Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
JCI Insight ; 9(2)2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38061015

ABSTRACT

Infection of immature mice with rhinovirus (RV) induces an asthma-like phenotype consisting of type 2 inflammation, mucous metaplasia, eosinophilic inflammation, and airway hyperresponsiveness that is dependent on IL-25 and type 2 innate lymphoid cells (ILC2s). Doublecortin-like kinase 1-positive (DCLK1+) tuft cells are a major source of IL-25. We sought to determine the requirement of tuft cells for the RV-induced asthma phenotype in wild-type mice and mice deficient in Pou2f3, a transcription factor required for tuft cell development. C57BL/6J mice infected with RV-A1B on day 6 of life and RV-A2 on day 13 of life showed increased DCLK1+ tuft cells in the large airways. Compared with wild-type mice, RV-infected Pou2f3-/- mice showed reductions in IL-25 mRNA and protein expression, ILC2 expansion, type 2 cytokine expression, mucous metaplasia, lung eosinophils, and airway methacholine responsiveness. We conclude that airway tuft cells are required for the asthma phenotype observed in immature mice undergoing repeated RV infections. Furthermore, RV-induced tuft cell development provides a mechanism by which early-life viral infections could potentiate type 2 inflammatory responses to future infections.


Subject(s)
Asthma , Enterovirus Infections , Animals , Mice , Immunity, Innate , Rhinovirus , Tuft Cells , Lymphocytes/metabolism , Mice, Inbred C57BL , Asthma/metabolism , Inflammation , Phenotype , Metaplasia
2.
Front Immunol ; 13: 952509, 2022.
Article in English | MEDLINE | ID: mdl-36032072

ABSTRACT

Wheezing-associated rhinovirus (RV) infections are associated with asthma development. We have shown that infection of immature mice with RV induces type 2 cytokine production and mucous metaplasia which is dependent on IL-33 and type 2 innate lymphoid cells (ILC2s) and intensified by a second heterologous RV infection. We hypothesize that M2a macrophages are required for the exaggerated inflammation and mucous metaplasia in response to heterologous RV infection. Wild-type C57Bl/6J mice and LysMCre IL4Rα KO mice lacking M2a macrophages were treated as follows: (1) sham infection on day 6 of life plus sham on day 13 of life, (2) RV-A1B on day 6 plus sham on day 13, (3) sham on day 6 and RV-A2 on day 13, or (4) RV-A1B on day 6 and RV-A2 on day 13. Lungs were harvested one or seven days after the second infection. Wild-type mice infected with RV-A1B at day 6 showed an increased number of Arg1- and Retnla-expressing lung macrophages, indicative of M2a polarization. Compared to wild-type mice infected with RV on day 6 and 13 of life, the lungs of LysMCre IL4Rα KO mice undergoing heterologous RV infection showed decreased protein abundance of the epithelial-derived innate cytokines IL-33, IL-25 and TSLP, decreased ILC2s, decreased mRNA expression of IL-13 and IL-5, and decreased PAS staining. Finally, mRNA analysis and immunofluorescence microscopy of double-infected LysMCre IL4Rα KO mice showed reduced airway epithelial cell IL-33 expression, and treatment with IL-33 restored the exaggerated muco-inflammatory phenotype. Conclusion: Early-life RV infection alters the macrophage response to subsequent heterologous infection, permitting enhanced IL-33 expression, ILC2 expansion and intensified airway inflammation and mucous metaplasia.


Subject(s)
Interleukin-33 , Rhinovirus , Animals , Immunity, Innate , Inflammation , Lymphocytes , Macrophages , Metaplasia , Mice , RNA, Messenger
3.
Methods Mol Biol ; 2506: 43-56, 2022.
Article in English | MEDLINE | ID: mdl-35771462

ABSTRACT

Rhinovirus (RV) infection is linked to early life wheezing and exacerbation of adult asthma. RV infection can be modeled in adult and neonatal mice. This chapter outlines methods for the production of standardized human rhinovirus A1B and mouse infection. The chapter also describes methods to couple infections with allergen (ovalbumin and house dust mite) administrations. The production of the virus involves its amplification, purification, and concentration. In order to standardize the concentrated RV stock, a plaque assay on HeLa cells is outlined as a method of calibrating infectivity. Once the number of plaque-forming units is determined, the standardized virus is used for mouse infection.


Subject(s)
Asthma , Enterovirus Infections , Picornaviridae Infections , Animals , HeLa Cells , Humans , Mice , Pyroglyphidae , Rhinovirus
4.
Mucosal Immunol ; 14(6): 1369-1380, 2021 11.
Article in English | MEDLINE | ID: mdl-34354243

ABSTRACT

Compared to other RV species, RV-C has been associated with more severe respiratory illness and is more likely to occur in children with a history of asthma or who develop asthma. We therefore inoculated 6-day-old mice with sham, RV-A1B, or RV-C15. Inflammasome priming and activation were assessed, and selected mice treated with recombinant IL-1ß. Compared to RV-A1B infection, RV-C15 infection induced an exaggerated asthma phenotype, with increased mRNA expression of Il5, Il13, Il25, Il33, Muc5ac, Muc5b, and Clca1; increased lung lineage-negative CD25+CD127+ST2+ ILC2s; increased mucous metaplasia; and increased airway responsiveness. Lung vRNA, induction of pro-inflammatory type 1 cytokines, and inflammasome priming (pro-IL-1ß and NLRP3) were not different between the two viruses. However, inflammasome activation (mature IL-1ß and caspase-1 p12) was reduced in RV-C15-infected mice compared to RV-A1B-infected mice. A similar deficiency was found in cultured macrophages. Finally, IL-1ß treatment decreased RV-C-induced type 2 cytokine and mucus-related gene expression, ILC2s, mucous metaplasia, and airway responsiveness but not lung vRNA level. We conclude that RV-C induces an enhanced asthma phenotype in immature mice. Compared to RV-A, RV-C-induced macrophage inflammasome activation and IL-1ß are deficient, permitting exaggerated type 2 inflammation and mucous metaplasia.


