Severity and Delayed Recovery in the Syrian Hamster. Viruses 2021; 13:v13122506. [PMID: 34960775 PMCID: PMC8703573 DOI: 10.3390/v13122506] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 12/15/2022] Open
Abstract
Pre-existing comorbidities such as obesity or metabolic diseases can adversely affect the clinical outcome of COVID-19. Chronic metabolic disorders are globally on the rise and often a consequence of an unhealthy diet, referred to as a Western Diet. For the first time in the Syrian hamster model, we demonstrate the detrimental impact of a continuous high-fat high-sugar diet on COVID-19 outcome. We observed increased weight loss and lung pathology, such as exudate, vasculitis, hemorrhage, fibrin, and edema, delayed viral clearance and functional lung recovery, and prolonged viral shedding. This was accompanied by an altered, but not significantly different, systemic IL-10 and IL-6 profile, as well as a dysregulated serum lipid response dominated by polyunsaturated fatty acid-containing phosphatidylethanolamine, partially recapitulating cytokine and lipid responses associated with severe human COVID-19. Our data support the hamster model for testing restrictive or targeted diets and immunomodulatory therapies to mediate the adverse effects of metabolic disease on COVID-19.
Collapse
|
17
|
Routhier J, Pons S, Freidja ML, Dalstein V, Cutrona J, Jonquet A, Lalun N, Mérol JC, Lathrop M, Stitzel JA, Kervoaze G, Pichavant M, Gosset P, Tournier JM, Birembaut P, Dormoy V, Maskos U. An innate contribution of human nicotinic receptor polymorphisms to COPD-like lesions. Nat Commun 2021; 12:6384. [PMID: 34737286 PMCID: PMC8568944 DOI: 10.1038/s41467-021-26637-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/14/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic Obstructive Pulmonary Disease is a generally smoking-linked major cause of morbidity and mortality. Genome-wide Association Studies identified a locus including a non-synonymous single nucleotide polymorphism in CHRNA5, rs16969968, encoding the nicotinic acetylcholine receptor α5 subunit, predisposing to both smoking and Chronic Obstructive Pulmonary Disease. Here we report that nasal polyps from rs16969968 non-smoking carriers exhibit airway epithelium remodeling and inflammation. These hallmarks of Chronic Obstructive Pulmonary Disease occur spontaneously in mice expressing human rs16969968. They are significantly amplified after exposure to porcine pancreatic elastase, an emphysema model, and to oxidative stress with a polymorphism-dependent alteration of lung function. Targeted rs16969968 expression in epithelial cells leads to airway remodeling in vivo, increased proliferation and production of pro-inflammatory cytokines through decreased calcium entry and increased adenylyl-cyclase activity. We show that rs16969968 directly contributes to Chronic Obstructive Pulmonary Disease-like lesions, sensitizing the lung to the action of oxidative stress and injury, and represents a therapeutic target. Human polymorphisms in nicotinic acetylcholine receptor genes have been linked to both smoking and lung diseases like Chronic Obstructive Pulmonary Disease (COPD) or lung cancer. Here the authors identify a direct role for a human coding polymorphism in COPD-like lesions independent of smoke or nicotine exposure.
Collapse
Affiliation(s)
- Julie Routhier
- Université de Reims Champagne-Ardenne, Inserm, P3Cell UMR-S1250, Reims, France
| | - Stéphanie Pons
- Institut Pasteur, Université de Paris, Integrative Neurobiology of Cholinergic Systems, CNRS UMR 3571, Paris, France
| | - Mohamed Lamine Freidja
- Université de Reims Champagne-Ardenne, Inserm, P3Cell UMR-S1250, Reims, France.,Department of Biochemistry and Microbiology, Faculty of Sciences, University of M'sila, M'sila, Algeria
| | - Véronique Dalstein
- Université de Reims Champagne-Ardenne, Inserm, P3Cell UMR-S1250, Reims, France.,Department of Biopathology, CHU of Reims, Reims, France
| | - Jérôme Cutrona
- Université de Reims Champagne-Ardenne, Inserm, P3Cell UMR-S1250, Reims, France
| | - Antoine Jonquet
- Université de Reims Champagne-Ardenne, Inserm, P3Cell UMR-S1250, Reims, France
| | - Nathalie Lalun
- Université de Reims Champagne-Ardenne, Inserm, P3Cell UMR-S1250, Reims, France
| | - Jean-Claude Mérol
- Université de Reims Champagne-Ardenne, Inserm, P3Cell UMR-S1250, Reims, France.,Department of Otorhinolaryngology, CHU of Reims, Reims, France
| | - Mark Lathrop
- McGill University Genome Center, Montréal, QC, Canada
| | - Jerry A Stitzel
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO, USA
| | - Gwenola Kervoaze
- University of Lille, CNRS UMR9017, Inserm U1019, CHU Lille, Institut Pasteur de Lille, CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Muriel Pichavant
- University of Lille, CNRS UMR9017, Inserm U1019, CHU Lille, Institut Pasteur de Lille, CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Philippe Gosset
- University of Lille, CNRS UMR9017, Inserm U1019, CHU Lille, Institut Pasteur de Lille, CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Jean-Marie Tournier
- Université de Reims Champagne-Ardenne, Inserm, P3Cell UMR-S1250, Reims, France
| | - Philippe Birembaut
- Université de Reims Champagne-Ardenne, Inserm, P3Cell UMR-S1250, Reims, France.,Department of Biopathology, CHU of Reims, Reims, France
| | - Valérian Dormoy
- Université de Reims Champagne-Ardenne, Inserm, P3Cell UMR-S1250, Reims, France.
| | - Uwe Maskos
- Institut Pasteur, Université de Paris, Integrative Neurobiology of Cholinergic Systems, CNRS UMR 3571, Paris, France.
| |
Collapse
|
18
|
An TJ, Kim JH, Park CK, Yoon HK. Tiotropium bromide has a more potent effect than corticosteroid in the acute neutrophilic asthma mouse model. Tuberc Respir Dis (Seoul) 2021; 85:18-24. [PMID: 34727490 PMCID: PMC8743638 DOI: 10.4046/trd.2021.0118] [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: 07/29/2021] [Accepted: 10/30/2021] [Indexed: 11/24/2022] Open
Abstract
Background Neutrophilic asthma (NeuA) is usually resistant to corticosteroids. Tiotropium bromide (TIO) is a bronchodilator that is used as an add-on therapy to inhaled corticosteroid and long-acting β2 agonist in asthma treatment. However, the role of TIO in NeuA is not fully known. Thus, the aim of this study was to evaluate the effect of TIO on NeuA compared to that of corticosteroids. Methods C57BL/6 female mice were sensitized with ovalbumin and lipopolysaccharide to induce neutrophilic inflammation. Dexamethasone (DEX) was administered on days 14, 17, 20, and 23. TIO was inhaled on days 21, 21, and 23. On day 24, mice were sacrificed. Airway hyper-responsiveness, levels of cytokines in bronchoalveolar lavage (BAL) and lung homogenates, and lung tissue histopathology were compared between the two groups. Results Neutrophil counts, T helper 2 cells (TH2)/TH17 cytokines, and pro-inflammatory cytokine in BAL fluids were elevated in the NeuA group. TIO group showed lower total cells, neutrophil counts, and eosinophil counts in BAL fluids than the DEX group (p<0.001, p<0.05, and p<0.001, respectively). Airway resistance was attenuated in the TIO group but elevated in the NeuA group (p<0.001). Total protein, interleukin (IL)-5, and IL-17A levels in BAL fluids were lower in the TIO group than in the NeuA group (all p<0.05). Conclusion TIO showed more potent effects than DEX in improving airway inflammation and attenuating airway resistance in NeuA.
Collapse
Affiliation(s)
- Tai Joon An
- Division of Pulmonary and Critical Care Medicine, Department of Internal medicine, Yeouido St. Mary`s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ji Hye Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal medicine, Yeouido St. Mary`s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chan Kwon Park
- Division of Pulmonary and Critical Care Medicine, Department of Internal medicine, Yeouido St. Mary`s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyoung Kyu Yoon
- Division of Pulmonary and Critical Care Medicine, Department of Internal medicine, Yeouido St. Mary`s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
19
|
Fujioka N, Kitabatake M, Ouji-Sageshima N, Ibaraki T, Kumamoto M, Fujita Y, Hontsu S, Yamauchi M, Yoshikawa M, Muro S, Ito T. Human Adipose-Derived Mesenchymal Stem Cells Ameliorate Elastase-Induced Emphysema in Mice by Mesenchymal-Epithelial Transition. Int J Chron Obstruct Pulmon Dis 2021; 16:2783-2793. [PMID: 34675503 PMCID: PMC8517419 DOI: 10.2147/copd.s324952] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/13/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose Chronic obstructive pulmonary disease (COPD) is a worldwide problem because of its high prevalence and mortality. However, there is no fundamental treatment to ameliorate their pathological change in COPD lung. Recently, adipose-derived mesenchymal stem cells (ADSCs) have attracted attention in the field of regenerative medicine to repair damaged organs. Moreover, their utility in treating respiratory diseases has been reported in some animal models. However, the detailed mechanism by which ADSCs improve chronic respiratory diseases, including COPD, remains to be elucidated. We examined whether human ADSCs (hADSCs) ameliorated elastase-induced emphysema and whether hADSCs differentiated into alveolar epithelial cells in a murine model of COPD. Methods Female SCID-beige mice (6 weeks old) were divided into the following four groups according to whether they received an intratracheal injection of phosphate-buffered saline or porcine pancreatic elastase, and whether they received an intravenous injection of saline or hADSCs 3 days after intratracheal injection; Control group, hADSC group, Elastase group, and Elastase-hADSC group. We evaluated the lung function, assessed histological changes, and compared gene expression between hADSCs isolated from the lung of Elastase-hADSC group and naïve hADSCs 28 days after saline or elastase administration. Results hADSCs improved the pathogenesis of COPD, including the mean linear intercept and forced expiratory volume, in an elastase-induced emphysema model in mice. Furthermore, hADSCs were observed in the lungs of elastase-treated mice at 25 days after administration. These cells expressed genes related to mesenchymal–epithelial transition and surface markers of alveolar epithelial cells, such as TTF-1, β-catenin, and E-cadherin. Conclusion hADSCs have the potential to improve the pathogenesis of COPD by differentiating into alveolar epithelial cells by mesenchymal–epithelial transition.
