16 research outputs found

    Inhibition of uric acid or IL- 1β ameliorates respiratory syncytial virus immunopathology and development of asthma

    Full text link
    BackgroundRespiratory syncytial virus (RSV) affects most infants early in life and is associated with increased asthma risk. The specific mechanism remains unknown.ObjectiveTo investigate the role of uric acid (UA) and IL- 1β in RSV immunopathology and asthma predisposition.MethodsTracheal aspirates from human infants with and without RSV were collected and analyzed for pro- IL- 1β mRNA and protein to establish a correlation in human disease. Neonatal mouse models of RSV were employed, wherein mice infected at 6- 7 days of life were analyzed at 8 days postinfection, 5 weeks postinfection, or after a chronic cockroach allergen asthma model. A xanthine oxidase inhibitor or IL- 1 receptor antagonist was administered during RSV infection.ResultsHuman tracheal aspirates from RSV- infected infants showed elevated pro- IL- 1β mRNA and protein. Inhibition of UA or IL- 1β during neonatal murine RSV infection decreased mucus production, reduced cellular infiltrates to the lung (especially ILC2s), and decreased type 2 immune responses. Inhibition of either UA or IL- 1β during RSV infection led to chronic reductions in pulmonary immune cell composition and reduced type 2 immune responses and reduced similar responses after challenge with cockroach antigen.ConclusionsInhibiting UA and IL- 1β during RSV infection ameliorates RSV immunopathology, reduces the consequences of allergen- induced asthma, and presents new therapeutic targets to reduce early- life viral- induced asthma development.Neonatal RSV infection is associated with increases in pulmonary uric acid and IL- 1β and lung immunopathology. XOI or IL- 1RA administration during neonatal RSV infection leads to reduced RSV immunopathology. XOI or IL- 1RA administration during neonatal RSV infection leads to reduced type 2 immune responses during a subsequent model of asthma.Abbreviations: IL- 1RA, IL- 1 receptor antagonist; RSV: Respiratory syncytial virus; XOI, xanthine oxidase inhibitor.Peer Reviewedhttp://deepblue.lib.umich.edu/bitstream/2027.42/162774/3/all14310.pdfhttp://deepblue.lib.umich.edu/bitstream/2027.42/162774/2/all14310_am.pdfhttp://deepblue.lib.umich.edu/bitstream/2027.42/162774/1/all14310-sup-0005-TableS1.pd

    Marine Algal Toxin Azaspiracid Is an Open-State Blocker of hERG Potassium Channels

    Get PDF
    Azaspiracids (AZA) are polyether marine dinoflagellate toxins that accumulate in shellfish and represent an emerging human health risk. Although human exposure is primarily manifested by severe and protracted diarrhea, this toxin class has been shown to be highly cytotoxic, a teratogen to developing fish, and a possible carcinogen in mice. Until now, AZA's molecular target(s) has not yet been determined. Using three independent methods (voltage clamp, channel binding assay, and thallium flux assay), we have for the first time demonstrated that AZA1, AZA2, and AZA3 each bind to and block the hERG (human ether-à-go-go related gene) potassium channel heterologously expressed in HEK-293 mammalian cells. Inhibition of K+ current for each AZA analogue was concentration-dependent (IC50 value range: 0.64 - 0.84 μM). The mechanism of hERG channel inhibition by AZA1 was investigated further in Xenopus oocytes where it was shown to be an open state-dependent blocker and, using mutant channels, to interact with F656 but not with Y652 within the S6 transmembrane domain that forms the channel's central pore. AZA1, AZA2, and AZA3 were each shown to inhibit [3H]dofetilide binding to the hERG channel and thallium ion flux through the channel (IC50 value range: 2.1 – 6.6 μM). AZA1 did not block K+ current of the closely related EAG1 channel. Collectively, these data suggest that the AZAs physically block the K+ conductance pathway of hERG1 channels by occluding the cytoplasmic mouth of the open pore. Although the concentrations necessary to block hERG channels are relatively high, AZA-induced blockage may prove to contribute to the toxicological properties of the AZAs