Subject(s)
Asthma/etiology , Asthma/metabolism , Coxsackievirus Infections/complications , Coxsackievirus Infections/virology , Enterovirus , Inflammasomes/metabolism , Phenotype , Animals , Asthma/diagnosis , Biomarkers , Cell Line , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Disease Susceptibility , Enterovirus/physiology , Humans , Immunity, Innate , Immunophenotyping , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice
5.
Front Immunol ; 12: 649520, 2021.
Article in English | MEDLINE | ID: mdl-33968043

ABSTRACT

Rhinovirus C (RV-C) infection is associated with severe asthma exacerbations. Since type 2 inflammation is an important disease mechanism in asthma, we hypothesized that RV-C infection, in contrast to RV-A, preferentially stimulates type 2 inflammation, leading to exacerbated eosinophilic inflammation. To test this, we developed a mouse model of RV-C15 airways disease. RV-C15 was generated from the full-length cDNA clone and grown in HeLa-E8 cells expressing human CDHR3. BALB/c mice were inoculated intranasally with 5 x 106 ePFU RV-C15, RV-A1B or sham. Mice inoculated with RV-C15 showed lung viral titers of 1 x 105 TCID50 units 24 h after infection, with levels declining thereafter. IFN-α, ß, γ and λ2 mRNAs peaked 24-72 hrs post-infection. Immunofluorescence verified colocalization of RV-C15, CDHR3 and acetyl-α-tubulin in mouse ciliated airway epithelial cells. Compared to RV-A1B, mice infected with RV-C15 demonstrated higher bronchoalveolar eosinophils, mRNA expression of IL-5, IL-13, IL-25, Muc5ac and Gob5/Clca, protein production of IL-5, IL-13, IL-25, IL-33 and TSLP, and expansion of type 2 innate lymphoid cells. Analogous results were found in mice treated with house dust mite before infection, including increased airway responsiveness. In contrast to Rorafl/fl littermates, RV-C-infected Rorafl/flIl7rcre mice deficient in ILC2s failed to show eosinophilic inflammation or mRNA expression of IL-13, Muc5ac and Muc5b. We conclude that, compared to RV-A1B, RV-C15 infection induces ILC2-dependent type 2 airway inflammation, providing insight into the mechanism of RV-C-induced asthma exacerbations.


Subject(s)
Asthma/immunology , Coxsackievirus Infections/immunology , Enterovirus/immunology , Eosinophilia/immunology , Lymphocytes/immunology , Animals , Asthma/blood , Asthma/diagnosis , Asthma/virology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cadherin Related Proteins , Cadherins/genetics , Cadherins/metabolism , Coxsackievirus Infections/blood , Coxsackievirus Infections/complications , Coxsackievirus Infections/virology , Disease Models, Animal , Enterovirus/metabolism , Eosinophilia/blood , Eosinophilia/virology , Eosinophils/immunology , Female , HeLa Cells , Humans , Immunity, Innate , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Symptom Flare Up
6.
Article in English | MEDLINE | ID: mdl-32984077

ABSTRACT

Exposure to respiratory pathogens is a leading cause of exacerbations of airway diseases such as asthma and chronic obstructive pulmonary disease (COPD). Pellino-1 is an E3 ubiquitin ligase known to regulate virally-induced inflammation. We wished to determine the role of Pellino-1 in the host response to respiratory viruses in health and disease. Pellino-1 expression was examined in bronchial sections from patients with GOLD stage two COPD and healthy controls. Primary bronchial epithelial cells (PBECs) in which Pellino-1 expression had been knocked down were extracellularly challenged with the TLR3 agonist poly(I:C). C57BL/6 Peli1-/- mice and wild type littermates were subjected to intranasal infection with clinically-relevant respiratory viruses: rhinovirus (RV1B) and influenza A. We found that Pellino-1 is expressed in the airways of normal subjects and those with COPD, and that Pellino-1 regulates TLR3 signaling and responses to airways viruses. In particular we observed that knockout of Pellino-1 in the murine lung resulted in increased production of proinflammatory cytokines IL-6 and TNFα upon viral infection, accompanied by enhanced recruitment of immune cells to the airways, without any change in viral replication. Pellino-1 therefore regulates inflammatory airway responses without altering replication of respiratory viruses.