Collapse
Affiliation(s)
- Nobuhiro Fujioka
- Department of Respiratory Medicine, Nara Medical University, Kashihara, Nara, Japan
| | | | | | - Takahiro Ibaraki
- Department of Respiratory Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Makiko Kumamoto
- Department of Respiratory Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Yukio Fujita
- Department of Respiratory Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Shigeto Hontsu
- Department of Respiratory Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Motoo Yamauchi
- Department of Respiratory Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Masanori Yoshikawa
- Department of Respiratory Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Shigeo Muro
- Department of Respiratory Medicine, Nara Medical University, Kashihara, Nara, Japan
| | - Toshihiro Ito
- Department of Immunology, Nara Medical University, Kashihara, Nara, Japan
| |
Collapse
|
20
|
Cass SP, Mekhael O, Thayaparan D, McGrath JJC, Revill SD, Fantauzzi MF, Wang P, Reihani A, Hayat AI, Stevenson CS, Dvorkin-Gheva A, Botelho FM, Stämpfli MR, Ask K. Increased Monocyte-Derived CD11b + Macrophage Subpopulations Following Cigarette Smoke Exposure Are Associated With Impaired Bleomycin-Induced Tissue Remodelling. Front Immunol 2021; 12:740330. [PMID: 34603325 PMCID: PMC8481926 DOI: 10.3389/fimmu.2021.740330] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/30/2021] [Indexed: 01/16/2023] Open
Abstract
Rationale The accumulation of macrophages in the airways and the pulmonary interstitium is a hallmark of cigarette smoke-associated inflammation. Notably, pulmonary macrophages are not a homogenous population but consist of several subpopulations. To date, the manner in which cigarette smoke exposure affects the relative composition and functional capacity of macrophage subpopulations has not been elucidated. Methods Using a whole-body cigarette smoke exposure system, we investigated the impact of cigarette smoke on macrophage subpopulations in C57BL/6 mice using flow cytometry-based approaches. Moreover, we used bromodeoxyuridine labelling plus Il1a-/- and Il1r1-/- mice to assess the relative contribution of local proliferation and monocyte recruitment to macrophage accumulation. To assess the functional consequences of altered macrophage subpopulations, we used a model of concurrent bleomycin-induced lung injury and cigarette smoke exposure to examine tissue remodelling processes. Main Results Cigarette smoke exposure altered the composition of pulmonary macrophages increasing CD11b+ subpopulations including monocyte-derived alveolar macrophages (Mo-AM) as well as interstitial macrophages (IM)1, -2 and -3. The increase in CD11b+ subpopulations was observed at multiple cigarette smoke exposure timepoints. Bromodeoxyuridine labelling and studies in Il1a-/- mice demonstrated that increased Mo-AM and IM3 turnover in the lungs of cigarette smoke-exposed mice was IL-1α dependent. Compositional changes in macrophage subpopulations were associated with impaired induction of fibrogenesis including decreased α-smooth muscle actin positive cells following intratracheal bleomycin treatment. Mechanistically, in vivo and ex vivo assays demonstrated predominant macrophage M1 polarisation and reduced matrix metallopeptidase 9 activity in cigarette smoke-exposed mice. Conclusion Cigarette smoke exposure modified the composition of pulmonary macrophage by expanding CD11b+ subpopulations. These compositional changes were associated with attenuated fibrogenesis, as well as predominant M1 polarisation and decreased fibrotic activity. Overall, these data suggest that cigarette smoke exposure altered the composition of pulmonary macrophage subpopulations contributing to impaired tissue remodelling.
Collapse
Affiliation(s)
- Steven P Cass
- Medical Sciences Graduate Program, McMaster University, Hamilton, ON, Canada
| | - Olivia Mekhael
- Medical Sciences Graduate Program, McMaster University, Hamilton, ON, Canada
| | - Danya Thayaparan
- Medical Sciences Graduate Program, McMaster University, Hamilton, ON, Canada
| | - Joshua J C McGrath
- Medical Sciences Graduate Program, McMaster University, Hamilton, ON, Canada
| | - Spencer D Revill
- Medical Sciences Graduate Program, McMaster University, Hamilton, ON, Canada.,Department of Medicine, Firestone Institute for Respiratory Health, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, ON, Canada
| | - Matthew F Fantauzzi
- Medical Sciences Graduate Program, McMaster University, Hamilton, ON, Canada
| | - Peiyao Wang
- Department Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Amir Reihani
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, ON, Canada
| | - Aaron I Hayat
- Medical Sciences Graduate Program, McMaster University, Hamilton, ON, Canada
| | - Christopher S Stevenson
- Janssen Disease Interception Accelerator, Janssen Pharmaceutical Companies of Johnson and Johnson, Raritan, NJ, United States
| | - Anna Dvorkin-Gheva
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Fernando M Botelho
- Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Martin R Stämpfli
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, ON, Canada.,Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Kjetil Ask
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, ON, Canada.,Department of Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
21
|
Lu P, ElMallah MK, Liu Z, Wu C, Chen J, Lifshitz LM, ZhuGe R. Genetic deletion of the Tas2r143/Tas2r135/Tas2r126 cluster reveals that TAS2Rs may not mediate bitter tastant-induced bronchodilation. J Cell Physiol 2021; 236:6407-6423. [PMID: 33559206 PMCID: PMC8223514 DOI: 10.1002/jcp.30315] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 01/19/2021] [Accepted: 01/27/2021] [Indexed: 11/09/2022]
Abstract
Bitter taste receptors (TAS2Rs) and their signaling elements are detected throughout the body, and bitter tastants induce a wide variety of biological responses in tissues and organs outside the mouth. However, the roles of TAS2Rs in these responses remain to be tested and established genetically. Here, we employed the CRISPR/Cas9 gene-editing technique to delete three bitter taste receptors-Tas2r143/Tas2r135/Tas2r126 (i.e., Tas2r triple knockout [TKO]) in mice. The fidelity and effectiveness of the Tas2r deletions were validated genetically at DNA and messenger RNA levels and functionally based on the tasting of TAS2R135 and TAS2R126 agonists. Bitter tastants are known to relax airways completely. However, TAS2R135 or TAS2R126 agonists either failed to induce relaxation of pre-contracted airways in wild-type mice and Tas2r TKO mice or relaxed them dose-dependently, but to the same extent in both types of mice. These results indicate that TAS2Rs are not required for bitter tastant-induced bronchodilation. The Tas2r TKO mice also provide a valuable model to resolve whether TAS2Rs mediate bitter tastant-induced responses in many other extraoral tissues.
Collapse
Affiliation(s)
- Ping Lu
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Mai K ElMallah
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Zeyu Liu
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Chan Wu
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Jun Chen
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Lawrence M Lifshitz
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Ronghua ZhuGe
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
22
|
Mekhael O, Naiel S, Vierhout M, Hayat AI, Revill SD, Abed S, Inman MD, Kolb MRJ, Ask K. Mouse Models of Lung Fibrosis. Methods Mol Biol 2021; 2299:291-321. [PMID: 34028751 DOI: 10.1007/978-1-0716-1382-5_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
The drug discovery pipeline, from discovery of therapeutic targets through preclinical and clinical development phases, to an approved product by health authorities, is a time-consuming and costly process, where a lead candidates' success at reaching the final stage is rare. Although the time from discovery to final approval has been reduced over the last decade, there is still potential to further optimize and streamline the evaluation process of each candidate as it moves through the different development phases. In this book chapter, we describe our preclinical strategies and overall decision-making process designed to evaluate the tolerability and efficacy of therapeutic candidates suitable for patients diagnosed with fibrotic lung disease. We also describe the benefits of conducting preliminary discovery trials, to aid in the selection of suitable primary and secondary outcomes to be further evaluated and assessed in subsequent internal and external validation studies. We outline all relevant research methodologies and protocols routinely performed by our research group and hope that these strategies and protocols will be a useful guide for biomedical and translational researchers aiming to develop safe and beneficial therapies for patients with fibrotic lung disease.
Collapse
Affiliation(s)
- Olivia Mekhael
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, ON, Canada
| | - Safaa Naiel
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, ON, Canada
| | - Megan Vierhout
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, ON, Canada
| | - Aaron I Hayat
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, ON, Canada
| | - Spencer D Revill
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, ON, Canada
| | - Soumeya Abed
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, ON, Canada
| | - Mark D Inman
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, ON, Canada
| | - Martin R J Kolb
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, ON, Canada
| | - Kjetil Ask
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University and The Research Institute of St. Joe's Hamilton, Hamilton, ON, Canada.
| |
Collapse
|
23
|
Case JB, Chen RE, Cao L, Ying B, Winkler ES, Johnson M, Goreshnik I, Pham MN, Shrihari S, Kafai NM, Bailey AL, Xie X, Shi PY, Ravichandran R, Carter L, Stewart L, Baker D, Diamond MS. Ultrapotent miniproteins targeting the SARS-CoV-2 receptor-binding domain protect against infection and disease. Cell Host Microbe 2021; 29:1151-1161.e5. [PMID: 34192518 PMCID: PMC8221914 DOI: 10.1016/j.chom.2021.06.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/23/2021] [Accepted: 06/11/2021] [Indexed: 12/13/2022]
Abstract
Despite the introduction of public health measures and spike protein-based vaccines to mitigate the COVID-19 pandemic, SARS-CoV-2 infections and deaths continue to have a global impact. Previously, we used a structural design approach to develop picomolar range miniproteins targeting the SARS-CoV-2 spike receptor-binding domain. Here, we investigated the capacity of modified versions of one lead miniprotein, LCB1, to protect against SARS-CoV-2-mediated lung disease in mice. Systemic administration of LCB1-Fc reduced viral burden, diminished immune cell infiltration and inflammation, and completely prevented lung disease and pathology. A single intranasal dose of LCB1v1.3 reduced SARS-CoV-2 infection in the lung when given as many as 5 days before or 2 days after virus inoculation. Importantly, LCB1v1.3 protected in vivo against a historical strain (WA1/2020), an emerging B.1.1.7 strain, and a strain encoding key E484K and N501Y spike protein substitutions. These data support development of LCB1v1.3 for prevention or treatment of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- James Brett Case
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rita E Chen
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Longxing Cao
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Baoling Ying
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Emma S Winkler
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Max Johnson
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Inna Goreshnik
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Minh N Pham
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Swathi Shrihari
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Natasha M Kafai
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Adam L Bailey
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xuping Xie
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA; Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Rashmi Ravichandran
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Lauren Carter
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Lance Stewart
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
24
|
Cinar R, Park JK, Zawatsky CN, Coffey NJ, Bodine SP, Abdalla J, Yokoyama T, Jourdan T, Jay L, Zuo MXG, O'Brien KJ, Huang J, Mackie K, Alimardanov A, Iyer MR, Gahl WA, Kunos G, Gochuico BR, Malicdan MCV. CB 1 R and iNOS are distinct players promoting pulmonary fibrosis in Hermansky-Pudlak syndrome. Clin Transl Med 2021; 11:e471. [PMID: 34323400 PMCID: PMC8255071 DOI: 10.1002/ctm2.471] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 06/02/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023] Open
Abstract
Hermansky-Pudlak syndrome (HPS) is a rare genetic disorder which, in its most common and severe form, HPS-1, leads to fatal adult-onset pulmonary fibrosis (PF) with no effective treatment. We evaluated the role of the endocannabinoid/CB1 R system and inducible nitric oxide synthase (iNOS) for dual-target therapeutic strategy using human bronchoalveolar lavage fluid (BALF), lung samples from patients with HPS and controls, HPS-PF patient-derived lung fibroblasts, and bleomycin-induced PF in pale ear mice (HPS1ep/ep ). We found overexpression of CB1 R and iNOS in fibrotic lungs of HPSPF patients and bleomycin-infused pale ear mice. The endocannabinoid anandamide was elevated in BALF and negatively correlated with pulmonary function parameters in HPSPF patients and pale ear mice with bleomycin-induced PF. Simultaneous targeting of CB1 R and iNOS by MRI-1867 yielded greater antifibrotic efficacy than inhibiting either target alone by attenuating critical pathologic pathways. Moreover, MRI-1867 treatment abrogated bleomycin-induced increases in lung levels of the profibrotic interleukin-11 via iNOS inhibition and reversed mitochondrial dysfunction via CB1 R inhibition. Dual inhibition of CB1 R and iNOS is an effective antifibrotic strategy for HPSPF.