    The militarisation of English schools: Troops to Teaching and the implications for Initial Teacher Education and race equality

    Get PDF
    This article considers the implications of the Troops to Teaching (TtT) programme, to be introduced in England in autumn 2013, for Initial Teacher Education (ITE) and race equality. TtT will fast-track ex-armed service members to teach in schools, without necessarily the requirement of a university degree. Employing theories of white supremacy, and Althusser’s (1971) concept of Ideological and Repressive State Apparatus, I argue that this initiative both stems from, and contributes to, a system of social privilege and oppression in education. Despite appearing to be aimed at all young people, the planned TtT initiative is actually aimed at poor and racially subordinated youth. This is likely to further entrench polarisation in a system which already provides two tier educational provision: TtT will be a programme for the inner-city disadvantaged, whilst wealthier, whiter schools will mostly continue to get highly qualified teachers. Moreover, TtT contributes to a wider devaluing of current ITE; ITE itself is rendered virtually irrelevant, as it seems TtT teachers will not be subject specialists, rather will be expected to provide military-style discipline, the skills for which they will be expected to bring with them. More sinister, I argue that TtT is part of the wider militarisation of education. This military-industrial-education complex seeks to contain and police young people who are marginalised along lines of race and class, and contributes to a wider move to increase ideological support for foreign wars - both aims ultimately in the service of neoliberal objectives which will feed social inequalities

    Sirtuin 1 Regulates Dendritic Cell Activation and Autophagy during Respiratory Syncytial Virus–Induced Immune Responses

    No full text
    Respiratory syncytial virus (RSV) is the major cause of lower respiratory tract infection in children worldwide. Sirtuin 1 (SIRT1), an NAD(+) dependent deacetylase, has been associated with the induction of autophagy and the regulation of inflammatory mediators. We found that Sirt1 was upregulated in mouse lung after RSV infection. Infected animals that received EX-527, a selective SIRT1 inhibitor, displayed exacerbated lung pathology, with increased mucus production, elevated viral load, and enhanced Th2 cytokine production. Gene expression analysis of isolated cell populations revealed that Sirt1 was most highly upregulated in RSV-treated dendritic cells (DCs). Upon RSV infection, EX-527-treated DCs, Sirt1 siRNA-treated DCs, or DCs from conditional knockout (Sirt1(f/f)-CD11c–Cre(+)) mice showed downregulated inflammatory cytokine gene expression and attenuated autophagy. Finally, RSV infection of Sirt1(f/f)-CD11c–Cre(+) mice resulted in altered lung and lymph node cytokine responses, leading to exacerbated pathology. These data indicate that SIRT1 promotes DC activation associated with autophagy-mediated processes during RSV infection, thereby directing efficient antiviral immune responses

    Intranasal delivery of allergen in a nanoemulsion adjuvant inhibits allergen- specific reactions in mouse models of allergic airway disease