Subject(s)
Picornaviridae Infections , Pulmonary Disease, Chronic Obstructive , Virus Diseases , Animals , Humans , Mice , Mice, Inbred C57BL , Nuclear Proteins , Rhinovirus , Ubiquitin-Protein Ligases/genetics
7.
J Allergy Clin Immunol ; 146(3): 571-582.e3, 2020 09.
Article in English | MEDLINE | ID: mdl-32344055

ABSTRACT

BACKGROUND: Early-life wheezing-associated respiratory tract infection by rhinovirus (RV) is a risk factor for asthma development. Infants are infected with many different RV strains per year. OBJECTIVE: We previously showed that RV infection of 6-day-old BALB/c mice induces a mucous metaplasia phenotype that is dependent on type 2 innate lymphoid cells (ILC2s). We hypothesized that early-life RV infection alters the response to subsequent heterologous infection, inducing an exaggerated asthma-like phenotype. METHODS: Wild-type BALB/c mice and Rorafl/flIl7rcre mice lacking ILC2s were treated as follows: (1) sham on day 6 of life plus sham on day 13 of life, (2) RV-A1B on day 6 plus sham on day 13, (3) sham on day 6 plus RV-A2 on day 13, and (4) RV-A1B on day 6 plus RV-A2 on day 13. RESULTS: Mice infected with RV-A1B at day 6 and sham at day 13 showed an increased number of bronchoalveolar lavage eosinophils and increased expression of IL-13 mRNA but not expression of IFN-γ mRNA (which is indicative of a type 2 immune response), whereas mice infected with sham on day 6 and RV-A2 on day 13 of life demonstrated increased IFN-γ expression (which is a mature antiviral response). In contrast, mice infected with RV-A1B on day 6 before RV-A2 infection on day 13 showed increased expression of IL-13, IL-5, Gob5, Muc5b, and Muc5ac mRNA; increased numbers of eosinophils and IL-13-producing ILC2s; and exaggerated mucus metaplasia and airway hyperresponsiveness. Compared with Rorafl/fl mice, Rorafl/flIl7rcre mice showed complete suppression of bronchoalveolar lavage eosinophils and mucous metaplasia. CONCLUSION: Early-life RV infection alters the response to subsequent heterologous infection, inducing an intensified asthma-like phenotype that is dependent on ILC2s.


Subject(s)
Asthma/immunology , Eosinophils/immunology , Picornaviridae Infections/immunology , Rhinovirus/physiology , Th2 Cells/immunology , Adverse Childhood Experiences , Animals , Animals, Newborn , Disease Progression , Humans , Immunity, Innate , Infant, Newborn , Interleukin-13/genetics , Interleukin-13/metabolism , Mice , Mice, Inbred BALB C , Phenotype , Respiratory Sounds
8.
Allergy ; 75(8): 2005-2019, 2020 08.
Article in English | MEDLINE | ID: mdl-32086822

ABSTRACT

BACKGROUND: Early-life wheezing-associated respiratory infection with human rhinovirus (RV) is associated with asthma development. RV infection of 6-day-old immature mice causes mucous metaplasia and airway hyperresponsiveness which is associated with the expansion of IL-13-producing type 2 innate lymphoid cells (ILC2s) and dependent on IL-25 and IL-33. We examined regulation of this asthma-like phenotype by IL-1ß. METHODS: Six-day-old wild-type or NRLP3-/- mice were inoculated with sham or RV-A1B. Selected mice were treated with IL-1 receptor antagonist (IL-1RA), anti-IL-1ß, or recombinant IL-1ß. RESULTS: Rhinovirus infection induced Il25, Il33, Il4, Il5, Il13, muc5ac, and gob5 mRNA expression, ILC2 expansion, mucus metaplasia, and airway hyperresponsiveness. RV also induced lung mRNA and protein expression of pro-IL-1ß and NLRP3 as well as cleavage of caspase-1 and pro-IL-1ß, indicating inflammasome priming and activation. Lung macrophages were a major source of IL-1ß. Inhibition of IL-1ß signaling with IL-1RA, anti-IL-1ß, or NLRP3 KO increased RV-induced type 2 cytokine immune responses, ILC2 number, and mucus metaplasia, while decreasing IL-17 mRNA expression. Treatment with IL-1ß had the opposite effect, decreasing IL-25, IL-33, and mucous metaplasia while increasing IL-17 expression. IL-1ß and IL-17 each suppressed Il25, Il33, and muc5ac mRNA expression in cultured airway epithelial cells. Finally, RV-infected 6-day-old mice showed reduced IL-1ß mRNA and protein expression compared to mature mice. CONCLUSION: Macrophage IL-1ß limits type 2 inflammation and mucous metaplasia following RV infection by suppressing epithelial cell innate cytokine expression. Reduced IL-1ß production in immature animals provides a mechanism permitting asthma development after early-life viral infection.