Collapse
Affiliation(s)
- Resat Cinar
- Section on Fibrotic DisordersNational Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthMarylandUSA
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
| | - Joshua K. Park
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
| | - Charles N. Zawatsky
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
| | - Nathan J. Coffey
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
| | - Steven P. Bodine
- Section of Human Biochemical GeneticsMedical Genetics BranchNational Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Jasmina Abdalla
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
| | - Tadafumi Yokoyama
- Section of Human Biochemical GeneticsMedical Genetics BranchNational Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
- Present address:
Department of PediatricsKanazawa UniversityKanazawaJapan
| | - Tony Jourdan
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
- Present address:
INSERM Lipids, Nutrition, Cancer UMR1231University of Burgundy and Franche‐ComtéDijonFrance
| | - Lindsey Jay
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
| | - Mei Xing G. Zuo
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
| | - Kevin J. O'Brien
- Section of Human Biochemical GeneticsMedical Genetics BranchNational Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Junfeng Huang
- Therapeutics Development BranchDivision of Preclinical InnovationNational Center for Advancing Translational SciencesNational Institutes of HealthRockvilleMarylandUSA
| | - Ken Mackie
- Department of Psychological and Brain SciencesIndiana UniversityBloomingtonIndianaUSA
| | - Asaf Alimardanov
- Therapeutics Development BranchDivision of Preclinical InnovationNational Center for Advancing Translational SciencesNational Institutes of HealthRockvilleMarylandUSA
| | - Malliga R. Iyer
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
| | - William A. Gahl
- Section of Human Biochemical GeneticsMedical Genetics BranchNational Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
- NIH Undiagnosed Diseases Program and Office of the Clinical DirectorNational Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - George Kunos
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
| | - Bernadette R. Gochuico
- Section of Human Biochemical GeneticsMedical Genetics BranchNational Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - May Christine V. Malicdan
- Section of Human Biochemical GeneticsMedical Genetics BranchNational Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
- NIH Undiagnosed Diseases Program and Office of the Clinical DirectorNational Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
25
|
Jaeger N, McDonough RT, Rosen AL, Hernandez-Leyva A, Wilson NG, Lint MA, Russler-Germain EV, Chai JN, Bacharier LB, Hsieh CS, Kau AL. Airway Microbiota-Host Interactions Regulate Secretory Leukocyte Protease Inhibitor Levels and Influence Allergic Airway Inflammation. Cell Rep 2021; 33:108331. [PMID: 33147448 PMCID: PMC7685510 DOI: 10.1016/j.celrep.2020.108331] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 08/22/2020] [Accepted: 10/08/2020] [Indexed: 01/04/2023] Open
Abstract
Homeostatic mucosal immune responses are fine-tuned by naturally evolved interactions with native microbes, and integrating these relationships into experimental models can provide new insights into human diseases. Here, we leverage a murine-adapted airway microbe, Bordetella pseudohinzii (Bph), to investigate how chronic colonization impacts mucosal immunity and the development of allergic airway inflammation (AAI). Colonization with Bph induces the differentiation of interleukin-17A (IL-17A)-secreting T-helper cells that aid in controlling bacterial abundance. Bph colonization protects from AAI and is associated with increased production of secretory leukocyte protease inhibitor (SLPI), an antimicrobial peptide with anti-inflammatory properties. These findings are additionally supported by clinical data showing that higher levels of upper respiratory SLPI correlate both with greater asthma control and the presence of Haemophilus, a bacterial genus associated with AAI. We propose that SLPI could be used as a biomarker of beneficial host-commensal relationships in the airway. Asthma is known to be modified by airway microbes. Jaeger et al. use a murine-adapted bacterium to show that airway colonization evokes a Th17 response associated with increased SLPI, an antimicrobial peptide, and protection from lung inflammation. In people, SLPI was correlated with airway microbiota composition.
Collapse
Affiliation(s)
- Natalia Jaeger
- Division of Allergy and Immunology, Department of Medicine and Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ryan T McDonough
- Division of Allergy and Immunology, Department of Medicine and Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Anne L Rosen
- Division of Allergy and Immunology, Department of Medicine and Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ariel Hernandez-Leyva
- Division of Allergy and Immunology, Department of Medicine and Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Naomi G Wilson
- Division of Allergy and Immunology, Department of Medicine and Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael A Lint
- Division of Allergy and Immunology, Department of Medicine and Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Emilie V Russler-Germain
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jiani N Chai
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Leonard B Bacharier
- Division of Allergy, Immunology and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chyi-Song Hsieh
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrew L Kau
- Division of Allergy and Immunology, Department of Medicine and Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
26
|
Khadangi F, Forgues AS, Tremblay-Pitre S, Dufour-Mailhot A, Henry C, Boucher M, Beaulieu MJ, Morissette M, Fereydoonzad L, Brunet D, Robichaud A, Bossé Y. Intranasal versus intratracheal exposure to lipopolysaccharides in a murine model of acute respiratory distress syndrome. Sci Rep 2021; 11:7777. [PMID: 33833346 PMCID: PMC8032690 DOI: 10.1038/s41598-021-87462-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
Due to frequent and often severe lung affections caused by COVID-19, murine models of acute respiratory distress syndrome (ARDS) are increasingly used in experimental lung research. The one induced by a single lipopolysaccharide (LPS) exposure is practical. However, whether it is preferable to administer LPS intranasally or intratracheally remains an open question. Herein, female C57Bl/6 J mice were exposed intranasally or intratracheally to one dose of either saline or 3 mg/kg of LPS. They were studied 24 h later. The groups treated with LPS, either intranasally or intratracheally, exhibited a pronounced neutrophilic inflammation, signs of lung tissue damage and protein extravasation into the alveoli, and mild lung dysfunction. The magnitude of the response was generally not different between groups exposed intranasally versus intratracheally. However, the variability of some the responses was smaller in the LPS-treated groups exposed intranasally versus intratracheally. Notably, the saline-treated mice exposed intratracheally demonstrated a mild neutrophilic inflammation and alterations of the airway epithelium. We conclude that an intranasal exposure is as effective as an intratracheal exposure in a murine model of ARDS induced by LPS. Additionally, the groups exposed intranasally demonstrated less variability in the responses to LPS and less complications associated with the sham procedure.
Collapse
Affiliation(s)
- Fatemeh Khadangi
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Pavillon Mallet, M2694, 2725, chemin Sainte-Foy, Quebec, QC, G1V 4G5, Canada
| | - Anne-Sophie Forgues
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Pavillon Mallet, M2694, 2725, chemin Sainte-Foy, Quebec, QC, G1V 4G5, Canada
| | - Sophie Tremblay-Pitre
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Pavillon Mallet, M2694, 2725, chemin Sainte-Foy, Quebec, QC, G1V 4G5, Canada
| | - Alexis Dufour-Mailhot
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Pavillon Mallet, M2694, 2725, chemin Sainte-Foy, Quebec, QC, G1V 4G5, Canada
| | - Cyndi Henry
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Pavillon Mallet, M2694, 2725, chemin Sainte-Foy, Quebec, QC, G1V 4G5, Canada
| | - Magali Boucher
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Pavillon Mallet, M2694, 2725, chemin Sainte-Foy, Quebec, QC, G1V 4G5, Canada
| | - Marie-Josée Beaulieu
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Pavillon Mallet, M2694, 2725, chemin Sainte-Foy, Quebec, QC, G1V 4G5, Canada
| | - Mathieu Morissette
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Pavillon Mallet, M2694, 2725, chemin Sainte-Foy, Quebec, QC, G1V 4G5, Canada
| | | | - David Brunet
- SCIREQ - Scientific Respiratory Equipment Inc., Montreal, Canada
| | | | - Ynuk Bossé
- Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Pavillon Mallet, M2694, 2725, chemin Sainte-Foy, Quebec, QC, G1V 4G5, Canada.
| |
Collapse
|
27
|
Lundblad LKA, Robichaud A. Oscillometry of the respiratory system: a translational opportunity not to be missed. Am J Physiol Lung Cell Mol Physiol 2021; 320:L1038-L1056. [PMID: 33822645 PMCID: PMC8203417 DOI: 10.1152/ajplung.00222.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Airway oscillometry has become the de facto standard for quality assessment of lung physiology in laboratory animals and has demonstrated its usefulness in understanding diseases of small airways. Nowadays, it is seeing extensive use in daily clinical practice and research; however, a question that remains unanswered is how well physiological findings in animals and humans correlate? Methodological and device differences are obvious between animal and human studies. However, all devices deliver an oscillated airflow test signal and output respiratory impedance. In addition, despite analysis differences, there are ways to interpret animal and human oscillometry data to allow suitable comparisons. The potential with oscillometry is its ability to reveal universal features of the respiratory system across species, making translational extrapolation likely to be predictive. This means that oscillometry can thus help determine if an animal model displays the same physiological characteristics as the human disease. Perhaps more importantly, it can also be useful to determine whether an intervention is effective as well as to understand if it affects the desired region of the respiratory system, e.g., the periphery of the lung. Finally, findings in humans can also inform preclinical scientists and give indications as to what type of physiological changes should be observed in animal models to make them relevant as models of human disease. The present article will attempt to demonstrate the potential of oscillometry in respiratory research, an area where the development of novel therapies is plagued with a failure rate higher than in other disease areas.
