1
|
Li D, van der Veen TA, de Groot LES, de Jager MH, Lan A, Baarsma HA, Lutter R, van der Graaf K, Gosens R, Schmidt M, Melgert BN. Pre-Existing Allergic Inflammation Alters Both Innate and Adaptive Immune Responses in Mice Co-Infected with Influenza Virus. Int J Mol Sci 2025; 26:3483. [PMID: 40331962 PMCID: PMC12027099 DOI: 10.3390/ijms26083483] [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: 01/18/2025] [Revised: 03/08/2025] [Accepted: 03/12/2025] [Indexed: 05/08/2025] Open
Abstract
Asthma, a chronic airway disease, is marked by allergic inflammation, hyperresponsiveness, and tissue remodeling. Influenza infections in asthma patients can cause severe exacerbations, though the underlying mechanisms remain unclear. This study investigated how pre-existing allergic inflammation affects immune responses to influenza infection in mice exposed to house dust mite (HDM). Mice were repeatedly exposed to HDM, followed by infection with the influenza A virus, and were sacrificed three days post-infection. Plasma was analyzed for HDM-specific immunoglobulins, while lung tissue was used for immune cell flow cytometry and RNA sequencing analysis. HDM exposure induced allergic inflammation, evidenced by more HDM-specific IgE, IgG1, IgG2, eosinophils, neutrophils, Th1, and Th17 cells compared to controls. Upon influenza infection, the effects of HDM and influenza co-infection interacted, showing fewer Th1 cells and regulatory T cells and more Th2 cells compared to mice exposed to the influenza virus alone. Interestingly, RNA-seq analysis revealed less upregulation of Th1-related genes and antiviral pathways in co-exposed mice, suggesting impaired Th1 immunity and antiviral responses. Pre-existing allergic inflammation significantly altered immune responses in mice co-infected with influenza, revealing underdeveloped antiviral responses as early as three days post-infection. These findings may explain the increased susceptibility of patients with asthma to severe viral diseases.
Collapse
Affiliation(s)
- Dan Li
- Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands; (D.L.); (T.A.v.d.V.); (M.H.d.J.); (A.L.); (H.A.B.); (R.G.); (M.S.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - T. Anienke van der Veen
- Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands; (D.L.); (T.A.v.d.V.); (M.H.d.J.); (A.L.); (H.A.B.); (R.G.); (M.S.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Linsey E. S. de Groot
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands (R.L.)
| | - Marina H. de Jager
- Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands; (D.L.); (T.A.v.d.V.); (M.H.d.J.); (A.L.); (H.A.B.); (R.G.); (M.S.)
| | - Andy Lan
- Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands; (D.L.); (T.A.v.d.V.); (M.H.d.J.); (A.L.); (H.A.B.); (R.G.); (M.S.)
| | - Hoeke A. Baarsma
- Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands; (D.L.); (T.A.v.d.V.); (M.H.d.J.); (A.L.); (H.A.B.); (R.G.); (M.S.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - René Lutter
- Department of Respiratory Medicine, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands (R.L.)
| | | | - Reinoud Gosens
- Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands; (D.L.); (T.A.v.d.V.); (M.H.d.J.); (A.L.); (H.A.B.); (R.G.); (M.S.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Martina Schmidt
- Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands; (D.L.); (T.A.v.d.V.); (M.H.d.J.); (A.L.); (H.A.B.); (R.G.); (M.S.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Barbro N. Melgert
- Groningen Research Institute of Pharmacy, Department of Molecular Pharmacology, University of Groningen, 9713 AV Groningen, The Netherlands; (D.L.); (T.A.v.d.V.); (M.H.d.J.); (A.L.); (H.A.B.); (R.G.); (M.S.)
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| |
Collapse
|
2
|
Ledesma-Aparicio J, Salazar-Guerrero G, Soto-Muñoz A, Ramírez-Estudillo C, Gómez-Esquivel ML, Reyes-Grajeda JP, Reyes-López CA, Vega-López MA. Evaluation of a translational swine model of respiratory hypersensitivity induced by exposure to Phleum pratense pollen allergens. FRONTIERS IN ALLERGY 2025; 6:1557656. [PMID: 40259949 PMCID: PMC12009831 DOI: 10.3389/falgy.2025.1557656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/24/2025] [Indexed: 04/23/2025] Open
Abstract
Introduction Asthma is a disease characterized by chronic inflammation of the airway mucosa that causes tissue remodeling and a reversible decrease in airflow. The causative agent of asthma is still unknown; however, several studies have shown that environmental factors such as allergens present in pollens are involved. This project's objective was to develop and evaluate a model of respiratory hypersensitivity in Vietnamese minipigs, which is closer in many aspects to humans than rodents, using Phleum pratense allergenic pollen extract. Methods In this hypersensitivity model, human-like signs were observed during a challenge with the allergens. Intradermal and passive anaphylaxis tests confirmed that specific IgE mediated the response. Results Significant changes in lung tissue remodeling, high levels of serum allergen-specific IgA, IgG, and to a lesser extent IgE were found in the sensitized pigs, which could favor tolerance and pathogenesis. However, since chronic pathology did not develop, elevated levels of cytokines were not proven. Discussion This work demonstrated that the immunization protocol in this experimental model can induce a type I respiratory hypersensitivity-like response mediated by antigen-specific IgE, with pathophysiological similarities to those of humans and prospective for translational basic and applied research.