Subject(s)
Picornaviridae Infections , Rhinovirus , Animals , Cytokines , Immunity, Innate , Lymphocytes , Metaplasia , Mice , Mucus
9.
Mucosal Immunol ; 12(4): 958-968, 2019 07.
Article in English | MEDLINE | ID: mdl-31089187

ABSTRACT

Activation of the inflammasome is a key function of the innate immune response that regulates inflammation in response to microbial substances. Inflammasome activation by human rhinovirus (RV), a major cause of asthma exacerbations, has not been well studied. We examined whether RV induces inflammasome activation in vivo, molecular mechanisms underlying RV-stimulated inflammasome priming and activation, and the contribution of inflammasome activation to RV-induced airway inflammation and exacerbation. RV infection triggered lung mRNA and protein expression of pro-IL-1ß and NLRP3, indicative of inflammasome priming, as well as cleavage of caspase-1 and pro-IL-1ß, completing inflammasome activation. Immunofluorescence staining showed IL-1ß in lung macrophages. Depletion with clodronate liposomes and adoptive transfer experiments showed macrophages to be required and sufficient for RV-induced inflammasome activation. TLR2 was required for RV-induced inflammasome priming in vivo. UV irradiation blocked inflammasome activation and RV genome was sufficient for inflammasome activation in primed cells. Naive and house dust mite-treated NLRP3-/- and IL-1ß-/- mice, as well as IL-1 receptor antagonist-treated mice, showed attenuated airway inflammation and responsiveness following RV infection. We conclude that RV-induced inflammasome activation is required for maximal airway inflammation and hyperresponsiveness in naive and allergic mice. The inflammasome represents a molecular target for RV-induced asthma exacerbations.


Subject(s)
Allergens/immunology , Inflammasomes/metabolism , Picornaviridae Infections/immunology , Picornaviridae Infections/metabolism , Respiratory Tract Infections/immunology , Respiratory Tract Infections/metabolism , Rhinovirus/immunology , Animals , Disease Models, Animal , Humans , Immunization , Interleukin-1beta/genetics , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Picornaviridae Infections/virology , Pyroglyphidae/immunology , Respiratory Tract Infections/virology , Rhinovirus/genetics , Toll-Like Receptor 2/metabolism
10.
Am J Physiol Lung Cell Mol Physiol ; 317(1): L57-L70, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30908938

ABSTRACT

Asthma exacerbations are often caused by rhinovirus (RV). We and others have shown that Toll-like receptor 2 (TLR2), a membrane surface receptor that recognizes bacterial lipopeptides and lipoteichoic acid, is required and sufficient for RV-induced proinflammatory responses in vitro and in vivo. We hypothesized that viral protein-4 (VP4), an internal capsid protein that is myristoylated upon viral replication and externalized upon viral binding, is a ligand for TLR2. Recombinant VP4 and myristoylated VP4 (MyrVP4) were purified by Ni-affinity chromatography. MyrVP4 was also purified from RV-A1B-infected HeLa cells by urea solubilization and anti-VP4 affinity chromatography. Finally, synthetic MyrVP4 was produced by chemical peptide synthesis. MyrVP4-TLR2 interactions were assessed by confocal fluorescence microscopy, fluorescence resonance energy transfer (FRET), and monitoring VP4-induced cytokine mRNA expression in the presence of anti-TLR2 and anti-VP4. MyrVP4 and TLR2 colocalized in TLR2-expressing HEK-293 cells, mouse bone marrow-derived macrophages, human bronchoalveolar macrophages, and human airway epithelial cells. Colocalization was absent in TLR2-null HEK-293 cells and blocked by anti-TLR2 and anti-VP4. Cy3-labeled MyrVP4 and Cy5-labeled anti-TLR2 showed an average fractional FRET efficiency of 0.24 ± 0.05, and Cy5-labeled anti-TLR2 increased and unlabeled MyrVP4 decreased FRET efficiency. MyrVP4-induced chemokine mRNA expression was higher than that elicited by VP4 alone and was attenuated by anti-TLR2 and anti-VP4. Cytokine expression was similarly increased by MyrVP4 purified from RV-infected HeLa cells and synthetic MyrVP4. We conclude that, during RV infection, MyrVP4 and TLR2 interact to generate a proinflammatory response.


Subject(s)
Asthma/genetics , Capsid Proteins/genetics , Eosinophilia/genetics , Picornaviridae Infections/genetics , Protein Processing, Post-Translational , Toll-Like Receptor 2/genetics , Viral Proteins/genetics , Adolescent , Amino Acid Sequence , Animals , Asthma/immunology , Asthma/pathology , Asthma/virology , Capsid Proteins/immunology , Child , Eosinophilia/immunology , Eosinophilia/pathology , Eosinophilia/virology , Epithelial Cells/immunology , Epithelial Cells/virology , Female , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Macrophages/immunology , Macrophages/virology , Male , Mice , Mice, Inbred C57BL , Myristic Acids/immunology , Myristic Acids/metabolism , Picornaviridae Infections/immunology , Picornaviridae Infections/pathology , Picornaviridae Infections/virology , Protein Binding , Rhinovirus/immunology , Rhinovirus/pathogenicity , Signal Transduction , Toll-Like Receptor 2/immunology , Viral Proteins/immunology , Virus Replication
11.
JCI Insight ; 3(16)2018 08 23.
Article in English | MEDLINE | ID: mdl-30135310