Collapse
Affiliation(s)
- Lennart K A Lundblad
- Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada.,THORASYS Thoracic Medical Systems Inc., Montreal, Quebec, Canada
| | - Annette Robichaud
- SCIREQ Scientific Respiratory Equipment Inc., Montreal, Quebec, Canada
| |
Collapse
|
28
|
Winkler ES, Gilchuk P, Yu J, Bailey AL, Chen RE, Chong Z, Zost SJ, Jang H, Huang Y, Allen JD, Case JB, Sutton RE, Carnahan RH, Darling TL, Boon ACM, Mack M, Head RD, Ross TM, Crowe JE, Diamond MS. Human neutralizing antibodies against SARS-CoV-2 require intact Fc effector functions for optimal therapeutic protection. Cell 2021; 184:1804-1820.e16. [PMID: 33691139 PMCID: PMC7879018 DOI: 10.1016/j.cell.2021.02.026] [Citation(s) in RCA: 264] [Impact Index Per Article: 88.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/17/2020] [Accepted: 02/05/2021] [Indexed: 02/06/2023]
Abstract
SARS-CoV-2 has caused the global COVID-19 pandemic. Although passively delivered neutralizing antibodies against SARS-CoV-2 show promise in clinical trials, their mechanism of action in vivo is incompletely understood. Here, we define correlates of protection of neutralizing human monoclonal antibodies (mAbs) in SARS-CoV-2-infected animals. Whereas Fc effector functions are dispensable when representative neutralizing mAbs are administered as prophylaxis, they are required for optimal protection as therapy. When given after infection, intact mAbs reduce SARS-CoV-2 burden and lung disease in mice and hamsters better than loss-of-function Fc variant mAbs. Fc engagement of neutralizing antibodies mitigates inflammation and improves respiratory mechanics, and transcriptional profiling suggests these phenotypes are associated with diminished innate immune signaling and preserved tissue repair. Immune cell depletions establish that neutralizing mAbs require monocytes and CD8+ T cells for optimal clinical and virological benefit. Thus, potently neutralizing mAbs utilize Fc effector functions during therapy to mitigate lung infection and disease.
Collapse
Affiliation(s)
- Emma S Winkler
- Department of Medicine, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Pavlo Gilchuk
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jinsheng Yu
- Department of Genetics, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Adam L Bailey
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Rita E Chen
- Department of Medicine, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Zhenlu Chong
- Department of Medicine, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Seth J Zost
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Hyesun Jang
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30605, USA
| | - Ying Huang
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30605, USA
| | - James D Allen
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30605, USA
| | - James Brett Case
- Department of Medicine, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Rachel E Sutton
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Robert H Carnahan
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Tamarand L Darling
- Department of Medicine, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Adrianus C M Boon
- Department of Medicine, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Matthias Mack
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Richard D Head
- Department of Genetics, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30605, USA
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
29
|
Case JB, Chen RE, Cao L, Ying B, Winkler ES, Goreshnik I, Shrihari S, Kafai NM, Bailey AL, Xie X, Shi PY, Ravichandran R, Carter L, Stewart L, Baker D, Diamond MS. Ultrapotent miniproteins targeting the receptor-binding domain protect against SARS-CoV-2 infection and disease in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.03.01.433110. [PMID: 33688650 PMCID: PMC7941621 DOI: 10.1101/2021.03.01.433110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Despite the introduction of public health measures and spike protein-based vaccines to mitigate the COVID-19 pandemic, SARS-CoV-2 infections and deaths continue to rise. Previously, we used a structural design approach to develop picomolar range miniproteins targeting the SARS-CoV-2 receptor binding domain. Here, we investigated the capacity of modified versions of one lead binder, LCB1, to protect against SARS-CoV-2-mediated lung disease in human ACE2-expressing transgenic mice. Systemic administration of LCB1-Fc reduced viral burden, diminished immune cell infiltration and inflammation, and completely prevented lung disease and pathology. A single intranasal dose of LCB1v1.3 reduced SARS-CoV-2 infection in the lung even when given as many as five days before or two days after virus inoculation. Importantly, LCB1v1.3 protected in vivo against a historical strain (WA1/2020), an emerging B.1.1.7 strain, and a strain encoding key E484K and N501Y spike protein substitutions. These data support development of LCB1v1.3 for prevention or treatment of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- James Brett Case
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Rita E. Chen
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Longxing Cao
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Baoling Ying
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Emma S. Winkler
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Inna Goreshnik
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Swathi Shrihari
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Natasha M. Kafai
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Adam L. Bailey
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Xuping Xie
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston TX, USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Rashmi Ravichandran
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Lauren Carter
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Lance Stewart
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Michael S. Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
30
|
Khan A, Markus A, Rittmann T, Albers J, Alves F, Hülsmann S, Dullin C. Simple low dose radiography allows precise lung volume assessment in mice. Sci Rep 2021; 11:4163. [PMID: 33602964 PMCID: PMC7893164 DOI: 10.1038/s41598-021-83319-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/08/2021] [Indexed: 02/07/2023] Open
Abstract
X-ray based lung function (XLF) as a planar method uses dramatically less X-ray dose than computed tomography (CT) but so far lacked the ability to relate its parameters to pulmonary air volume. The purpose of this study was to calibrate the functional constituents of XLF that are biomedically decipherable and directly comparable to that of micro-CT and whole-body plethysmography (WBP). Here, we developed a unique set-up for simultaneous assessment of lung function and volume using XLF, micro-CT and WBP on healthy mice. Our results reveal a strong correlation of lung volumes obtained from radiographic XLF and micro-CT and demonstrate that XLF is superior to WBP in sensitivity and precision to assess lung volumes. Importantly, XLF measurement uses only a fraction of the radiation dose and acquisition time required for CT. Therefore, the redefined XLF approach is a promising tool for preclinical longitudinal studies with a substantial potential of clinical translation.
Collapse
Affiliation(s)
- Amara Khan
- Translational Molecular Imaging, Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Straße 3, 37075, Göttingen, Germany
| | - Andrea Markus
- Translational Molecular Imaging, Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Straße 3, 37075, Göttingen, Germany
| | - Thomas Rittmann
- 4th Physical Institute - Solids and Nanostructures, University of Göttingen, Friedrich-Hund-Platz 1, 37077, Göttingen, Germany
| | - Jonas Albers
- Translational Molecular Imaging, Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Straße 3, 37075, Göttingen, Germany
- Institute for Diagnostic and Interventional Radiology, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
| | - Frauke Alves
- Translational Molecular Imaging, Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Straße 3, 37075, Göttingen, Germany
- Institute for Diagnostic and Interventional Radiology, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany
- Clinic for Hematology and Medical Oncology, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Swen Hülsmann
- Clinic for Anesthesiology, University Medical Center Göttingen, Humboldtallee 23, 37073, Göttingen, Germany
| | - Christian Dullin
- Translational Molecular Imaging, Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Straße 3, 37075, Göttingen, Germany.
- Institute for Diagnostic and Interventional Radiology, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075, Göttingen, Germany.
| |
Collapse
|
31
|
Ariyoshi T, Tezuka J, Yasudo H, Sakata Y, Nakamura T, Matsushige T, Hasegawa H, Nakajima N, Ainai A, Oga A, Itoh H, Shirabe K, Toda S, Atsuta R, Ohga S, Hasegawa S. Enhanced airway hyperresponsiveness in asthmatic children and mice with A(H1N1)pdm09 infection. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:457-465. [PMID: 33470564 PMCID: PMC8127572 DOI: 10.1002/iid3.406] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/27/2020] [Accepted: 12/28/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Severe asthma exacerbation is an important comorbidity of the 2009 HIN1 pandemic (A(H1N1)pdm09) in asthmatic patients. However, the mechanisms underlying severe asthma exacerbation remain unknown. In this study, airway hyperresponsiveness (AHR) was measured in pediatric asthma patients infected with A(H1N1)pdm09. We also evaluated AHR in asthmatic mice with A(H1N1)pdm09 infection and those with seasonal influenza for comparison. METHODS AHRs in asthmatic children were defined as the provocative acetylcholine concentration causing a 20% reduction in forced expiratory volume in 1 s (PC20 ). To investigate the pathophysiology using animal models, BALB/c mice aged 6-8 weeks were sensitized and challenged with ovalbumin. Either mouse-adapted A(H1N1)pdm09, seasonal H1N1 virus (1 × 105 pfu/20 μl), or mock treatment as a control was administered intranasally. At 3, 7, and 10 days after infection, each group of mice was evaluated for AHR by methacholine challenge using an animal ventilator, flexiVent. Lung samples were resected and observed using light microscopy to assess the degree of airway inflammation. RESULTS AHRs in the children with bronchial asthma were temporarily increased, and alleviated by 3 months after discharge. AHR was significantly enhanced in A(H1N1)pdm09-infected asthmatic mice compared to that in seasonal H1N1-infected mice (p < .001), peaking at 7 days postinfection and then becoming similar to control levels by 10 days postinfection. Histopathological examination of lung tissues showed more intense infiltration of inflammatory cells and severe tissue destruction in A(H1N1)pdm09-infected mice at 7 days postinfection than at 10 days postinfection. CONCLUSION Our results suggest that enhanced AHR could contribute to severe exacerbation in human asthmatic patients with A(H1N1)pdm09 infection.
Collapse
Affiliation(s)
- Taira Ariyoshi
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Junichiro Tezuka
- Department of Allergy and Pulmonology, Fukuoka Children's Hospital, Fukuoka, Japan.,Department of Pediatrics, National Hospital Organization Fukuokahigashi Medical Center, Fukuoka, Japan
| | - Hiroki Yasudo
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Yasufumi Sakata
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Tamaki Nakamura
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Takeshi Matsushige
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hideki Hasegawa
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Noriko Nakajima
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Akira Ainai
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Atsunori Oga
- Department of Molecular Pathology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hiroshi Itoh
- Department of Molecular Pathology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Komei Shirabe
- Yamaguchi Prefectural Institute of Public Health and Environment, Yamaguchi, Japan
| | - Shoichi Toda
- Yamaguchi Prefectural Institute of Public Health and Environment, Yamaguchi, Japan
| | | | - Shouichi Ohga
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan.,Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shunji Hasegawa
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| |
Collapse
|
32
|
Winkler ES, Gilchuk P, Yu J, Bailey AL, Chen RE, Zost SJ, Jang H, Huang Y, Allen JD, Case JB, Sutton RE, Carnahan RH, Darling TL, Boon ACM, Mack M, Head RD, Ross TM, Crowe JE, Diamond MS. Human neutralizing antibodies against SARS-CoV-2 require intact Fc effector functions and monocytes for optimal therapeutic protection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 33398272 DOI: 10.1101/2020.12.28.424554] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SARS-CoV-2 has caused the global COVID-19 pandemic. Although passively delivered neutralizing antibodies against SARS-CoV-2 show promise in clinical trials, their mechanism of action in vivo is incompletely understood. Here, we define correlates of protection of neutralizing human monoclonal antibodies (mAbs) in SARS-CoV-2-infected animals. Whereas Fc effector functions are dispensable when representative neutralizing mAbs are administered as prophylaxis, they are required for optimal protection as therapy. When given after infection, intact mAbs reduce SARS-CoV-2 burden and lung disease in mice and hamsters better than loss-of-function Fc variant mAbs. Fc engagement of neutralizing antibodies mitigates inflammation and improves respiratory mechanics, and transcriptional profiling suggests these phenotypes are associated with diminished innate immune signaling and preserved tissue repair. Immune cell depletions establish that neutralizing mAbs require monocytes for therapeutic efficacy. Thus, potently neutralizing mAbs require Fc effector functions for maximal therapeutic benefit during therapy to modulate protective immune responses and mitigate lung disease.