Collapse
Affiliation(s)
- Jessica Ledesma-Aparicio
- Laboratorio de Estructura de Proteínas, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Gustavo Salazar-Guerrero
- Laboratorio de Bioquímica Estructural, Escuela Nacional de Medicina y Homeopatía del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Axel Soto-Muñoz
- Departamento de Infectómica y Patogénesis Molecular, Cinvestav, Zacatenco, Mexico City, México
| | | | - Mónica Luz Gómez-Esquivel
- Laboratorio de Bioquímica Estructural, Escuela Nacional de Medicina y Homeopatía del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Juan Pablo Reyes-Grajeda
- Laboratorio de Estructura de Proteínas, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - César A. Reyes-López
- Laboratorio de Bioquímica Estructural, Escuela Nacional de Medicina y Homeopatía del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Marco A. Vega-López
- Departamento de Infectómica y Patogénesis Molecular, Cinvestav, Zacatenco, Mexico City, México
| |
Collapse
|
3
|
Ralchev N, Bradyanova S, Kerekov N, Tchorbanov A, Mihaylova N. Suppression of Pathological Allergen-Specific B Cells by Protein-Engineered Molecules in a Mouse Model of Chronic House Dust Mite Allergy. Int J Mol Sci 2024; 25:13661. [PMID: 39769423 PMCID: PMC11728213 DOI: 10.3390/ijms252413661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/10/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
Der p1 is one of the major allergens causing house dust mite (HDM) allergy. Pathological Der p1-specific B cells play a key role in allergic inflammation as producers of allergen-specific antibodies. Crosslinking the inhibitory FcγRIIb with the B cell receptor triggers a high-affinity suppressive signal in B cells. Selective elimination of allergen-specific cells could potentially be achieved by administering chimeric molecules that combine, through protein engineering, the FcγRIIb-targeting monoclonal 2.4G2 antibody with the epitope-carrying Dp52-71 peptides from Der p1. We tested this hypothesis, in a chronic mouse model of HDM allergy induced in BalB/c mice, using FACS and ELISA assays, along with histopathological and correlational analyses. Dp52-71chimera treatment of HDM-challenged mice led to a decrease in serum anti-HDM IgG1 antibodies, a reduction in BALF β-hexosaminidase levels, a lowered number of SiglecFhigh CD11clow eosinophils, and an improved lung PAS score. Furthermore, we observed overexpression of FcγRIIb on the surface of CD19 cells in the lungs of HDM-challenged animals, which negatively correlated with the levels of lung alveolar macrophages, neutrophils, and BALF IL-13. Taken together, these results suggest that the use of FcγRIIb overexpression, combined with the expansion of chimeric protein technology to include more epitopes, could improve the outcome of inflammation.
Collapse
Affiliation(s)
| | | | | | | | - Nikolina Mihaylova
- Department of Immunology, Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria; (N.R.); (S.B.); (N.K.); (A.T.)
| |
Collapse
|
4
|
Magnusen AF, Pandey MK. Complement System and Adhesion Molecule Skirmishes in Fabry Disease: Insights into Pathogenesis and Disease Mechanisms. Int J Mol Sci 2024; 25:12252. [PMID: 39596318 PMCID: PMC11594573 DOI: 10.3390/ijms252212252] [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] [Received: 09/12/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Fabry disease is a rare X-linked lysosomal storage disorder caused by mutations in the galactosidase alpha (GLA) gene, resulting in the accumulation of globotriaosylceramide (Gb3) and its deacetylated form, globotriaosylsphingosine (Lyso-Gb3) in various tissues and fluids throughout the body. This pathological accumulation triggers a cascade of processes involving immune dysregulation and complement system activation. Elevated levels of complement 3a (C3a), C5a, and their precursor C3 are observed in the plasma, serum, and tissues of patients with Fabry disease, correlating with significant endothelial cell abnormalities and vascular dysfunction. This review elucidates how the complement system, particularly through the activation of C3a and C5a, exacerbates disease pathology. The activation of these pathways leads to the upregulation of adhesion molecules, including vascular cell adhesion molecule 1 (VCAM1), intercellular adhesion molecule 1 (ICAM1), platelet and endothelial cell adhesion molecule 1 (PECAM1), and complement receptor 3 (CR3) on leukocytes and endothelial cells. This upregulation promotes the excessive recruitment of leukocytes, which in turn exacerbates disease pathology. Targeting complement components C3a, C5a, or their respective receptors, C3aR (C3a receptor) and C5aR1 (C5a receptor 1), could potentially reduce inflammation, mitigate tissue damage, and improve clinical outcomes for individuals with Fabry disease.
Collapse
Affiliation(s)
- Albert Frank Magnusen
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
| | - Manoj Kumar Pandey
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
- Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|
5
|
Schnabel CL, Jentsch MC, Lübke S, Kaiser-Thom S, Gerber V, Vrtala S, Huang HJ, Rhyner C, Wagner B, Hoffmann R, Volke D. Immunoproteomics reveal increased serum IgG3/5 binding to Dermatophagoides and yeast protein antigens in severe equine asthma in a preliminary study. Front Immunol 2023; 14:1293684. [PMID: 38162673 PMCID: PMC10754955 DOI: 10.3389/fimmu.2023.1293684] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/21/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction Severe equine asthma (SEA) is a common, chronic respiratory disease of horses characterized by hyperreactivity to hay dust which has many similarities to severe neutrophilic asthma in humans. SEA-provoking antigens have not been comprehensively characterized, but molds and mites have been suggested as relevant sources. Here, we identified relevant antigen candidates using immunoproteomics with IgG isotype-binding analyses. Methods Proteins from Dermatophagoides pteronyssinus (Der p) were separated by two-dimensional gel electrophoresis followed by immunoblotting (2D immunoblots) resulting in a characteristic pattern of 440 spots. After serum incubation, antibody (Ig)-binding of all Ig (Pan-Ig) and IgG isotypes (type-2-associated IgG3/5, type-1-associated IgG4/7) was quantified per each spot and compared between asthmatic and healthy horses' sera (n=5 per group). Results Ig binding differences were detected in 30 spots. Pan-Ig binding was higher with asthmatics compared to healthy horses' sera on four spots, and IgG3/5 binding was higher on 18 spots. Small IgG4/7 binding differences were detected on 10 spots with higher binding with asthmatics' sera on four but higher binding with healthy horses' sera on six spots. Proteins from the spots with group differences including mite and yeast proteins were identified by liquid chromatography mass spectrometry. The latter likely originated from the feeding substrate of the Der p culture. Prioritized antigen candidates amongst the proteins identified were Der p 1, Der p 11, group 15 allergens, myosin heavy chain, and uncharacterized Der p proteins. Additionally, yeast enolases, alcohol dehydrogenase (ADH), phosphoglycerate kinase (PGK), glyceraldehyde-3-phosphate dehydrogenase, and heat shock proteins were prioritized. Eleven antigen candidates were tested for confirmation by ELISAs using the respective proteins separately. Differences in asthmatics vs. healthy horses' serum Ig binding to Der p 1, Der p 18, and three yeast enzymes (enolase, ADH, and PGK) confirmed these as promising antigens of immune responses in SEA. Discussion Antigens with relevance in SEA were newly identified by immunoproteomics, and yeast antigens were considered for SEA for the first time. Serum IgG3/5 binding to relevant antigens was increased in SEA and is a novel feature that points to increased type-2 responses in SEA but requires confirmation of the corresponding cellular responses.