ABSTRACT

Enterovirus D68 (EV-D68) shares biologic features with rhinovirus (RV). In 2014, a nationwide outbreak of EV-D68 was associated with severe asthma-like symptoms. We sought to develop a mouse model of EV-D68 infection and determine the mechanisms underlying airway disease. BALB/c mice were inoculated intranasally with EV-D68 (2014 isolate), RV-A1B, or sham, alone or in combination with anti-IL-17A or house dust mite (HDM) treatment. Like RV-A1B, lung EV-D68 viral RNA peaked 12 hours after infection. EV-D68 induced airway inflammation, expression of cytokines (TNF-α, IL-6, IL-12b, IL-17A, CXCL1, CXCL2, CXCL10, and CCL2), and airway hyperresponsiveness, which were suppressed by anti-IL-17A antibody. Neutrophilic inflammation and airway responsiveness were significantly higher after EV-D68 compared with RV-A1B infection. Flow cytometry showed increased lineage-, NKp46-, RORγt+ IL-17+ILC3s and γδ T cells in the lungs of EV-D68-treated mice compared with those in RV-treated mice. EV-D68 infection of HDM-exposed mice induced additive or synergistic increases in BAL neutrophils and eosinophils and expression of IL-17, CCL11, IL-5, and Muc5AC. Finally, patients from the 2014 epidemic period with EV-D68 showed significantly higher nasopharyngeal IL-17 mRNA levels compared with patients with RV-A infection. EV-D68 infection induces IL-17-dependent airway inflammation and hyperresponsiveness, which is greater than that generated by RV-A1B, consistent with the clinical picture of severe asthma-like symptoms.


Subject(s)
Asthma/immunology , Enterovirus D, Human/immunology , Enterovirus Infections/immunology , Interleukin-17/metabolism , Neutrophils/immunology , Allergens/administration & dosage , Allergens/immunology , Animals , Asthma/pathology , Asthma/virology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Line, Tumor , Child , Child, Preschool , Disease Models, Animal , Enterovirus/immunology , Enterovirus/isolation & purification , Enterovirus D, Human/isolation & purification , Enterovirus Infections/pathology , Enterovirus Infections/virology , Female , Humans , Infant , Infant, Newborn , Interleukin-17/antagonists & inhibitors , Interleukin-17/genetics , Interleukin-17/immunology , Lung/cytology , Lung/pathology , Male , Mice , Nasopharynx/immunology , Nasopharynx/pathology , Nasopharynx/virology , Neutrophils/drug effects , Neutrophils/metabolism , Pyroglyphidae/immunology , RNA, Messenger/metabolism
12.
Influenza Other Respir Viruses ; 12(6): 717-727, 2018 11.
Article in English | MEDLINE | ID: mdl-30120824

ABSTRACT

BACKGROUND: Rhinovirus (RV) causes the common cold and asthma exacerbations. The RV genome is a 7.3 kb single-strand positive-sense RNA. OBJECTIVE: Using minor group RV1A as a backbone, we sought to design and generate a recombinant RV1A accommodating fluorescent marker expression, thereby allowing tracking of viral infection. METHOD: Recombinant RV1A infectious cDNA clones harboring the coding sequence of green fluorescent protein (GFP), Renilla luciferase, or iLOV (for light, oxygen, or voltage sensing) were engineered and constructed. RV-infected cells were determined by flow cytometry, immunohistochemistry, and immunofluorescence microscopy. RESULTS: RV1A-GFP showed a cytopathic effect in HeLa cells but failed to express GFP or Renilla luciferase due to deletion. The smaller fluorescent protein construct, RV1A-iLOV, was stably expressed in infected cells. RV1A-iLOV expression was used to examine the antiviral effect of bafilomycin in HeLa cells. Compared to parental virus, RV1A-iLOV infection of BALB/c mice yielded a similar viral load and level of cytokine mRNA expression. However, imaging of fixed lung tissue failed to reveal a fluorescent signal, likely due to the oxidation and bleaching of iLOV-bound flavin mononucleotide. We therefore employed an anti-iLOV antibody for immunohistochemical and immunofluorescence imaging. The iLOV signal was identified in airway epithelial cells and CD45+ CD11b+ lung macrophages. CONCLUSIONS: These results suggest that RV1A-iLOV is a useful molecular tool for studying RV pathogenesis. The construction strategy for RV1A-iLOV could be applied to other RV serotypes. However, the detection of iLOV-expressing RV in fixed tissue required the use of an anti-iLOV antibody, limiting the value of this construct.


Subject(s)
Luminescent Proteins/analysis , Picornaviridae Infections/virology , Rhinovirus/growth & development , Staining and Labeling/methods , Animals , Cytopathogenic Effect, Viral , Flow Cytometry , Gene Expression , Genomic Instability , HeLa Cells , Humans , Immunohistochemistry , Luminescent Proteins/genetics , Mice, Inbred BALB C , Microscopy, Fluorescence , Picornaviridae Infections/pathology , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Rhinovirus/genetics , Viral Load
13.
J Pediatr Surg ; 53(10): 1896-1903, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29960740