Collapse
|
33
|
Xu C, Zhao J, Li Q, Hou L, Wang Y, Li S, Jiang F, Zhu Z, Tian L. Exosomes derived from three-dimensional cultured human umbilical cord mesenchymal stem cells ameliorate pulmonary fibrosis in a mouse silicosis model. Stem Cell Res Ther 2020; 11:503. [PMID: 33239075 PMCID: PMC7687745 DOI: 10.1186/s13287-020-02023-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023] Open
Abstract
Background Silicosis is an occupational respiratory disease caused by long-term excessive silica inhalation, which is most commonly encountered in industrial settings. Unfortunately, there is no effective therapy to delay and cure the progress of silicosis. In the recent years, stem cell therapy has emerged as an attractive tool against pulmonary fibrosis (PF) owing to its unique biological characteristics. However, the direct use of stem cells remains limitation by many risk factors for therapeutic purposes. The exclusive utility of exosomes secreted from stem cells, rather than cells, has been considered a promising alternative to overcome the limitations of cell-based therapy while maintaining its advantages. Methods and results In this study, we first employed a three-dimensional (3D) dynamic system to culture human umbilical cord mesenchymal stem cell (hucMSC) spheroids in a microcarrier suspension to yield exosomes from serum-free media. Experimental silicosis was induced in C57BL/6J mice by intratracheal instillation of a silica suspension, with/without exosomes derived from hucMSC (hucMSC-Exos), injection via the tail vein afterwards. The results showed that the gene expression of collagen I (COL1A1) and fibronectin (FN) was upregulated in the silica group as compared to that in the control group; however, this change decreased with hucMSC-Exo treatment. The value of FEV0.1 decreased in the silica group as compared to that in the control group, and this change diminished with hucMSC-Exo treatment. These findings suggested that hucMSC-Exos could inhibit silica-induced PF and regulate pulmonary function. We also performed in vitro experiments to confirm these findings; the results revealed that hucMSC-Exos decreased collagen deposition in NIH-3T3 cells exposed to silica. Conclusions Taken together, these studies support a potential role for hucMSC-Exos in ameliorating pulmonary fibrosis and provide new evidence for improving clinical treatment induced by silica. Supplementary information The online version contains supplementary material available at 10.1186/s13287-020-02023-9.
Collapse
Affiliation(s)
- Chunjie Xu
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xitoutiao Youanmen Street, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Jing Zhao
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xitoutiao Youanmen Street, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Qiuyue Li
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xitoutiao Youanmen Street, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Lin Hou
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xitoutiao Youanmen Street, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Yan Wang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xitoutiao Youanmen Street, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Siling Li
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xitoutiao Youanmen Street, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Fuyang Jiang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xitoutiao Youanmen Street, Beijing, 100069, China.,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Zhonghui Zhu
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xitoutiao Youanmen Street, Beijing, 100069, China. .,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| | - Lin Tian
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xitoutiao Youanmen Street, Beijing, 100069, China. .,Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
34
|
Biseugenol Exhibited Anti-Inflammatory and Anti-Asthmatic Effects in an Asthma Mouse Model of Mixed-Granulocytic Asthma. Molecules 2020; 25:molecules25225384. [PMID: 33217892 PMCID: PMC7698799 DOI: 10.3390/molecules25225384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 11/16/2022] Open
Abstract
In the present work, the anti-inflammatory and antiasthmatic potential of biseugenol, isolated as the main component from n-hexane extract from leaves of Nectandra leucantha and chemically prepared using oxidative coupling from eugenol, was evaluated in an experimental model of mixed-granulocytic asthma. Initially, in silico studies of biseugenol showed good predictions for drug-likeness, with adherence to Lipinski’s rules of five (RO5), good Absorption, Distribution, Metabolism and Excretion (ADME) properties and no alerts for Pan-Assay Interference Compounds (PAINS), indicating adequate adherence to perform in vivo assays. Biseugenol (20 mg·kg−1) was thus administered intraperitoneally (four days of treatment) and resulted in a significant reduction in both eosinophils and neutrophils of bronchoalveolar lavage fluid in ovalbumin-sensitized mice with no statistical difference from dexamethasone (5 mg·kg−1). As for lung function parameters, biseugenol (20 mg·kg−1) significantly reduced airway and tissue damping in comparison to ovalbumin group, with similar efficacy to positive control dexamethasone. Airway hyperresponsiveness to intravenous methacholine was reduced with biseugenol but was inferior to dexamethasone in higher doses. In conclusion, biseugenol displayed antiasthmatic effects, as observed through the reduction of inflammation and airway hyperresponsiveness, with similar effects to dexamethasone, on mixed-granulocytic ovalbumin-sensitized mice.
Collapse
|
35
|
Haider SH, Veerappan A, Crowley G, Caraher EJ, Ostrofsky D, Mikhail M, Lam R, Wang Y, Sunseri M, Kwon S, Prezant DJ, Liu M, Schmidt AM, Nolan A. Multiomics of World Trade Center Particulate Matter-induced Persistent Airway Hyperreactivity. Role of Receptor for Advanced Glycation End Products. Am J Respir Cell Mol Biol 2020; 63:219-233. [PMID: 32315541 DOI: 10.1165/rcmb.2019-0064oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pulmonary disease after World Trade Center particulate matter (WTC-PM) exposure is associated with dyslipidemia and the receptor for advanced glycation end products (RAGE); however, the mechanisms are not well understood. We used a murine model and a multiomics assessment to understand the role of RAGE in the pulmonary long-term effects of a single high-intensity exposure to WTC-PM. After 1 month, WTC-PM-exposed wild-type (WT) mice had airway hyperreactivity, whereas RAGE-deficient (Ager-/-) mice were protected. PM-exposed WT mice also had histologic evidence of airspace disease, whereas Ager-/- mice remained unchanged. Inflammatory mediators such as G-CSF (granulocyte colony-stimulating factor), IP-10 (IFN-γ-induced protein 10), and KC (keratinocyte chemoattractant) were differentially expressed after WTC-PM exposure. WTC-PM induced α-SMA, DIAPH1 (protein diaphanous homolog 1), RAGE, and significant lung collagen deposition in WT compared with Ager-/- mice. Compared with WT mice with PM exposure, relative expression of phosphorylated to total CREB (cAMP response element-binding protein) and JNK (c-Jun N-terminal kinase) was significantly increased in the lung of PM-exposed Ager-/- mice, whereas Akt (protein kinase B) was decreased. Random forests of the refined lung metabolomic profile classified subjects with 92% accuracy; principal component analysis captured 86.7% of the variance in three components and demonstrated prominent subpathway involvement, including known mediators of lung disease such as vitamin B6 metabolites, sphingolipids, fatty acids, and phosphatidylcholines. Treatment with a partial RAGE antagonist, pioglitazone, yielded similar fold-change expression of metabolites (N6-carboxymethyllysine, 1-methylnicotinamide, N1+N8-acetylspermidine, and succinylcarnitine [C4-DC]) between WT and Ager-/- mice exposed to WTC-PM. RAGE can mediate WTC-PM-induced airway hyperreactivity and warrants further investigation.
Collapse
Affiliation(s)
- Syed H Haider
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - Arul Veerappan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - George Crowley
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - Erin J Caraher
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - Dean Ostrofsky
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - Mena Mikhail
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - Rachel Lam
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - Yuyan Wang
- Division of Biostatistics, Department of Population Health
| | - Maria Sunseri
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - Sophia Kwon
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine
| | - David J Prezant
- Bureau of Health Services and Office of Medical Affairs, Fire Department of New York, Brooklyn, New York; and.,Division of Pulmonary Medicine, Department of Medicine, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, New York
| | - Mengling Liu
- Division of Biostatistics, Department of Population Health.,Department of Environmental Medicine, and
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, New York
| | - Anna Nolan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine.,Department of Environmental Medicine, and.,Bureau of Health Services and Office of Medical Affairs, Fire Department of New York, Brooklyn, New York; and
| |
Collapse
|
36
|
Alessandrini F, Musiol S, Schneider E, Blanco-Pérez F, Albrecht M. Mimicking Antigen-Driven Asthma in Rodent Models-How Close Can We Get? Front Immunol 2020; 11:575936. [PMID: 33101301 PMCID: PMC7555606 DOI: 10.3389/fimmu.2020.575936] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/31/2020] [Indexed: 12/22/2022] Open
Abstract
Asthma is a heterogeneous disease with increasing prevalence worldwide characterized by chronic airway inflammation, increased mucus secretion and bronchial hyperresponsiveness. The phenotypic heterogeneity among asthmatic patients is accompanied by different endotypes, mainly Type 2 or non-Type 2. To investigate the pathomechanism of this complex disease many animal models have been developed, each trying to mimic specific aspects of the human disease. Rodents have classically been employed in animal models of asthma. The present review provides an overview of currently used Type 2 vs. non-Type 2 rodent asthma models, both acute and chronic. It further assesses the methods used to simulate disease development and exacerbations as well as to quantify allergic airway inflammation, including lung physiologic, cellular and molecular immunologic responses. Furthermore, the employment of genetically modified animals, which provide an in-depth understanding of the role of a variety of molecules, signaling pathways and receptors implicated in the development of this disease as well as humanized models of allergic inflammation, which have been recently developed to overcome differences between the rodent and human immune systems, are discussed. Nevertheless, differences between mice and humans should be carefully considered and limits of extrapolation should be wisely taken into account when translating experimental results into clinical use.
Collapse
Affiliation(s)
- Francesca Alessandrini
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Stephanie Musiol
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Evelyn Schneider
- Center of Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Frank Blanco-Pérez
- Molecular Allergology/Vice President's Research Group, Paul-Ehrlich-Institut, Langen, Germany
| | - Melanie Albrecht
- Molecular Allergology/Vice President's Research Group, Paul-Ehrlich-Institut, Langen, Germany
| |
Collapse
|
37
|
Prada-Dacasa P, Urpi A, Sánchez-Benito L, Bianchi P, Quintana A. Measuring Breathing Patterns in Mice Using Whole-body Plethysmography. Bio Protoc 2020; 10:e3741. [PMID: 33659401 DOI: 10.21769/bioprotoc.3741] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 07/23/2020] [Accepted: 07/27/2020] [Indexed: 11/02/2022] Open
Abstract
Respiratory dysfunction is among the main cause of severe and fatal pathologies worldwide. The use of effective experimental models and methodologies for the study of the pulmonary pathophysiology is necessary to prevent, control and cure these diseases. Plethysmography, a technique for the assessment of lung function, has been widely applied in mice for the characterization of respiratory physiology. However, classical plethysmography methods present technical limitations such as the use of anesthesia and animal immobilization. Whole-body plethysmography (WBP) avoids these issues providing a non-invasive approach for the assessment of the respiratory function in conscious animals. WBP relies on the recording of pressure changes that are produced by the spontaneous breathing activity of an animal placed inside an airtight chamber. During normal respiration, pressure variation is directly proportional to the respiratory pattern of the animal allowing the measurement of the respiratory rate and tidal volume. These parameters are commonly used to evaluate pulmonary function in different physiological and disease models. In contrast to classical plethysmography methods, WBP technique allows reproducible serial measurements as it avoids animal restraint or the use of anesthesia. These key features rend WBP a suitable approach for longitudinal studies allowing the assessment of progressive respiratory alterations in physiological and pathological conditions. This protocol describes the procedures for the measurement of the breathing patterns in mice using the WBP method, the data analysis and results interpretation.