    Full text link
    BackgroundAtopic diseases are an increasing problem that involve both immediate hypersensitivity reactions mediated by IgE and unique cellular inflammation. Many forms of specific immunotherapy involve the administration of allergen to suppress allergic immune responses but are focused on IgE- mediated reactions. In contrast, the effect of allergen- specific immunotherapy on allergic inflammation is complex, not entirely consistent and not well understood. We have previously demonstrated the ability of allergen administered in a nanoemulsion (NE) mucosal adjuvant to suppress IgE- mediated allergic responses and protect from allergen challenge in murine food allergy models. This activity was associated with decreases in allergen- specific IL- 10 and reductions in allergic cytokines and increases in regulatory T cells.ObjectiveHere, we extend these studies to using 2 distinct models, the ovalbumin (OVA) and cockroach (CRA) models of allergic airway disease, which are based predominantly on allergic inflammation.MethodsAcute or chronic allergic airway disease was induced in mice using ovalbumin and cockroach allergen models. Mice received three therapeutic immunizations with allergen in NE, and reactivity to airway challenge was determined.ResultsTherapeutic immunization with cockroach or OVA allergen in NE markedly reduced pathology after airway challenge. The 2 models demonstrated protection from allergen challenge- induced pathology that was associated with suppression of Th2- polarized immune responses in the lung. In addition, the reduction in ILC2 numbers in the lungs of allergic mice along with reduction in epithelial cell alarmins, IL- 25 and IL- 33, suggests an overall change in the lung immune environment induced by the NE immunization protocol.Conclusions and Clinical RelevanceThese results demonstrate that suppression of allergic airway inflammation and bronchial hyper- reactivity can be achieved using allergen- specific immunotherapy without significant reductions in allergen- specific IgE and suggest that ILC2 cells may be critical targets for this activity.The ability of intranasal vaccines to modulate allergic immune responses and inflammation in the lung was studied in two distinct models of allergic airway disease. Mice were sensitized to ovalbumin or cockroach allergen and subsequently received three immunizations with allergen in nanoemulsion adjuvant. Nanoemulsion vaccines modulated the allergen- specific cytokine milieu in the lungs to suppress Th2 cytokine production, alarmin expression and recruitment of ILC2s. The immune modulation in the lungs was associated with protection from allergen challenge- induced inflammation and reactivity.Peer Reviewedhttp://deepblue.lib.umich.edu/bitstream/2027.42/170274/1/cea13903.pdfhttp://deepblue.lib.umich.edu/bitstream/2027.42/170274/2/cea13903_am.pd

    NLRP3-Inflammasome Inhibition during Respiratory Virus Infection Abrogates Lung Immunopathology and Long-Term Airway Disease Development

    No full text
    Respiratory syncytial virus (RSV) infects most infants by two years of age. It can cause severe disease leading to an increased risk of developing asthma later in life. Previously, our group has shown that RSV infection in mice and infants promotes IL-1β production. Here, we characterized the role of NLRP3-Inflammasome activation during RSV infection in adult mice and neonates. We observed that the inhibition of NLRP3 activation using the small molecule inhibitor, MCC950, or in genetically modified NLRP3 knockout (Nlrp3−/−) mice during in vivo RSV infection led to decreased lung immunopathology along with a reduced expression of the mucus-associated genes and reduced production of innate cytokines (IL-1β, IL-33 and CCL2) linked to severe RSV disease while leading to significant increases in IFN-β. NLRP3-inflammasome inhibition or deletion diminished Th2 cytokines and inflammatory cell infiltration into the lungs. Furthermore, NLRP3 inhibition or deletion during early-life RSV infection led to reducing viral-exacerbated allergic response in a mouse model of RSV-induced allergy exacerbation. Here, we demonstrated the critical role of NLRP3-inflammasome activation in RSV immunopathology and the related long-term airway alteration. Moreover, these findings suggest the NLRP3-inflammasome as a potential therapeutic target to attenuate severe RSV disease and limit childhood asthma development

    Inhibition of uric acid or IL‐1β ameliorates respiratory syncytial virus immunopathology and development of asthma

    No full text
    BackgroundRespiratory syncytial virus (RSV) affects most infants early in life and is associated with increased asthma risk. The specific mechanism remains unknown.ObjectiveTo investigate the role of uric acid (UA) and IL- 1β in RSV immunopathology and asthma predisposition.MethodsTracheal aspirates from human infants with and without RSV were collected and analyzed for pro- IL- 1β mRNA and protein to establish a correlation in human disease. Neonatal mouse models of RSV were employed, wherein mice infected at 6- 7 days of life were analyzed at 8 days postinfection, 5 weeks postinfection, or after a chronic cockroach allergen asthma model. A xanthine oxidase inhibitor or IL- 1 receptor antagonist was administered during RSV infection.ResultsHuman tracheal aspirates from RSV- infected infants showed elevated pro- IL- 1β mRNA and protein. Inhibition of UA or IL- 1β during neonatal murine RSV infection decreased mucus production, reduced cellular infiltrates to the lung (especially ILC2s), and decreased type 2 immune responses. Inhibition of either UA or IL- 1β during RSV infection led to chronic reductions in pulmonary immune cell composition and reduced type 2 immune responses and reduced similar responses after challenge with cockroach antigen.ConclusionsInhibiting UA and IL- 1β during RSV infection ameliorates RSV immunopathology, reduces the consequences of allergen- induced asthma, and presents new therapeutic targets to reduce early- life viral- induced asthma development.Neonatal RSV infection is associated with increases in pulmonary uric acid and IL- 1β and lung immunopathology. XOI or IL- 1RA administration during neonatal RSV infection leads to reduced RSV immunopathology. XOI or IL- 1RA administration during neonatal RSV infection leads to reduced type 2 immune responses during a subsequent model of asthma.Abbreviations: IL- 1RA, IL- 1 receptor antagonist; RSV: Respiratory syncytial virus; XOI, xanthine oxidase inhibitor.Peer Reviewedhttp://deepblue.lib.umich.edu/bitstream/2027.42/162774/3/all14310.pdfhttp://deepblue.lib.umich.edu/bitstream/2027.42/162774/2/all14310_am.pdfhttp://deepblue.lib.umich.edu/bitstream/2027.42/162774/1/all14310-sup-0005-TableS1.pd