ABSTRACT

PURPOSE: An artificial placenta (AP) utilizing extracorporeal life support (ECLS) could avoid the harm of mechanical ventilation (MV) while allowing the lungs to develop. METHODS: AP lambs (n = 5) were delivered at 118 days gestational age (GA; term = 145 days) and placed on venovenous ECLS (VV-ECLS) with jugular drainage and umbilical vein reinfusion. Lungs remained fluid-filled. After 10 days, lambs were ventilated. MV control lambs were delivered at 118 ("early MV"; n = 5) or 128 days ("late MV"; n = 5), and ventilated. Compliance and oxygenation index (OI) were calculated. After sacrifice, lungs were procured and H&E-stained slides scored for lung injury. Slides were also immunostained for PDGFR-α and α-actin; alveolar development was quantified by the area fraction of alveolar septal tips staining double-positive for both markers. RESULTS: Compliance of AP lambs was 2.79 ±â€¯0.81 Cdyn compared to 0.83 ±â€¯0.19 and 3.04 ±â€¯0.99 for early and late MV, respectively. OI in AP lambs was lower than early MV lambs (6.20 ±â€¯2.10 vs. 36.8 ±â€¯16.8) and lung injury lower as well (1.8 ±â€¯1.6 vs. 6.0 ±â€¯1.2). Double-positive area fractions were higher in AP lambs (0.012 ±â€¯0.003) than early (0.003 ±â€¯0.0005) and late (0.004 ±â€¯0.002) MV controls. CONCLUSIONS: Lung development continues and lungs are protected from injury during AP support relative to mechanical ventilation. LEVEL OF EVIDENCE: n/a (basic/translational science).


Subject(s)
Artificial Organs , Extracorporeal Membrane Oxygenation , Lung/growth & development , Premature Birth/therapy , Animals , Animals, Newborn , Disease Models, Animal , Female , Gestational Age , Lung/physiology , Placenta/physiology , Pregnancy , Sheep
14.
Neonatology ; 113(4): 313-321, 2018.
Article in English | MEDLINE | ID: mdl-29478055

ABSTRACT

BACKGROUND: Extremely premature neonates suffer high morbidity and mortality. An artificial placenta (AP) using extracorporeal life support (ECLS) is a promising therapy. OBJECTIVES: We hypothesized that intratracheal perfluorocarbon (PFC) instillation during AP support would reduce lung injury and promote lung development relative to intratracheal amniotic fluid or crystalloid. METHODS: Lambs at an estimated gestational age (EGA) 116-121 days (term 145 days) were placed on venovenous ECLS with jugular drainage and umbilical vein reinfusion and intubated. Airways were managed by the instillation of amniotic fluid and tracheal occlusion (TO; n = 4), or lactated Ringer's (LR; n = 4) or perfluorodecalin (a PFC) without occlusion (n = 4). After 7 days, the animals were sacrificed. Early (EGA 116-121 days) and late (EGA 125-131 days) tissue control lambs were delivered and sacrificed. Lungs were formalin-inflated to 30 cm H2O and sectioned for histology. Injury was scored by an unbiased pathologist. Slides were immunostained for PDGFR-α and α-actin; development was quantified by the area fraction of double-positive tips. Surfactant protein-C (SP-C) concentration in bronchoalveolar lavage fluid was quantified using ELISA. RESULTS: Total injury scores were lower in PFC lungs (1.8 ± 1.7) than in TO (6.5 ± 2.1; p = 0.01) and LR lungs (5.5 ± 2.4; p = 0.01). The area fraction of double-positive alveolar tips appeared higher in PFC lungs than in TO lungs (0.18 ± 0.007 vs. 0.008 ± 0.004; p = 0.07). SP-C concentration was higher in PFC lungs than in TO lungs (37.9 ± 7.6 vs. 20.0 ± 5.4 pg/mL; p = 0.005), and both early (12.4 ± 1.7 g/mL; p = 0.007) and late tissue control lungs (15.1 ± 5.0 pg/mL; p = 0.0008). CONCLUSION: During AP support, intratracheal PFC prevents lung injury and promotes normal lung development better than crystalloid or amniotic fluid with TO.


Subject(s)
Animals, Newborn , Artificial Organs , Extracorporeal Membrane Oxygenation , Fluorocarbons/administration & dosage , Lung Injury/prevention & control , Animals , Female , Lung/growth & development , Placenta/physiology , Pregnancy , Sheep
15.
J Immunol ; 199(4): 1308-1318, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28701507

ABSTRACT

Early-life respiratory viral infection is a risk factor for asthma development. Rhinovirus (RV) infection of 6-d-old mice, but not mature mice, causes mucous metaplasia and airway hyperresponsiveness that are associated with the expansion of lung type 2 innate lymphoid cells (ILC2s) and are dependent on IL-13 and the innate cytokine IL-25. However, contributions of the other innate cytokines, IL-33 and thymic stromal lymphopoietin (TSLP), to the observed asthma-like phenotype have not been examined. We reasoned that IL-33 and TSLP expression are also induced by RV infection in immature mice and are required for maximum ILC2 expansion and mucous metaplasia. We inoculated 6-d-old BALB/c (wild-type) and TSLP receptor-knockout mice with sham HeLa cell lysate or RV. Selected mice were treated with neutralizing Abs to IL-33 or recombinant IL-33, IL-25, or TSLP. ILC2s were isolated from RV-infected immature mice and treated with innate cytokines ex vivo. RV infection of 6-d-old mice increased IL-33 and TSLP protein abundance. TSLP expression was localized to the airway epithelium, whereas IL-33 was expressed in epithelial and subepithelial cells. RV-induced mucous metaplasia, ILC2 expansion, airway hyperresponsiveness, and epithelial cell IL-25 expression were attenuated by anti-IL-33 treatment and in TSLP receptor-knockout mice. Administration of intranasal IL-33 and TSLP was sufficient for mucous metaplasia. Finally, TSLP was required for maximal ILC2 gene expression in response to IL-25 and IL-33. The generation of mucous metaplasia in immature RV-infected mice involves a complex interplay among the innate cytokines IL-25, IL-33, and TSLP.