Collapse
Affiliation(s)
- Patricia Prada-Dacasa
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Andrea Urpi
- Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Laura Sánchez-Benito
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Patrizia Bianchi
- Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Albert Quintana
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.,Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
38
|
Szafran BN, Pinkston R, Perveen Z, Ross MK, Morgan T, Paulsen DB, Penn AL, Kaplan BLF, Noël A. Electronic-Cigarette Vehicles and Flavoring Affect Lung Function and Immune Responses in a Murine Model. Int J Mol Sci 2020; 21:E6022. [PMID: 32825651 PMCID: PMC7504509 DOI: 10.3390/ijms21176022] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/15/2020] [Accepted: 08/19/2020] [Indexed: 12/17/2022] Open
Abstract
The use of electronic nicotine delivery systems (ENDS), also known as electronic-cigarettes (e-cigs), has raised serious public health concerns, especially in light of the 2019 outbreak of e-cig or vaping product use-associated acute lung injury (EVALI). While these cases have mostly been linked to ENDS that contain vitamin E acetate, there is limited research that has focused on the chronic pulmonary effects of the delivery vehicles (i.e., without nicotine and flavoring). Thus, we investigated lung function and immune responses in a mouse model following exposure to the nearly ubiquitous e-cig delivery vehicles, vegetable glycerin (VG) and propylene glycol (PG), used with a specific 70%/30% ratio, with or without vanilla flavoring. We hypothesized that mice exposed sub-acutely to these e-cig aerosols would exhibit lung inflammation and altered lung function. Adult female C57BL/6 mice (n = 11-12 per group) were exposed to filtered air, 70%/30% VG/PG, or 70%/30% VG/PG with a French vanilla flavoring for 2 h a day for 6 weeks. Prior to sacrifice, lung function was assessed. At sacrifice, broncho-alveolar lavage fluid and lung tissue were collected for lipid mediator analysis, flow cytometry, histopathology, and gene expression analyses. Exposures to VG/PG + vanilla e-cig aerosol increased lung tidal and minute volumes and tissue damping. Immunophenotyping of lung immune cells revealed an increased number of dendritic cells, CD4+ T cells, and CD19+ B cells in the VG/PG-exposed group compared to air, irrespective of the presence of vanilla flavoring. Quantification of bioactive lung lipids demonstrated a >3-fold increase of 2-arachidonoylglycerol (2-AG), an anti-inflammatory mediator, and a 2-fold increase of 12-hydroxyeicosatetraenoic acid (12-HETE), another inflammatory mediator, following VG/PG exposure, with or without vanilla flavoring. This suggests that e-cig aerosol vehicles may affect immunoregulatory molecules. We also found that the two e-cig aerosols dysregulated the expression of lung genes. Ingenuity Pathway Analysis revealed that the gene networks that are dysregulated by the VG/PG e-cig aerosol are associated with metabolism of cellular proteins and lipids. Overall, our findings demonstrate that VG and PG, the main constituents of e-liquid formulations, when aerosolized through an e-cig device, are not harmless to the lungs, since they disrupt immune homeostasis.
Collapse
Affiliation(s)
- Brittany N. Szafran
- Center for Environmental Health Sciences, Department of Basic Sciences, Mississippi State University College of Veterinary Medicine, Mississippi State, MS 39762, USA; (B.N.S.); (M.K.R.); (B.L.F.K.)
| | - Rakeysha Pinkston
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; (R.P.); (Z.P.); (A.L.P.)
- Department of Environmental Toxicology, Southern University, Baton Rouge, LA 70803, USA
| | - Zakia Perveen
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; (R.P.); (Z.P.); (A.L.P.)
| | - Matthew K. Ross
- Center for Environmental Health Sciences, Department of Basic Sciences, Mississippi State University College of Veterinary Medicine, Mississippi State, MS 39762, USA; (B.N.S.); (M.K.R.); (B.L.F.K.)
| | - Timothy Morgan
- Department of Pathobiology and Population Medicine, Mississippi State University College of Veterinary Medicine, Mississippi State, MS 39762, USA;
| | - Daniel B. Paulsen
- Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA;
| | - Arthur L. Penn
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; (R.P.); (Z.P.); (A.L.P.)
| | - Barbara L. F. Kaplan
- Center for Environmental Health Sciences, Department of Basic Sciences, Mississippi State University College of Veterinary Medicine, Mississippi State, MS 39762, USA; (B.N.S.); (M.K.R.); (B.L.F.K.)
| | - Alexandra Noël
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; (R.P.); (Z.P.); (A.L.P.)
| |
Collapse
|
39
|
Lee JU, Lee HJ, Kim JN, Kim MK, Kim SR, Chang HS, Park CS, Park JS. Effects of Ammonium Chloride on Ozone-induced Airway Inflammation: the Role of Slc26a4 in the Lungs of Mice. J Korean Med Sci 2020; 35:e272. [PMID: 32808511 PMCID: PMC7431289 DOI: 10.3346/jkms.2020.35.e272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 06/24/2020] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Exposure to ozone (O₃) induces neutrophilic inflammation and goblet cell hyperplasia in humans and experimental animals. Because the solute carrier family 26-member 4 (Slc26a4; pendrin) gene induces mucin production and intraluminal acidification in the airways, it was hypothesized to be a key molecule in O₃-induced airway injury. Thus, we evaluated the role of Slc26a4 and the protective effects of ammonium chloride (NH₄Cl) in O₃-induced airway injury in mice. METHODS Six-week-old female BALB/c mice were exposed to filtered air or O₃ for 21 days (2 ppm for 3 hr/day). NH₄Cl (0, 0.1, 1, and 10 mM) was administered intratracheally into the airways. Airway resistance was measured using a flexiVent system, and bronchoalveolar lavage fluid (BALF) cells were differentially counted. Slc26a4 and Muc5ac proteins and mRNA were measured via western blotting, real-time polymerase chain reaction, and immunostaining. Tumor necrosis factor (TNF)-α, interferon (IFN)-γ, interleukin (IL)-17, IL-1β, and caspase-1 were analyzed via western blotting. RESULTS The levels Slc26a4 protein and mRNA significantly increased in lung tissues from Day 7 to Day 21 of O₃ exposure, with concomitant increases in lung resistance, numbers of goblet cells in lung tissues, and inflammatory cells and thiocyanate (SCN-) levels in BALF in a time-dependent manner. Treatment with NH₄Cl significantly reduced these changes to levels similar to those of sham-treated mice, with a concomitant reduction of Slc26a4 proteins in lung lysates and SCN- levels in BALF. Slc26a4 protein was co-expressed with muc5ac protein in the bronchial epithelium, as indicated by immunofluorescence staining. NH₄Cl treatment also significantly attenuated the O₃-induced increases in IFN-γ, TNF-α, IL-17, IL-1β, and p20-activated caspase-1. CONCLUSION Slc26a4 may be involved in O₃-induced inflammatory and epithelial changes in the airways via activation of the inflammasome and the induction of IL-17 and IFN-γ. NH₄Cl shows a potential as a therapeutic agent for controlling O₃-induced airway inflammation and epithelial damage by modulating Slc26a4 expression.
Collapse
Affiliation(s)
- Jong Uk Lee
- Division of Allergy and Respiratory Diseases, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Hyeon Ju Lee
- Division of Allergy and Respiratory Diseases, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Ji Na Kim
- Division of Allergy and Respiratory Diseases, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Min Kyung Kim
- Division of Allergy and Respiratory Diseases, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Sung Roul Kim
- Department of Environmental Health Sciences, Soonchunhyang University, Asan, Korea
| | - Hun Soo Chang
- Division of Allergy and Respiratory Diseases, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
- Soonchunhyang Medical Research Institute, Soonchunhyang University College of Medicine, Cheonan, Korea
| | - Choon Sik Park
- Division of Allergy and Respiratory Diseases, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Jong Sook Park
- Division of Allergy and Respiratory Diseases, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea.
| |
Collapse
|
40
|
Martinez ME, Harder OE, Rosas LE, Joseph L, Davis IC, Niewiesk S. Pulmonary function analysis in cotton rats after respiratory syncytial virus infection. PLoS One 2020; 15:e0237404. [PMID: 32776985 PMCID: PMC7416943 DOI: 10.1371/journal.pone.0237404] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/25/2020] [Indexed: 01/31/2023] Open
Abstract
The cotton rat (Sigmodon hispidus) is an excellent small animal model for human respiratory viral infections such as human respiratory syncytial virus (RSV) and human metapneumovirus (HMPV). These respiratory viral infections, as well as other pulmonary inflammatory diseases such as asthma, are associated with lung mechanic disturbances. So far, the pathophysiological effects of viral infection and allergy on cotton rat lungs have not been measured, although this information might be an important tool to determine the efficacy of vaccine and drug candidates. To characterize pulmonary function in the cotton rat, we established forced oscillation technique in uninfected, RSV infected and HDM sensitized cotton rats, and characterized pulmonary inflammation, mucus production, pulmonary edema, and oxygenation. There was a gender difference after RSV infection, with females demonstrating airway hyper-responsiveness while males did not. Female cotton rats 2dpi had a mild increase in pulmonary edema (wet: dry weight ratios). At day 4 post infection, female cotton rats demonstrated mild pulmonary inflammation, no increase in mucus production or reduction in oxygenation. Pulmonary function was not significantly impaired after RSV infection. In contrast, cotton rats sensitized to HDM demonstrated airway hyper-responsiveness with a significant increase in pulmonary inflammation, increase in baseline tissue damping, and a decrease in baseline pulmonary compliance. In summary, we established baseline data for forced oscillation technique and other respiratory measures in the cotton rat and used it to analyze respiratory diseases in cotton rats.