Collapse
Affiliation(s)
- Christiane L. Schnabel
- Institute of Immunology, Faculty of Veterinary Medicine, and Center for Biotechnology and Biomedicine, Leipzig University, Leipzig, Germany
| | - Maria-Christin Jentsch
- Institute of Immunology, Faculty of Veterinary Medicine, and Center for Biotechnology and Biomedicine, Leipzig University, Leipzig, Germany
| | - Sabrina Lübke
- Institute of Immunology, Faculty of Veterinary Medicine, and Center for Biotechnology and Biomedicine, Leipzig University, Leipzig, Germany
| | - Sarah Kaiser-Thom
- Swiss Institute of Equine Medicine (ISME), Department of Clinical Veterinary Medicine, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Vinzenz Gerber
- Swiss Institute of Equine Medicine (ISME), Department of Clinical Veterinary Medicine, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Susanne Vrtala
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Huey-Jy Huang
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Claudio Rhyner
- Christine Kühne Center for Allergy, Research, and Education (CK-CARE), Davos, Switzerland
- Swiss Institute of Allergy and Asthma Research (SIAF), Davos, Switzerland
| | - Bettina Wagner
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Ralf Hoffmann
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy and Center for Biotechnology and Biomedicine, Leipzig University, Leipzig, Germany
| | - Daniela Volke
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy and Center for Biotechnology and Biomedicine, Leipzig University, Leipzig, Germany
| |
Collapse
|
6
|
Udoye CC, Ehlers M, Manz RA. The B Cell Response and Formation of Allergenic and Anti-Allergenic Antibodies in Food Allergy. BIOLOGY 2023; 12:1501. [PMID: 38132327 PMCID: PMC10740584 DOI: 10.3390/biology12121501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/30/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023]
Abstract
Food allergies are a growing public health concern worldwide, especially in children and young adults. Allergen-specific IgE plays a central role in the pathogenesis of food allergies, but their titers poorly correlate with allergy development. Host immune systems yield allergen-specific immunoglobulin (Ig)A, IgE and IgG subclasses with low or high affinities and differential Fc N-glycosylation patterns that can affect the allergic reaction to food in multiple ways. High-affinity IgE is required to induce strong mast cell activation eventually leading to allergic anaphylaxis, while low-affinity IgE can even inhibit the development of clinically relevant allergic symptoms. IgA and IgG antibodies can inhibit IgE-mediated mast cell activation through various mechanisms, thereby protecting IgE-positive individuals from allergy development. The production of IgE and IgG with differential allergenic potential seems to be affected by the signaling strength of individual B cell receptors, and by cytokines from T cells. This review provides an overview of the diversity of the B cell response and the diverse roles of antibodies in food allergy.
Collapse
Affiliation(s)
- Christopher C. Udoye
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany
| | - Marc Ehlers
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, 23538 Lübeck, Germany
| | - Rudolf A. Manz
- Institute for Systemic Inflammation Research, University of Lübeck, 23562 Lübeck, Germany
| |
Collapse
|
7
|
Gokhale S, Victor E, Tsai J, Spirollari E, Matracz B, Takatsuka S, Jung J, Kitamura D, Xie P. Upregulated Expression of the IL-9 Receptor on TRAF3-Deficient B Lymphocytes Confers Ig Isotype Switching Responsiveness to IL-9 in the Presence of Antigen Receptor Engagement and IL-4. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1059-1073. [PMID: 36883978 PMCID: PMC10073299 DOI: 10.4049/jimmunol.2200563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 02/06/2023] [Indexed: 03/09/2023]
Abstract
The pleiotropic cytokine IL-9 signals to target cells by binding to a heterodimeric receptor consisting of the unique subunit IL-9R and the common subunit γ-chain shared by multiple cytokines of the γ-chain family. In the current study, we found that the expression of IL-9R was strikingly upregulated in mouse naive follicular B cells genetically deficient in TNFR-associated factor 3 (TRAF3), a critical regulator of B cell survival and function. The highly upregulated IL-9R on Traf3-/- follicular B cells conferred responsiveness to IL-9, including IgM production and STAT3 phosphorylation. Interestingly, IL-9 significantly enhanced class switch recombination to IgG1 induced by BCR crosslinking plus IL-4 in Traf3-/- B cells, which was not observed in littermate control B cells. We further demonstrated that blocking the JAK-STAT3 signaling pathway abrogated the enhancing effect of IL-9 on class switch recombination to IgG1 induced by BCR crosslinking plus IL-4 in Traf3-/- B cells. Our study thus revealed, to our knowledge, a novel pathway that TRAF3 suppresses B cell activation and Ig isotype switching by inhibiting IL-9R-JAK-STAT3 signaling. Taken together, our findings provide (to our knowledge) new insights into the TRAF3-IL-9R axis in B cell function and have significant implications for the understanding and treatment of a variety of human diseases involving aberrant B cell activation such as autoimmune disorders.
Collapse
Affiliation(s)
- Samantha Gokhale
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, New Jersey 08854
| | - Eton Victor
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854
| | - Jemmie Tsai
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854
| | - Eris Spirollari
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854
| | - Brygida Matracz
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854
| | - Shogo Takatsuka
- Division of Molecular Biology, Research Institute for Biomedical Sciences (RIBS), Tokyo University of Science, Noda, Japan
| | - Jaeyong Jung
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854
- Graduate Program in Cellular and Molecular Pharmacology, Rutgers University, Piscataway, New Jersey 08854
| | - Daisuke Kitamura
- Division of Molecular Biology, Research Institute for Biomedical Sciences (RIBS), Tokyo University of Science, Noda, Japan
| | - Ping Xie
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854
- Rutgers Cancer Institute of New Jersey
| |
Collapse
|
8
|
Makhijani P, Basso PJ, Chan YT, Chen N, Baechle J, Khan S, Furman D, Tsai S, Winer DA. Regulation of the immune system by the insulin receptor in health and disease. Front Endocrinol (Lausanne) 2023; 14:1128622. [PMID: 36992811 PMCID: PMC10040865 DOI: 10.3389/fendo.2023.1128622] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/08/2023] [Indexed: 03/14/2023] Open
Abstract
The signaling pathways downstream of the insulin receptor (InsR) are some of the most evolutionarily conserved pathways that regulate organism longevity and metabolism. InsR signaling is well characterized in metabolic tissues, such as liver, muscle, and fat, actively orchestrating cellular processes, including growth, survival, and nutrient metabolism. However, cells of the immune system also express the InsR and downstream signaling machinery, and there is increasing appreciation for the involvement of InsR signaling in shaping the immune response. Here, we summarize current understanding of InsR signaling pathways in different immune cell subsets and their impact on cellular metabolism, differentiation, and effector versus regulatory function. We also discuss mechanistic links between altered InsR signaling and immune dysfunction in various disease settings and conditions, with a focus on age related conditions, such as type 2 diabetes, cancer and infection vulnerability.