    A steroid‐resistant cockroach allergen model is associated with lung and cecal microbiome changes

    No full text
    Abstract The pathogenesis of asthma has been partially linked to lung and gut microbiome. We utilized a steroid‐resistant chronic model of cockroach antigen‐induced (CRA) asthma with corticosteroid (fluticasone) treatment to examine lung and gut microbiome during disease. The pathophysiology assessment demonstrated that mucus and airway hyperresponsiveness were increased in the chronic CRA with no alteration in the fluticasone (Flut)‐treated group, demonstrating steroid resistance. Analysis of mRNA from lungs showed no decrease of MUC5AC or Gob5 in the Flut‐treated group. Furthermore, flow‐cytometry in lung tissue showed eosinophils and neutrophils were not significantly reduced in the Flut‐treated group compared to the chronic CRA group. When the microbiome profiles were assessed, data showed that only the Flut‐treated animals were significantly different in the gut microbiome. Finally, a functional analysis of cecal microbiome metabolites using PiCRUSt showed several biosynthetic pathways were significantly enriched in the Flut‐treated group, with tryptophan pathway verified by ELISA with increased kynurenine in homogenized cecum samples. While the implications of these data are unclear, they may suggest a significant impact of steroid treatment on future disease pathogenesis through microbiome and associated metabolite pathway changes

    Myeloid- and Epithelial-Derived HBEGF Promotes Pulmonary Fibrosis

    No full text
    Idiopathic pulmonary fibrosis (IPF) is a poorly understood, progressive lethal lung disease with no known cure. In addition to alveolar epithelial cell (AEC) injury and excessive deposition of extracellular matrix proteins, chronic inflammation is a hallmark of IPF. Literature suggests that the persistent inflammation seen in IPF primarily consists of monocytes and macrophages. Recent work demonstrates that monocyte-derived alveolar macrophages (moAMs) drive lung fibrosis, but further characterization of critical moAM cell attributes is necessary. Heparin-binding epidermal growth factor-like growth factor (HB-EGF) is an important EGFR (epidermal growth factor receptor) ligand that has essential roles in angiogenesis, wound healing, keratinocyte migration, and epithelial-mesenchymal transition. Our past work has shown HB-EGF is a primary marker of profibrotic M2 macrophages, and this study seeks to characterize myeloid-derived HB-EGF and its primary mechanism of action in bleomycin-induced lung fibrosis using Hbegff/f;Lyz2Cre+ mice. Here, we show that IPF patients and fibrotic mice have increased expression of HB-EGF and that lung macrophages and transitional alveolar epithelial cells of fibrotic mice and humans all express HB-EGF. We also show that Hbegff/f;Lyz2Cre+ mice are protected from bleomycin-induced fibrosis and that this protection is likely multifactorial, caused by decreased CCL2-dependent monocyte migration, decreased fibroblast migration, and decreased contribution of HB-EGF from AEC sources when HB-EGF is removed under the Lyz2Cre promoter
    corecore