Collapse
Affiliation(s)
- Margaret E. Martinez
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Olivia E. Harder
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Lucia E. Rosas
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Lisa Joseph
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Ian C. Davis
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Stefan Niewiesk
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
41
|
Winkler ES, Bailey AL, Kafai NM, Nair S, McCune BT, Yu J, Fox JM, Chen RE, Earnest JT, Keeler SP, Ritter JH, Kang LI, Dort S, Robichaud A, Head R, Holtzman MJ, Diamond MS. SARS-CoV-2 infection in the lungs of human ACE2 transgenic mice causes severe inflammation, immune cell infiltration, and compromised respiratory function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32676600 DOI: 10.1101/2020.07.09.196188] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Severe Acute Respiratory Syndrome Coronavirus -2 (SARS-CoV-2) emerged in late 2019 and has spread worldwide resulting in the Coronavirus Disease 2019 (COVID-19) pandemic. Although animal models have been evaluated for SARS-CoV-2 infection, none have recapitulated the severe lung disease phenotypes seen in hospitalized human cases. Here, we evaluate heterozygous transgenic mice expressing the human ACE2 receptor driven by the epithelial cell cytokeratin-18 gene promoter (K18-hACE2) as a model of SARS-CoV-2 infection. Intranasal inoculation of SARS-CoV-2 in K18-hACE2 mice results in high levels of viral infection in lung tissues with additional spread to other organs. Remarkably, a decline in pulmonary function, as measured by static and dynamic tests of respiratory capacity, occurs 4 days after peak viral titer and correlates with an inflammatory response marked by infiltration into the lung of monocytes, neutrophils, and activated T cells resulting in pneumonia. Cytokine profiling and RNA sequencing analysis of SARS-CoV-2-infected lung tissues show a massively upregulated innate immune response with prominent signatures of NF-kB-dependent, type I and II interferon signaling, and leukocyte activation pathways. Thus, the K18-hACE2 model of SARS-CoV-2 infection recapitulates many features of severe COVID-19 infection in humans and can be used to define the mechanistic basis of lung disease and test immune and antiviral-based countermeasures.
Collapse
|
42
|
Crotoxin-Induced Mice Lung Impairment: Role of Nicotinic Acetylcholine Receptors and COX-Derived Prostanoids. Biomolecules 2020; 10:biom10050794. [PMID: 32443924 PMCID: PMC7277605 DOI: 10.3390/biom10050794] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/13/2020] [Accepted: 03/14/2020] [Indexed: 12/12/2022] Open
Abstract
Respiratory compromise in Crotalus durissus terrificus (C.d.t.) snakebite is an important pathological condition. Considering that crotoxin (CTX), a phospholipase A2 from C.d.t. venom, is the main component of the venom, the present work investigated the toxin effects on respiratory failure. Lung mechanics, morphology and soluble markers were evaluated from Swiss male mice, and mechanism determined using drugs/inhibitors of eicosanoids biosynthesis pathway and autonomic nervous system. Acute respiratory failure was observed, with an early phase (within 2 h) characterized by enhanced presence of eicosanoids, including prostaglandin E2, that accounted for the increased vascular permeability in the lung. The alterations of early phase were inhibited by indomethacin. The late phase (peaked 12 h) was marked by neutrophil infiltration, presence of pro-inflammatory cytokines/chemokines, and morphological alterations characterized by alveolar septal thickening and bronchoconstriction. In addition, lung mechanical function was impaired, with decreased lung compliance and inspiratory capacity. Hexamethonium, a nicotinic acetylcholine receptor antagonist, hampered late phase damages indicating that CTX-induced lung impairment could be associated with cholinergic transmission. The findings reported herein highlight the impact of CTX on respiratory compromise, and introduce the use of nicotinic blockers and prostanoids biosynthesis inhibitors as possible symptomatic therapy to Crotalus durissus terrificus snakebite.
Collapse
|
43
|
Park JK, Coffey NJ, Bodine SP, Zawatsky CN, Jay L, Gahl WA, Kunos G, Gochuico BR, Malicdan MCV, Cinar R. Bleomycin Induces Drug Efflux in Lungs. A Pitfall for Pharmacological Studies of Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2020; 62:178-190. [PMID: 31419911 DOI: 10.1165/rcmb.2018-0147oc] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
ATP-binding cassette (ABC) transporters are evolutionarily conserved membrane proteins that pump a variety of endogenous substrates across cell membranes. Certain subfamilies are known to interact with pharmaceutical compounds, potentially influencing drug delivery and treatment efficacy. However, the role of drug resistance-associated ABC transporters has not been examined in idiopathic pulmonary fibrosis (IPF) or its animal model: the bleomycin (BLM)-induced murine model. Here, we investigate the expression of two ABC transporters, P-gp (permeability glycoprotein) and BCRP (breast cancer resistance protein), in human IPF lung tissue and two different BLM-induced mouse models of pulmonary fibrosis. We obtained human IPF specimens from patients during lung transplantation and administered BLM to male C57BL/6J mice either by oropharyngeal aspiration (1 U/kg) or subcutaneous osmotic infusion (100 U/kg over 7 d). We report that P-gp and BCRP expression in lungs of patients with IPF was comparable to controls. However, murine lungs expressed increased levels of P-gp and BCRP after oropharyngeal and subcutaneous BLM administration. We localized this upregulation to multiple pulmonary cell types, including alveolar fibroblasts, endothelial cells, and type 2 epithelial cells. Functionally, this effect reduced murine lung exposure to nintedanib, a U.S. Food and Drug Administration-approved IPF therapy known to be a P-gp substrate. The study reveals a discrepancy between IPF pathophysiology and the common animal model of lung fibrosis. BLM-induced drug efflux in the murine lungs may present an uncontrolled confounding variable in the preclinical study of IPF drug candidates, and these findings will facilitate disease model validation and enhance new drug discoveries that will ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Joshua K Park
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland; and
| | - Nathan J Coffey
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland; and
| | - Steven P Bodine
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, and
| | - Charles N Zawatsky
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland; and
| | - Lindsey Jay
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland; and
| | - William A Gahl
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, and.,NIH Undiagnosed Diseases Program and Office of the Clinical Director, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - George Kunos
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland; and
| | - Bernadette R Gochuico
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, and
| | - May Christine V Malicdan
- Section of Human Biochemical Genetics, Medical Genetics Branch, National Human Genome Research Institute, and.,NIH Undiagnosed Diseases Program and Office of the Clinical Director, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Resat Cinar
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland; and
| |
Collapse
|
44
|
LaRivière WB, Liao S, McMurtry SA, Oshima K, Han X, Zhang F, Yan S, Haeger SM, Ransom M, Bastarache JA, Linhardt RJ, Schmidt EP, Yang Y. Alveolar heparan sulfate shedding impedes recovery from bleomycin-induced lung injury. Am J Physiol Lung Cell Mol Physiol 2020; 318:L1198-L1210. [PMID: 32320623 DOI: 10.1152/ajplung.00063.2020] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The pulmonary epithelial glycocalyx, an anionic cell surface layer enriched in glycosaminoglycans such as heparan sulfate and chondroitin sulfate, contributes to the alveolar barrier. Direct injury to the pulmonary epithelium induces shedding of heparan sulfate into the air space; the impact of this shedding on recovery after lung injury is unknown. Using mass spectrometry, we found that heparan sulfate was shed into the air space for up to 3 wk after intratracheal bleomycin-induced lung injury and coincided with induction of matrix metalloproteinases (MMPs), including MMP2. Delayed inhibition of metalloproteinases, beginning 7 days after bleomycin using the nonspecific MMP inhibitor doxycycline, attenuated heparan sulfate shedding and improved lung function, suggesting that heparan sulfate shedding may impair lung recovery. While we also observed an increase in air space heparanase activity after bleomycin, pharmacological and transgenic inhibition of heparanase in vivo failed to attenuate heparan sulfate shedding or protect against bleomycin-induced lung injury. However, experimental augmentation of airway heparanase activity significantly worsened post-bleomycin outcomes, confirming the importance of epithelial glycocalyx integrity to lung recovery. We hypothesized that MMP-associated heparan sulfate shedding contributed to delayed lung recovery, in part, by the release of large, highly sulfated fragments that sequestered lung-reparative growth factors such as hepatocyte growth factor. In vitro, heparan sulfate bound hepatocyte growth factor and attenuated growth factor signaling, suggesting that heparan sulfate shed into the air space after injury may directly impair lung repair. Accordingly, administration of exogenous heparan sulfate to mice after bleomycin injury increased the likelihood of death due to severe lung dysfunction. Together, our findings demonstrate that alveolar epithelial heparan sulfate shedding impedes lung recovery after bleomycin.
Collapse
Affiliation(s)
- W B LaRivière
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - S Liao
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - S A McMurtry
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - K Oshima
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - X Han
- Department of Chemistry, Rensselaer Polytechnic Institute, Troy, New York
| | - F Zhang
- Department of Chemistry, Rensselaer Polytechnic Institute, Troy, New York
| | - S Yan
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.,College of Life Sciences, Henan Normal University, Xinxiang, China
| | - S M Haeger
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - M Ransom
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - J A Bastarache
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - R J Linhardt
- Department of Chemistry, Rensselaer Polytechnic Institute, Troy, New York
| | - E P Schmidt
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.,Department of Medicine, Denver Health Medical Center, Denver, Colorado
| | - Y Yang
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
45
|
Dimori M, Heard-Lipsmeyer ME, Byrum SD, Mackintosh SG, Kurten RC, Carroll JL, Morello R. Respiratory defects in the CrtapKO mouse model of osteogenesis imperfecta. Am J Physiol Lung Cell Mol Physiol 2020; 318:L592-L605. [PMID: 32022592 PMCID: PMC7191481 DOI: 10.1152/ajplung.00313.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Respiratory disease is a leading cause of mortality in patients with osteogenesis imperfecta (OI), a connective tissue disease that causes severely reduced bone mass and is most commonly caused by dominant mutations in type I collagen genes. Previous studies proposed that impaired respiratory function in OI patients was secondary to skeletal deformities; however, recent evidence suggests the existence of a primary lung defect. Here, we analyzed the lung phenotype of Crtap knockout (KO) mice, a mouse model of recessive OI. While we confirm changes in the lung parenchyma that are reminiscent of emphysema, we show that CrtapKO lung fibroblasts synthesize type I collagen with altered posttranslation modifications consistent with those observed in bone and skin. Unrestrained whole body plethysmography showed a significant decrease in expiratory time, resulting in an increased ratio of inspiratory time over expiratory time and a concomitant increase of the inspiratory duty cycle in CrtapKO compared with WT mice. Closed-chest measurements using the forced oscillation technique showed increased respiratory system elastance, decreased respiratory system compliance, and increased tissue damping and elasticity in CrtapKO mice compared with WT. Pressure-volume curves showed significant differences in lung volumes and in the shape of the curves between CrtapKO mice and WT mice, with and without adjustment for body weight. This is the first evidence that collagen defects in OI cause primary changes in lung parenchyma and several respiratory parameters and thus negatively impact lung function.