Collapse
Affiliation(s)
- Priya Makhijani
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Buck Institute for Research in Aging, Novato, CA, United States
| | - Paulo José Basso
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Yi Tao Chan
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Nan Chen
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jordan Baechle
- Buck Institute for Research in Aging, Novato, CA, United States
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, United States
| | - Saad Khan
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
| | - David Furman
- Buck Institute for Research in Aging, Novato, CA, United States
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, United States
- Stanford 1, 000 Immunomes Project, Stanford School of Medicine, Stanford University, Stanford, CA, United States
- Instituto de Investigaciones en Medicina Traslacional (IIMT), Universidad Austral, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Pilar, Argentina
| | - Sue Tsai
- Department of Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Daniel A. Winer
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Buck Institute for Research in Aging, Novato, CA, United States
- Division of Cellular and Molecular Biology, Diabetes Research Group, Toronto General Hospital Research Institute (TGHRI), University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Buck Artificial Intelligence Platform, Buck Institute for Research on Aging, Novato, CA, United States
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
9
|
Trivedi VS, Magnusen AF, Rani R, Marsili L, Slavotinek AM, Prows DR, Hopkin RJ, McKay MA, Pandey MK. Targeting the Complement-Sphingolipid System in COVID-19 and Gaucher Diseases: Evidence for a New Treatment Strategy. Int J Mol Sci 2022; 23:14340. [PMID: 36430817 PMCID: PMC9695449 DOI: 10.3390/ijms232214340] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/22/2022] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2)-induced disease (COVID-19) and Gaucher disease (GD) exhibit upregulation of complement 5a (C5a) and its C5aR1 receptor, and excess synthesis of glycosphingolipids that lead to increased infiltration and activation of innate and adaptive immune cells, resulting in massive generation of pro-inflammatory cytokines, chemokines and growth factors. This C5a-C5aR1-glycosphingolipid pathway- induced pro-inflammatory environment causes the tissue damage in COVID-19 and GD. Strikingly, pharmaceutically targeting the C5a-C5aR1 axis or the glycosphingolipid synthesis pathway led to a reduction in glycosphingolipid synthesis and innate and adaptive immune inflammation, and protection from the tissue destruction in both COVID-19 and GD. These results reveal a common involvement of the complement and glycosphingolipid systems driving immune inflammation and tissue damage in COVID-19 and GD, respectively. It is therefore expected that combined targeting of the complement and sphingolipid pathways could ameliorate the tissue destruction, organ failure, and death in patients at high-risk of developing severe cases of COVID-19.
Collapse
Affiliation(s)
- Vyoma Snehal Trivedi
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
| | - Albert Frank Magnusen
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
| | - Reena Rani
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
| | - Luca Marsili
- Department of Neurology, James J. and Joan A. Gardner Center for Parkinson’s Disease and Movement Disorders, University of Cincinnati, 3113 Bellevue Ave, Cincinnati, OH 45219, USA
| | - Anne Michele Slavotinek
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, 3230 Eden Ave, Cincinnati, OH 45267, USA
| | - Daniel Ray Prows
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, 3230 Eden Ave, Cincinnati, OH 45267, USA
| | - Robert James Hopkin
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, 3230 Eden Ave, Cincinnati, OH 45267, USA
| | - Mary Ashley McKay
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
| | - Manoj Kumar Pandey
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, 3333 Burnet Avenue, Building R1, MLC 7016, Cincinnati, OH 45229, USA
- Department of Pediatrics, College of Medicine, University of Cincinnati, 3230 Eden Ave, Cincinnati, OH 45267, USA
| |
Collapse
|
10
|
Exploring the Potential Effects and Mechanisms of Asarum sieboldii Radix Essential Oil for Treatment of Asthma. Pharmaceutics 2022; 14:pharmaceutics14030558. [PMID: 35335934 PMCID: PMC8953372 DOI: 10.3390/pharmaceutics14030558] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 02/05/2023] Open
Abstract
Asthma, a common chronic pulmonary disorder characterized by airway remodeling, hyperresponsiveness and obstruction, can be aggravated by repeated exposure to particulate matter (PM). The potential effect and mechanisms of Asarum sieboldii Radix essential oil (AEO) against asthma were explored based on network pharmacology. AEO was pre-treated using a nebulizer for 3 weeks and the mice were sensitized to ovalbumin (OVA) and PM10 with the co-treatment of AEO for 4 weeks. In addition, A549 lung epithelial cells were sensitized with PM10 to investigate the underlying mechanisms of AEO regarding the lung-fibrosis-related mediators. The target genes of methyl eugenol, a main compound of AEO, were highly matched by 48% with the gene set of asthma. AEO markedly inhibited the increase in epithelial thickness through the accumulation of goblet cells in the airways. Collagen deposition in the lung tissues of OVA+PM10-challenged asthmatic mice was significantly decreased by AEO. AEO also inhibited the influx of inflammatory cells in the bronchoalveolar lavage fluid, as well as the increases in serum IgE and IgG2a and cytokines in the lung tissues. Furthermore, AEO regulated the expressions of fibrotic mediators, especially POSTN and TGF-β. In conclusion, we expect that AEO can be one of the effective alternative therapeutics to relieve asthma.
Collapse
|
11
|
Tomonaga T, Izumi H, Nishida C, Kato K, Yatera K, Kuroda E, Morimoto Y. Suppression of Airway Allergic Reactions by a Photocatalytic Filter Using Mouse Model. TOXICS 2022; 10:toxics10010040. [PMID: 35051082 PMCID: PMC8781178 DOI: 10.3390/toxics10010040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 12/10/2022]
Abstract
Photocatalytic filters installed in air purifiers have been used to purify spaces by decomposing allergenic substances. However, we have not found any reports that evaluate the effectiveness of photocatalytic filters in suppressing allergic reactions in living organisms. In this study, we intratracheally instilled ovalbumin (OVA) into OVA-sensitized mice after the OVA was photocatalyzed by a titanium dioxide (TiO2) filter, and verified the experimental model for evaluating the allergy-suppressing effect of photocatalysts. Mice were sensitized to OVA (10 µg/mouse) four times, and were intratracheally instilled with OVA (10 µg/mouse) after photocatalysis three times. Non-sensitized animals were instilled with normal saline following the same exposure schedule. The mice were dissected 24 h after final exposure. The OVA after photocatalysis significantly decreased the number of eosinophils in bronchoalveolar lavage fluid, and the concentration of OVA-specific IgE and IgG1 in serum, which were elevated in untreated OVA. Moreover, our experimental model showed the suppression of allergic reactions in mice, along with the decomposition of OVA after photocatalysis using the photocatalytic filter. Taken together, our experimental model for evaluating allergic reactions in the respiratory tract suggested that the allergy-suppressing effect of the photocatalytic filter can be evaluated.