Subject(s)
Cytokines/immunology , Interleukin-33/immunology , Interleukins/immunology , Lymphocyte Activation , Lymphocytes/physiology , Metaplasia/immunology , Picornaviridae Infections/immunology , Rhinovirus/immunology , Age Factors , Animals , Asthma/immunology , Asthma/virology , Cytokines/genetics , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/virology , Immunoglobulins/genetics , Immunoglobulins/immunology , Immunoglobulins/metabolism , Interleukin-33/genetics , Interleukins/genetics , Lymphocytes/immunology , Metaplasia/pathology , Metaplasia/virology , Mice , Mice, Knockout , Mucous Membrane/immunology , Mucous Membrane/pathology , Picornaviridae Infections/virology , Receptors, Cytokine/genetics , Receptors, Cytokine/immunology , Receptors, Cytokine/metabolism , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/virology , Thymic Stromal Lymphopoietin
16.
Am J Physiol Lung Cell Mol Physiol ; 312(6): L983-L993, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28360114

ABSTRACT

Early-life wheezing-associated respiratory tract infection by rhinovirus (RV) is considered a risk factor for asthma development. We have shown that RV infection of 6-day-old BALB/c mice, but not mature mice, induces an asthmalike phenotype that is associated with an increase in the population of type 2 innate lymphoid cells (ILC2s) and dependent on IL-13 and IL-25. We hypothesize that ILC2s are required and sufficient for development of the asthmalike phenotype in immature mice. Mice were infected with RV1B on day 6 of life and treated with vehicle or a chemical inhibitor of retinoic acid receptor-related orphan receptor-α (RORα), SR3335 (15 mg·kg-1·day-1 ip for 7 days). We also infected Rorasg/sg mice without functional ILC2s. ILC2s were identified as negative for lineage markers and positive for cluster of differentiation 25 (CD25)/IL-2Rα and CD127/IL-7Rα. Effects of SR3335 on proliferation and function of cultured ILC2s were determined. Finally, sorted ILC2s were transferred into naïve mice, and lungs were harvested 14 days later for assessment of gene expression and histology. SR3335 decreased the number of RV-induced lung lineage-negative, CD25+, CD127+ ILC2s in immature mice. SR3335 also attenuated lung mRNA expression of IL-13, Muc5ac, and Gob5 as well as mucous metaplasia. We also found reduced expansion of ILC2s in RV-infected Rorasg/sg mice. SR3335 also blocked IL-25 and IL-33-induced ILC2 proliferation and IL-13 production ex vivo. Finally, adoptive transfer of ILC2s led to development of asthmalike phenotype in immature and adult mice. RORα-dependent ILC2s are required and sufficient for type 2 cytokine expression and mucous metaplasia in immature mice.


Subject(s)
Aging/immunology , Immunity, Innate , Lymphocytes/immunology , Mucus/immunology , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Adoptive Transfer , Animals , Cell Proliferation/drug effects , Cell Separation , HeLa Cells , Humans , Immunity, Innate/drug effects , Lymphocytes/drug effects , Metaplasia , Mice, Inbred BALB C , Rhinovirus/drug effects , Sulfonamides , Thiophenes
17.
Am J Respir Cell Mol Biol ; 56(2): 242-251, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27679954

ABSTRACT

Early-life wheezing-associated infections with rhinovirus (RV) have been associated with asthma development in children. We have shown that RV infection of 6-day-old mice induces mucous metaplasia and airways hyperresponsiveness, which is dependent on IL-13, IL-25, and type 2 innate lymphoid cells (ILC2s). Infection of immature mice fails to induce lung IFN-γ expression, in contrast to mature 8-week-old mice with a robust IFN-γ response, consistent with the notion that deficient IFN-γ production in immature mice permits RV-induced type 2 immune responses. We therefore examined the effects of intranasal IFN-γ administration on RV-induced ILC2 expansion and IL-13 expression in 6-day-old BALB/c and IL-13 reporter mice. Airway responses were assessed by histology, immunofluorescence microscopy, quantitative polymerase chain reaction, ELISA, and flow cytometry. Lung ILC2s were also treated with IFN-γ ex vivo. We found that, compared with untreated RV-infected immature mice, IFN-γ treatment attenuated RV-induced IL-13 and Muc5ac mRNA expression and mucous metaplasia. IFN-γ also reduced ILC2 expansion and the percentage of IL-13-secreting ILC2s. IFN-γ had no effect on the mRNA or protein expression of IL-25, IL-33, or thymic stromal lymphoprotein. Finally, IFN-γ treatment of sorted ILC2s reduced IL-5, IL-13, IL-17RB, ST2, and GATA-3 mRNA expression. We conclude that, in immature mice, IFN-γ inhibits ILC2 expansion and IL-13 expression in vivo and ex vivo, thereby attenuating RV-induced mucous metaplasia. These findings demonstrate the antagonistic function of IFN-γ on ILC2 expansion and gene expression, the absence of which may contribute to the development of an asthma-like phenotype after early-life RV infection.