Collapse
Affiliation(s)
- Milena Dimori
- Department of Physiology & Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Melissa E Heard-Lipsmeyer
- Department of Physiology & Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Arkansas Children's Research Institute, Little Rock, Arkansas
| | - Samuel G Mackintosh
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Richard C Kurten
- Department of Physiology & Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - John L Carroll
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Roy Morello
- Department of Physiology & Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Division of Genetics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
46
|
Noël A, Hansen S, Zaman A, Perveen Z, Pinkston R, Hossain E, Xiao R, Penn A. In utero exposures to electronic-cigarette aerosols impair the Wnt signaling during mouse lung development. Am J Physiol Lung Cell Mol Physiol 2020; 318:L705-L722. [PMID: 32083945 DOI: 10.1152/ajplung.00408.2019] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Currently, more than 9 million American adults, including women of childbearing age, use electronic-cigarettes (e-cigs). Further, the prevalence of maternal vaping now approaching 10% is similar to that of maternal smoking. Little, however, is known about the effects of fetal exposures to nicotine-rich e-cig aerosols on lung development. In this study, we assessed whether in utero exposures to e-cig aerosols compromised lung development in mice. A third-generation e-cig device was used to expose pregnant BALB/c mice by inhalation to 36 mg/mL of nicotine cinnamon-flavored e-cig aerosols for 14-31 days. This included exposures for either 12 days before mating plus during gestation (preconception groups) or only during gestation (prenatal groups). Respective control mice were exposed to filtered air. Subgroups of offspring were euthanized at birth or at 4 wk of age. Compared with respective air-exposed controls, both preconception and prenatal exposures to e-cig aerosols significantly decreased the offspring birth weight and body length. In the preconception group, 7 inflammation-related genes were downregulated, including 4 genes common to both dams and fetuses, denoting an e-cig immunosuppressive effect. Lung morphometry assessments of preconception e-cig-exposed offspring showed a significantly increased tissue fraction at birth. This result was supported by the downregulation of 75 lung genes involved in the Wnt signaling, which is essential to lung organogenesis. Thus, our data indicate that maternal vaping impairs pregnancy outcomes, alters fetal lung structure, and dysregulates the Wnt signaling. This study provides experimental evidence for future regulations of e-cig products for pregnant women and developmentally vulnerable populations.
Collapse
Affiliation(s)
- Alexandra Noël
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Shannon Hansen
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Anusha Zaman
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Zakia Perveen
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Rakeysha Pinkston
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana.,Health Research Center, Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, Louisiana
| | - Ekhtear Hossain
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Rui Xiao
- Department of Anesthesiology, Columbia University Medical Center, New York, New York
| | - Arthur Penn
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| |
Collapse
|
47
|
Abstract
There is a growing recognition that aging is a risk factor for fibrosis that affects a number of organ systems, including the lung. Despite this understanding, most studies of experimental fibrosis have been conducted in young mice that typically resolve injury-induced lung fibrosis over the course of several months. Our studies demonstrate that aged mouse models may recapitulate human disease by generating a more persistent fibrotic response to injury. This is, in part, due to an imbalance in the expression and activity of NADPH oxidase (NOX) enzymes, in particular the NOX4 isoform, and a related deficiency in antioxidant responses in pathogenic myofibroblasts. These pathogenic myofibroblasts acquire features of cellular senescence and become resistant to apoptosis. In this chapter, we present methods and procedures to apply the aging model of lung fibrosis in mice that will allow interrogation of myofibroblast functions and the expression and activity of NOX4 in cells. We provide recommendations for best laboratory practices to assess the severity and resolution of fibrosis in murine models of aging.
Collapse
Affiliation(s)
- Karen Bernard
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Victor J Thannickal
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
48
|
Shah D, Torres C, Bhandari V. Adiponectin deficiency induces mitochondrial dysfunction and promotes endothelial activation and pulmonary vascular injury. FASEB J 2019; 33:13617-13631. [PMID: 31585050 PMCID: PMC6894062 DOI: 10.1096/fj.201901123r] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 09/03/2019] [Indexed: 01/15/2023]
Abstract
Adiponectin (APN), an adipocyte-derived adipokine, has been shown to limit lung injury originating from endothelial cell (EC) damage. Previously we reported that obese mice with low circulatory APN levels exhibited pulmonary vascular endothelial dysfunction. This study was designed to investigate the cellular and molecular mechanisms underlying the pulmonary endothelium-dependent protective effects of APN. Our results demonstrated that in APN-/- mice, there was an inherent state of endothelium mitochondrial dysfunction that could contribute to endothelial activation and increased susceptibility to LPS-induced acute lung injury (ALI). We noted that APN-/- mice showed decreased expression of mitochondrial biogenesis regulatory protein peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α) and its downstream proteins nuclear respiratory factor 1, transcription factor A, mitochondrial, and Sirtuin (Sirt)3 and Sirt1 expression in whole lungs and in freshly isolated lung ECs from these mice at baseline and subjected to LPS-induced ALI. We further showed that treating APN-/- mice with PGC-1α activator pyrroloquinoline quinone enhances mitochondrial biogenesis and function in lung endothelium and attenuation of ALI. These results suggest that the pulmonary endothelium-protective properties of APN are mediated, at least in part, by an enhancement of mitochondrial biogenesis through a mechanism involving PGC-1α activation.-Shah, D., Torres, C., Bhandari, V. Adiponectin deficiency induces mitochondrial dysfunction and promotes endothelial activation and pulmonary vascular injury.
Collapse
Affiliation(s)
- Dilip Shah
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Claudio Torres
- Department of Neurobiology and Anatomy, Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Vineet Bhandari
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
49
|
Veldhuizen RAW, McCaig LA, Pape C, Gill SE. The effects of aging and exercise on lung mechanics, surfactant and alveolar macrophages. Exp Lung Res 2019; 45:113-122. [PMID: 31195852 DOI: 10.1080/01902148.2019.1605633] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Purpose: Advancing age leads to changes to the respiratory system associated with increased susceptibility to lung diseases, and exercise may counteract this effect. To explore the underlying processes, we investigated the effects of aging and exercise on lung mechanics, alveolar macrophage function, and surfactant pools and activity, in mice. It was hypothesized that aging would impact lung mechanics, macrophage polarization, and the status of the surfactant system, and that these changes would be mitigated by exercise. Methods: Male C57BL/6 mice were housed from 2-3 to 22 months, for the aged group, or until 4 months of age for young mice. Mice in both groups were randomized to voluntarily running exercise or to non-exercise, for a 2-month period. Mice were euthanized and lung mechanics were analyzed using a flexiVent ventilator. Subsequently, the lungs were lavaged to obtain pulmonary surfactant and alveolar macrophages. Pulmonary surfactant was analyzed for pool sizes and activity whereas alveolar macrophages were examined for response to pro and anti-inflammatory stimuli. Results: Changes in lung mechanics, such as increased compliance and decreased airway resistance, were associated with aging but were not affected by exercise. The quantity as well as the biophysical activity of the pulmonary surfactant system was unaffected by either aging or exercise. More alveolar macrophages were recovered from exercising aged mice compared to both the young and non-exercising groups. Macrophages in this aged exercise group were more responsive to an anti-inflammatory stimulus. Conclusions: Our data supports previous literature that suggest the development of emphysema-like alterations to lung mechanics with aging. This effect was independent of exercise. Our data also indicates that surfactant is unaffected by aging and exercise. Alveolar macrophage properties and numbers were affected by exercise in the aging lung and may represent the main, potentially beneficial, effect of exercise on the pulmonary system.
Collapse
Affiliation(s)
- Ruud A W Veldhuizen
- a Departments of Physiology & Pharmacology, and Medicine , The University of Western Ontario , London , Ontario , Canada.,b Centre for Critical Illness Research, Lawson Health Research Institute , London , Ontario , Canada
| | - Lynda A McCaig
- a Departments of Physiology & Pharmacology, and Medicine , The University of Western Ontario , London , Ontario , Canada.,b Centre for Critical Illness Research, Lawson Health Research Institute , London , Ontario , Canada
| | - Cynthia Pape
- a Departments of Physiology & Pharmacology, and Medicine , The University of Western Ontario , London , Ontario , Canada.,b Centre for Critical Illness Research, Lawson Health Research Institute , London , Ontario , Canada
| | - Sean E Gill
- a Departments of Physiology & Pharmacology, and Medicine , The University of Western Ontario , London , Ontario , Canada.,b Centre for Critical Illness Research, Lawson Health Research Institute , London , Ontario , Canada
| |
Collapse
|
50
|
Zhai J, Insel M, Addison KJ, Stern DA, Pederson W, Dy A, Rojas-Quintero J, Owen CA, Sherrill DL, Morgan W, Wright AL, Halonen M, Martinez FD, Kraft M, Guerra S, Ledford JG. Club Cell Secretory Protein Deficiency Leads to Altered Lung Function. Am J Respir Crit Care Med 2019; 199:302-312. [PMID: 30543455 PMCID: PMC6363971 DOI: 10.1164/rccm.201807-1345oc] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 12/07/2018] [Indexed: 01/27/2023] Open
Abstract
RATIONALE CC16 (club cell secretory protein-16), a member of the secretoglobin family, is one of the most abundant proteins in normal airway secretions and has been described as a serum biomarker for obstructive lung diseases. OBJECTIVES To determine whether low CC16 is a marker for airway pathology or is implicated in the pathophysiology of progressive airway damage in these conditions. METHODS Using human data from the birth cohort of the Tucson Children's Respiratory Study, we examined the relation of circulating CC16 levels with pulmonary function and responses to bronchial methacholine challenge from childhood up to age 32 years. In wild-type and CC16-/- mice, we set out to comprehensively examine pulmonary physiology, inflammation, and remodeling in the naive airway. MEASUREMENTS AND MAIN RESULTS We observed that Tucson Children's Respiratory Study participants in the lowest tertile of serum CC16 had significant deficits in their lung function and enhanced airway hyperresponsiveness to methacholine challenge from 11 years throughout young adult life. Similarly, CC16-/- mice had significant deficits in lung function and enhanced airway hyperresponsiveness to methacholine as compared with wild-type mice, which were independent of inflammation and mucin production. As compared with wild-type mice, CC16-/- mice had significantly elevated gene expression of procollagen type I, procollagen type III, and α-smooth muscle actin, areas of pronounced collagen deposition and significantly enhanced smooth muscle thickness. CONCLUSIONS Our findings support clinical observations by providing evidence that lack of CC16 in the lung results in dramatically altered pulmonary function and structural alterations consistent with enhanced remodeling.
Collapse
Affiliation(s)
- Jing Zhai
- Asthma and Airway Disease Research Center
| | | | | | | | | | | | | | - Caroline A. Owen
- Brigham and Women’s Hospital/Harvard Medical School, Boston, Massachusetts
| | | | | | | | | | | | - Monica Kraft
- Asthma and Airway Disease Research Center
- Department of Medicine, and
| | - Stefano Guerra
- Asthma and Airway Disease Research Center
- Department of Medicine, and
- ISGlobal, Barcelona, Spain
| | - Julie G. Ledford
- Asthma and Airway Disease Research Center
- Department of Medicine, and
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| |
Collapse
|