Collapse
Affiliation(s)
- Taisuke Tomonaga
- Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu 807-8555, Japan; (H.I.); (Y.M.)
- Correspondence: ; Tel.: +81-93-691-7466; Fax: +81-93-691-4284
| | - Hiroto Izumi
- Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu 807-8555, Japan; (H.I.); (Y.M.)
| | - Chinatsu Nishida
- Department of Respiratory Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu 807-8555, Japan; (C.N.); (K.K.); (K.Y.)
| | - Kaori Kato
- Department of Respiratory Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu 807-8555, Japan; (C.N.); (K.K.); (K.Y.)
| | - Kazuhiro Yatera
- Department of Respiratory Medicine, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu 807-8555, Japan; (C.N.); (K.K.); (K.Y.)
| | - Etsushi Kuroda
- Department of Immunology, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya 663-8131, Japan;
| | - Yasuo Morimoto
- Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu 807-8555, Japan; (H.I.); (Y.M.)
| |
Collapse
|
12
|
Montamat G, Leonard C, Poli A, Klimek L, Ollert M. CpG Adjuvant in Allergen-Specific Immunotherapy: Finding the Sweet Spot for the Induction of Immune Tolerance. Front Immunol 2021; 12:590054. [PMID: 33708195 PMCID: PMC7940844 DOI: 10.3389/fimmu.2021.590054] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/04/2021] [Indexed: 01/16/2023] Open
Abstract
Prevalence and incidence of IgE-mediated allergic diseases have increased over the past years in developed and developing countries. Allergen-specific immunotherapy (AIT) is currently the only curative treatment available for allergic diseases that has long-term efficacy. Although AIT has been proven successful as an immunomodulatory therapy since its beginnings, it still faces several unmet needs and challenges today. For instance, some patients can experience severe side effects, others are non-responders, and prolonged treatment schedules can lead to lack of patient adherence and therapy discontinuation. A common strategy to improve AIT relies on the use of adjuvants and immune modulators to boost its effects and improve its safety. Among the adjuvants tested for their clinical efficacy, CpG oligodeoxynucleotide (CpG-ODN) was investigated with limited success and without reaching phase III trials for clinical allergy treatment. However, recently discovered immune tolerance-promoting properties of CpG-ODN place this adjuvant again in a prominent position as an immune modulator for the treatment of allergic diseases. Indeed, it has been shown that the CpG-ODN dose and concentration are crucial in promoting immune regulation through the recruitment of pDCs. While low doses induce an inflammatory response, high doses of CpG-ODN trigger a tolerogenic response that can reverse a pre-established allergic milieu. Consistently, CpG-ODN has also been found to stimulate IL-10 producing B cells, so-called B regulatory cells (Bregs). Accordingly, CpG-ODN has shown its capacity to prevent and revert allergic reactions in several animal models showing its potential as both preventive and active treatment for IgE-mediated allergy. In this review, we describe how CpG-ODN-based therapies for allergic diseases, despite having shown limited success in the past, can still be exploited further as an adjuvant or immune modulator in the context of AIT and deserves additional attention. Here, we discuss the past and current knowledge, which highlights CpG-ODN as a potential adjuvant to be reevaluated for the enhancement of AIT when used in appropriate conditions and formulations.
Collapse
Affiliation(s)
- Guillem Montamat
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Cathy Leonard
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Aurélie Poli
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Ludger Klimek
- Centre for Rhinology and Allergology, Wiesbaden, Germany
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Department of Dermatology and Allergy Centre, Odense University Hospital, Odense, Denmark
| |
Collapse
|
13
|
Lin J, Huang N, Li J, Liu X, Xiong Q, Hu C, Chen D, Guan L, Chang K, Li D, Tsui SKW, Zhong N, Liu Z, Yang PC. Cross-reactive antibodies against dust mite-derived enolase induce neutrophilic airway inflammation. Eur Respir J 2021; 57:13993003.02375-2019. [PMID: 32817257 DOI: 10.1183/13993003.02375-2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 07/30/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Neutrophilic inflammation is a hallmark of some specific asthma phenotypes; its aetiology is not yet fully understood. House dust mite (HDM) is the most common factor in the pathogenesis of airway inflammation. This study aims to elucidate the role of cross-antibodies against HDM-derived factors in the development of neutrophilic inflammation in the airway. METHODS Blood samples were collected from asthma patients with chronic neutrophilic asthma for analysis of HDM-specific cross-reactive antibodies. The role of an antibody against HDM-derived enolase (EnoAb) in the impairment of airway epithelial barrier function and induction of airway inflammation was assessed in a cell culture model and an animal model. RESULTS High similarity (72%) of the enolase gene sequences was identified between HDM and human. Serum EnoAb was detected in patients with chronic neutrophilic asthma. The EnoAb bound to airway epithelial cells to form complexes with enolase, which activated complement, impaired airway epithelial barrier functions and induced neutrophilic inflammation in the airway tissues. CONCLUSIONS HDM-derived enolase can induce specific cross-antibodies in humans, which induce neutrophilic inflammation in the airway.