Subject(s)
Asthma/drug therapy , Asthma/immunology , Immunity, Innate , Interferon-gamma/therapeutic use , Lymphocytes/immunology , Picornaviridae Infections/immunology , Picornaviridae Infections/virology , Rhinovirus/physiology , Animals , Animals, Newborn , Asthma/complications , Asthma/virology , Cell Lineage/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , HeLa Cells , Humans , Immunity, Innate/drug effects , Interferon-gamma/pharmacology , Interleukin-13/metabolism , Lymphocytes/drug effects , Metaplasia , Mice , Mice, Inbred BALB C , Mucus/metabolism , Phenotype , Picornaviridae Infections/complications , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rhinovirus/drug effects
18.
J Immunol ; 196(11): 4692-705, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27183577

ABSTRACT

Infants with a history of prematurity and bronchopulmonary dysplasia have a high risk of asthma and viral-induced exacerbations later in life. We hypothesized that hyperoxic exposure, a predisposing factor to bronchopulmonary dysplasia, modulates the innate immune response, producing an exaggerated proinflammatory reaction to viral infection. Two- to 3-d-old C57BL/6J mice were exposed to air or 75% oxygen for 14 d. Mice were infected intranasally with rhinovirus (RV) immediately after O2 exposure. Lung mRNA and protein expression, histology, dendritic cells (DCs), and airway responsiveness were assessed 1-12 d postinfection. Tracheal aspirates from premature human infants were collected for mRNA detection. Hyperoxia increased lung IL-12 expression, which persisted up to 12 d postexposure. Hyperoxia-exposed RV-infected mice showed further increases in IL-12 and increased expression of IFN-γ, TNF-α, CCL2, CCL3, and CCL4, as well as increased airway inflammation and responsiveness. In RV-infected, air-exposed mice, the response was not significant. Induced IL-12 expression in hyperoxia-exposed, RV-infected mice was associated with increased IL-12-producing CD103(+) lung DCs. Hyperoxia also increased expression of Clec9a, a CD103(+) DC-specific damaged cell-recognition molecule. Hyperoxia increased levels of ATP metabolites and expression of adenosine receptor A1, further evidence of cell damage and related signaling. In human preterm infants, tracheal aspirate Clec9a expression positively correlated with the level of prematurity. Hyperoxic exposure increases the activation of CD103(+), Clec9a(+) DCs, leading to increased inflammation and airway hyperresponsiveness upon RV infection. In premature infants, danger signal-induced DC activation may promote proinflammatory airway responses, thereby increasing respiratory morbidity.


Subject(s)
Hyperoxia/immunology , Respiratory Tract Infections/immunology , Rhinovirus/immunology , Signal Transduction/immunology , Animals , Animals, Newborn , Dendritic Cells/immunology , Dendritic Cells/pathology , Disease Models, Animal , Humans , Inflammation/immunology , Interleukin-12/biosynthesis , Interleukin-12/immunology , Lung/immunology , Lung/pathology , Mice , Mice, Inbred C57BL
19.
J Allergy Clin Immunol ; 138(6): 1619-1630, 2016 12.
Article in English | MEDLINE | ID: mdl-27084403

ABSTRACT

BACKGROUND: We have shown that rhinovirus, a cause of asthma exacerbation, colocalizes with CD68+ and CD11b+ airway macrophages after experimental infection in human subjects. We have also shown that rhinovirus-induced cytokine expression is abolished in Toll-like receptor (TLR2)-/- bone marrow-derived macrophages. OBJECTIVE: We hypothesize that TLR2+ macrophages are required and sufficient for rhinovirus-induced airway inflammation in vivo. METHODS: Naive and ovalbumin (OVA)-sensitized and challenged C57BL/6 wild-type and TLR2-/- mice were infected with RV1B, followed by IgG or anti-TLR2, to determine the requirement and sufficiency of TLR2 for rhinovirus-induced airway responses. Bone marrow chimera experiments using OVA-treated C57BL/6 and TLR2-/- mice were also performed. Finally, naive TLR2-/- mice underwent intranasal transfer of bone marrow-derived wild-type macrophages. RESULTS: RV1B infection of naive wild-type mice induced an influx of airway neutrophils and CD11b+ exudative macrophages, which was reduced in TLR2-/- mice. After allergen exposure, rhinovirus-induced neutrophilic and eosinophilic airway inflammation and hyperresponsiveness were reduced in TLR2-/- and anti-TLR2-treated mice. Transfer of TLR2-/- bone marrow into wild-type, OVA-treated C57BL/6 mice blocked rhinovirus-induced airway responses, whereas transfer of wild-type marrow to TLR2-/- mice restored them. Finally, transfer of wild-type macrophages to naive TLR2-/- mice was sufficient for neutrophilic inflammation after rhinovirus infection, whereas macrophages treated with IL-4 (to induce M2 polarization) were sufficient for eosinophilic inflammation, mucous metaplasia, and airways hyperresponsiveness. CONCLUSIONS: TLR2 is required for early inflammatory responses induced by rhinovirus, and TLR2+ macrophages are sufficient to confer airway inflammation to TLR2-/- mice, with the pattern of inflammation depending on the macrophage activation state.


Subject(s)
Asthma/immunology , Macrophages/physiology , Picornaviridae Infections/immunology , Rhinovirus/immunology , Toll-Like Receptor 2/metabolism , Animals , Cells, Cultured , Humans , Macrophage Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Toll-Like Receptor 2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...