Collapse
Affiliation(s)
- Jianli Lin
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen University School of Medicine, Shenzhen, China.,State Key Laboratory of Respiratory Disease, National Clinical Center for Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,These authors contributed equally to this work
| | - Nana Huang
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen University School of Medicine, Shenzhen, China.,These authors contributed equally to this work
| | - Jing Li
- State Key Laboratory of Respiratory Disease, National Clinical Center for Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,These authors contributed equally to this work
| | - Xiaoyu Liu
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen University School of Medicine, Shenzhen, China.,These authors contributed equally to this work
| | - Qing Xiong
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Chengshen Hu
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen University School of Medicine, Shenzhen, China
| | - Desheng Chen
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen University School of Medicine, Shenzhen, China
| | - Lvxin Guan
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen University School of Medicine, Shenzhen, China
| | - Kexin Chang
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen University School of Medicine, Shenzhen, China
| | - Dan Li
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen University School of Medicine, Shenzhen, China
| | | | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Center for Respiratory Diseases, Guangzhou Institute of Respiratory Diseases, First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Nanshan Zhong, Zhigang Liu and Ping-Chang Yang contributed equally to this article as lead authors and supervised the work
| | - Zhigang Liu
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen University School of Medicine, Shenzhen, China.,Nanshan Zhong, Zhigang Liu and Ping-Chang Yang contributed equally to this article as lead authors and supervised the work
| | - Ping-Chang Yang
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen University School of Medicine, Shenzhen, China.,Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China.,Nanshan Zhong, Zhigang Liu and Ping-Chang Yang contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
14
|
Blöchl C, Regl C, Huber CG, Winter P, Weiss R, Wohlschlager T. Towards middle-up analysis of polyclonal antibodies: subclass-specific N-glycosylation profiling of murine immunoglobulin G (IgG) by means of HPLC-MS. Sci Rep 2020; 10:18080. [PMID: 33093535 PMCID: PMC7581757 DOI: 10.1038/s41598-020-75045-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 10/09/2020] [Indexed: 12/21/2022] Open
Abstract
In recent years, advanced HPLC-MS strategies based on intact protein (“top-down”) or protein subunit (“middle-up/middle-down”) analysis have been implemented for the characterization of therapeutic monoclonal antibodies. Here, we assess feasibility of middle-up/middle-down analysis for polyclonal IgGs exhibiting extensive sequence variability. Specifically, we addressed IgGs from mouse, representing an important model system in immunological investigations. To obtain Fc/2 portions as conserved subunits of IgGs, we made use of the bacterial protease SpeB. For this purpose, we initially determined SpeB cleavage sites in murine IgGs. The resulting Fc/2 portions characteristic of different subclasses were subsequently analysed by ion-pair reversed-phase HPLC hyphenated to high-resolution mass spectrometry. This enabled simultaneous relative quantification of IgG subclasses and their N-glycosylation variants, both of which influence IgG effector functions. To assess method capabilities in an immunological context, we applied the analytical workflow to polyclonal antibodies obtained from BALB/c mice immunized with the grass pollen allergen Phl p 6. The study revealed a shift in IgG subclasses and Fc-glycosylation patterns in total and antigen-specific IgGs from different mouse cohorts, respectively. Eventually, Fc/2 characterization may reveal other protein modifications including oxidation, amino acid exchanges, and C-terminal lysine, and may thus be implemented for quality control of functional antibodies.
Collapse
Affiliation(s)
- Constantin Blöchl
- Department of Biosciences, Bioanalytical Research Labs, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria
| | - Christof Regl
- Department of Biosciences, Bioanalytical Research Labs, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria.,Christian Doppler Laboratory for Innovative Tools for Biosimilar Characterization, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria
| | - Christian G Huber
- Department of Biosciences, Bioanalytical Research Labs, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria.,Christian Doppler Laboratory for Innovative Tools for Biosimilar Characterization, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria
| | - Petra Winter
- Department of Biosciences, Division of Allergy and Immunology, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria
| | - Richard Weiss
- Department of Biosciences, Division of Allergy and Immunology, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria
| | - Therese Wohlschlager
- Department of Biosciences, Bioanalytical Research Labs, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria. .,Christian Doppler Laboratory for Innovative Tools for Biosimilar Characterization, University of Salzburg, Hellbrunner Straße 34, 5020, Salzburg, Austria.
| |
Collapse
|
15
|
Hu W, Ma L, Yang G, Zeng X, Liu J, Cheng B, Hu T, Zhao H, Liu Z. Der p2‑A20 DNA vaccine attenuates allergic inflammation in mice with allergic rhinitis. Mol Med Rep 2019; 20:4925-4932. [PMID: 31638224 PMCID: PMC6854600 DOI: 10.3892/mmr.2019.10760] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 08/30/2019] [Indexed: 02/07/2023] Open
Abstract
Allergic rhinitis (AR) is a common disease that requires more convenient, safe and effective antigen-specific immunotherapies. The present study aimed to investigate the therapeutic effect of intranasal administration of a eukaryotic expression vector co-expressing Der p2 and A20 protein (pVAX1-Der p2-A20) on mice with allergic rhinitis. The pVAX1-Der p2-A20 vaccine was prepared and encapsulated into poly(L-lactide-co-glycolide) (PLGA) nanoparticles. An allergic rhinitis Balb/c mouse model was established through intraperitoneal sensitization with recombinant Der p2 and cholera toxin followed by intranasal challenge with recombinant Der p2. The treatment effect of the DNA vaccine on nasal allergic inflammation was evaluated, and serum IgE, sIgE, IgG and cytokine levels were determined by ELISA. The percentage of CD4+CD25+Foxp3+Tregs in the spleen was detected by flow cytometry. The DNA vaccine co-expressing Der p2 and A20 was successfully constructed and encapsulated into PLGA nanoparticles. Der p2-A20 DNA vaccine intranasal administration markedly ameliorated Der p2-induced nasal allergic inflammation. The serum Der p2-specific IgE, IL-4 and IL-13 expression levels were inhibited, while the Der p2-specific IgG1, IgG2a and IFN-γ expression levels in the serum and splenic CD4+CD25+Foxp3+Treg population were significantly increased after Der p2-A20 DNA vaccine treatment. These results indicated that the Der p2-A20 DNA vaccine alleviates nasal allergic inflammation and promotes splenic Treg population in mice with allergic rhinitis.
Collapse
Affiliation(s)
- Wenhui Hu
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, Guangdong 518172, P.R. China
| | - Li Ma
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, Guangdong 518172, P.R. China
| | - Gui Yang
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, Guangdong 518172, P.R. China
| | - Xianhai Zeng
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, Guangdong 518172, P.R. China
| | - Jiangqi Liu
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, Guangdong 518172, P.R. China
| | - Baohui Cheng
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, Guangdong 518172, P.R. China
| | - Tianyong Hu
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, Guangdong 518172, P.R. China
| | - Hailiang Zhao
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, Guangdong 518172, P.R. China
| | - Zhiqiang Liu
- Department of Otolaryngology, Longgang ENT Hospital and Shenzhen Key Laboratory of ENT, Institute of ENT, Shenzhen, Guangdong 518172, P.R. China
| |
Collapse
|
16
|
Veloso-Teles R, Cerejeira R, Rodrigues D, Roque-Farinha R, von Buchwald C. Food-Specific IgE and IgG Antibodies in Patients With Chronic Rhinosinusitis With Nasal Polyps: A Case-Control Study. EAR, NOSE & THROAT JOURNAL 2019; 100:177-184. [PMID: 31547710 DOI: 10.1177/0145561319867668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
EPOS 2012 states that investigation is needed to study a possible role for food allergy in the initiation and perpetuation of chronic rhinosinusitis with nasal polyps (CRSwNP). Our main goal was to compare serum levels of food-specific immunoglobulin G (IgG) and IgE antibodies in patients with CRSwNP and controls. A prospective case-control study with 33 patients with CRSwNP and 31 controls without CRS was carried out. Clinical data were gathered through a systematic interview and blood sample was collected. Enzyme-linked immunosorbent assay tests using OmegaDiagnostics kit with 40 food allergens for detection of specific IgG antibodies were performed and food-specific IgE antibodies were determined by immunoassay using ImmunoCAP. Immunoglobulin classes and IgG subclasses levels were also evaluated. Statistical analysis was performed using SPSS v.23. The overall sum of food IgG antibodies was significantly lower in CRSwNP compared to control group, and this difference was also observed for different specific IgG antibodies (corn, soya, grain legumes, pear and apple, berries, citric fruit). In controls, a positive correlation between IgG1 and the sum of food IgG antibodies was seen, but in CRSwNP group a negative correlation was found. In addition, a significant higher level of IgG1 and lower IgG2 and IgG3 was found among patients with CRSwNP. Levels of serum-specific IgE antibodies against multiallergen food mix (fx5) and against shrimp, strawberry, orange, rye, or egg yolk, as well as the sum of food IgE antibodies, did not differ significantly between the groups. These findings suggest that food allergy does not have an important role in CRSwNP etiopathogenesis, whether it is IgG or IgE mediated. Moreover, the observed suppression of specific IgG antibodies against food allergens, its negative correlation with IgG1 and the IgG1 switching in CRSwNP, can be related to deviated IgG responses against other targets (eg, airborne particles) and warrants future investigation.
Collapse
Affiliation(s)
- Rafaela Veloso-Teles
- Faculty of Health Sciences, 70987University of Beira Interior, Covilhã, Portugal.,Department of Otorhinolaryngology, 162181Cova da Beira Hospital Centre (CHCB), Covilhã, Portugal.,Otorhinolaryngology Unit of Beira Interior (UOBI), Covilhã, Portugal
| | - Rui Cerejeira
- Faculty of Health Sciences, 70987University of Beira Interior, Covilhã, Portugal.,Department of Otorhinolaryngology, 162181Cova da Beira Hospital Centre (CHCB), Covilhã, Portugal.,Otorhinolaryngology Unit of Beira Interior (UOBI), Covilhã, Portugal
| | - Débora Rodrigues
- Department of Clinical Pathology, 162181Cova da Beira Hospital Centre (CHCB), Covilhã, Portugal
| | - Rosa Roque-Farinha
- Faculty of Health Sciences, 70987University of Beira Interior, Covilhã, Portugal
| | - Christian von Buchwald
- Department of Otorhinolaryngology, Head and Neck Surgery and Audiology, Rigshospitalet-Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
17
|
Prangtaworn P, Chaisri U, Seesuay W, Mahasongkram K, Onlamoon N, Reamtong O, Tungtrongchitr A, Indrawattana N, Chaicumpa W, Sookrung N. Tregitope-linked Refined Allergen Vaccines for Immunotherapy in Cockroach Allergy. Sci Rep 2018; 8:15480. [PMID: 30341299 PMCID: PMC6195530 DOI: 10.1038/s41598-018-33680-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 10/01/2018] [Indexed: 01/17/2023] Open
Abstract
Allergen-specific immunotherapy (AIT) facilitates long-term resolution of allergic morbidity resulting in reduced drug use and increased refractoriness to new sensitization. AIT effectiveness has been demonstrated in seasonal and perennial allergies, and insect stings. However, data and studies in AIT relative to cockroach (CR) allergy are relatively scarce. In this study, mice allergic to American CR (Periplaneta americana) were treated with a liposome (L)-entrapped vaccine made of mouse Tregitope289-Per a 9 of the CR, Tregitope167-Per a 9, or Per a 9 alone - or placebo. Allergic mice that received an individual vaccine intranasally had reduced Th2 response, reduced lung inflammation, and reduced respiratory tissue remodeling. However, only L-Tregitope289-Per a 9 and L-Tregitope167-Per a 9 induced expression of immunosuppressive cytokine genes (IL-10, TGF-β, and IL-35 for L-Tregitope289-Per a 9, and IL-10 and TGF-β for L-Tregitope167-Per a 9) and increment of idoleamine-2,3-dioxygenase 1 (IDO1), indicating that these vaccines caused allergic disease suppression and reversal of respiratory tissue remodeling via generation of regulatory lymphocytes. Liposome entrapped-recombinant Per a 9 (L-Per a 9) did not cause upregulation of immunosuppressive cytokine genes and IDO1 increment; rather, L-Per a 9 induced high expression of IFN-γ in lungs of treated mice, which resulted in mitigation of allergic manifestations. This study provides compelling evidence that both liposome-entrapped vaccines made of single refined major allergen alone and single refined major allergen linked with Tregitopes are effective for reducing allergen-mediated respiratory tissue inflammation and remodeling, but through different mechanisms.
Collapse
Affiliation(s)
- Pannathee Prangtaworn
- Graduate Program in Immunology, Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Urai Chaisri
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Watee Seesuay
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kodchakorn Mahasongkram
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nattawat Onlamoon
- Biomedical Research Incubator Unit, Department of Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Onrapak Reamtong
- Department of Tropical Molecular Biology and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Anchalee Tungtrongchitr
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nitaya Indrawattana
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wanpen Chaicumpa
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nitat Sookrung
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
- Biomedical Research Incubator Unit, Department of Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
18
|
Zhang J, Chen J, Zuo J, Newton GK, Stewart MR, Perrior TR, Garrod DR, Robinson C. Allergen Delivery Inhibitors: Characterisation of Potent and Selective Inhibitors of Der p 1 and Their Attenuation of Airway Responses to House Dust Mite Allergens. Int J Mol Sci 2018; 19:E3166. [PMID: 30326568 PMCID: PMC6214017 DOI: 10.3390/ijms19103166] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/05/2018] [Accepted: 10/05/2018] [Indexed: 12/21/2022] Open
Abstract
Group 1 allergens of house dust mites (HDM) are globally significant triggers of allergic disease. They are considered as initiator allergens because their protease activity enables the development of allergy to a spectrum of unrelated allergens from various sources. This initiator-perpetuator function identifies Group 1 HDM allergens as attractive drug design targets for the first small-molecule approach directed towards a non-human, root cause trigger of allergic disease. The purpose of this study was to: (i) identify exemplar inhibitors of these allergens using Der p 1 as a design template, and (ii) characterise the pharmacological profiles of these compounds using in vitro and in vivo models relevant to allergy. Potent inhibitors representing four different chemotypes and differentiated by mechanism of action were investigated. These compounds prevented the ab initio development of allergy to the full spectrum of HDM allergens and in established allergy they inhibited the recruitment of inflammatory cells and blunted acute allergic bronchoconstriction following aerosol challenge with the full HDM allergen repertoire. Collectively, the data obtained in these experiments demonstrate that the selective pharmacological targeting of Der p 1 achieves an attractive range of benefits against exposure to all HDM allergens, consistent with the initiator-perpetuator function of this allergen.
Collapse
Affiliation(s)
- Jihui Zhang
- Institute for Infection & Immunity, St George's, University of London, Cranmer Terrace, London SW17 0RE, UK.
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Jie Chen
- Institute for Infection & Immunity, St George's, University of London, Cranmer Terrace, London SW17 0RE, UK.
| | - Jie Zuo
- Institute for Infection & Immunity, St George's, University of London, Cranmer Terrace, London SW17 0RE, UK.
- BOE Technology Center, BOE Technology Group Co., Ltd., Beijing 100176, China.
| | - Gary K Newton
- Domainex Ltd., Chesterford Research Park, Little Chesterford, Saffron Walden, Essex CB10 1XL, UK.
| | - Mark R Stewart
- Domainex Ltd., Chesterford Research Park, Little Chesterford, Saffron Walden, Essex CB10 1XL, UK.
| | - Trevor R Perrior
- Domainex Ltd., Chesterford Research Park, Little Chesterford, Saffron Walden, Essex CB10 1XL, UK.
| | - David R Garrod
- Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PL, UK.
| | - Clive Robinson
- Institute for Infection & Immunity, St George's, University of London, Cranmer Terrace, London SW17 0RE, UK.
| |
Collapse
|
19
|
Peng X, Liang Y, Yin Y, Liao H, Li L. Development of a hollow mesoporous silica nanoparticles vaccine to protect against house dust mite induced allergic inflammation. Int J Pharm 2018; 549:115-123. [PMID: 30040973 DOI: 10.1016/j.ijpharm.2018.07.047] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 06/24/2018] [Accepted: 07/20/2018] [Indexed: 01/24/2023]
Abstract
Allergen specific immunotherapy (SIT) is the only specific therapeutic way for house dust mite (HDM) allergy. To improve the efficacy of SIT, hollow mesoporous silica nanoparticles (HMSNs) were used as vehicles for HDM allergen. The HMSNs were prepared and characterized. The major HDM allergen (Der f2) was loaded onto HMSNs, and the drug loading capacity and release profile were determined. Then the Der f2 loaded HMSNs were injected subcutaneously to mouse model of Der f2 induced allergic asthma and the preventive effects were evaluated. Our results showed that HMSNs were spherical (100 nm) with pore diameter of 2.897 nm and successfully loaded with Der f2 protein. The loading capacity is 90 μg Der f2/1 mg HMSNs. The Der f2 loaded on HMSNs released slowly in 72 h. Treatment with Der f2 loaded HMSNs could efficiently decrease Der f2 specific IgE levels, inflammatory cells infiltration in lung tissue, and Th2 cytokine IL4 levels in BALF. In the meanwhile, it could increase the Der f2 specific IgG levels, Th1 cytokine IFN-γ levels, and induce proliferation of splenocytes to Der f2 accompanied by increased IFN-γ levels. These results showed that Der f2 loaded HMSNs were efficient in preventing allergic inflammation, and HMSNs may be potential vehicles for SIT of HDM allergy.
Collapse
Affiliation(s)
- Xia Peng
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, 100 Haining Road, Shanghai, China
| | - Yuting Liang
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, 100 Haining Road, Shanghai, China
| | - Yue Yin
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, 100 Haining Road, Shanghai, China
| | - Huanjin Liao
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, 100 Haining Road, Shanghai, China
| | - Li Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, 100 Haining Road, Shanghai, China.
| |
Collapse
|
20
|
Allergic patients with and without allergen-specific immunotherapy mount protective immune responses to tick-borne encephalitis vaccination in absence of enhanced side effects or propagation of their Th2 bias. Vaccine 2018; 36:2816-2824. [PMID: 29673942 DOI: 10.1016/j.vaccine.2018.03.076] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 03/09/2018] [Accepted: 03/28/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Allergic diseases are caused by Th2-driven immune responses and their treatment with specific immunotherapy (SIT) leads to immunomodulation via IL10, TGF-ß and Th1/Tr1 shift. This phase IV, open-label clinical trial investigated whether allergies and SIT treatment influenced immune responses to routine vaccination. METHODS We studied three groups: 49 allergic patients (allergic group), 21 allergic patients receiving maintenance doses of SIT (SIT group), and 49 non-allergic controls. All subjects received tick-borne encephalitis (TBE) booster vaccines and humoral and cellular immune responses were evaluated after one week, four weeks and six months. RESULTS The levels and kinetics of neutralizing TBE-specific antibodies, reflecting protection against TBE, were not significantly different in the three groups. The allergic group showed Th2 polarization pre-booster as indicated by increased TBE-specific IgG1 and elevated mitogen-induced IL5 production. Alum-adjuvanted TBE vaccine led to Th2 biased immune responses in the controls, but to no further enhancement of Th2 polarization in the allergic and SIT group. Furthermore, in the SIT group cellular parameters reflected the induction of immunomodulation due to increased Tregs, elevated baseline IL10 and lack of TBE-specific IL5. Importantly, these cellular regulatory responses did not limit the ability to mount sufficient TBE-specific antibodies after the booster. All groups tolerated the vaccine well with no exacerbation of allergic symptoms. CONCLUSION TBE booster vaccinations were immunogenic and safe in both the allergic and SIT group and contributed to balanced immune responses. Our data indicate that all allergic patients, even when undergoing SIT, should be vaccinated without hesitation and at regular intervals according to standard recommendations. ClinicalTrials.gov (NCT02511535).
Collapse
|
21
|
Pandey MK, Grabowski GA, Köhl J. An unexpected player in Gaucher disease: The multiple roles of complement in disease development. Semin Immunol 2018; 37:30-42. [PMID: 29478824 DOI: 10.1016/j.smim.2018.02.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 12/14/2022]
Abstract
The complement system is well appreciated for its role as an important effector of innate immunity that is activated by the classical, lectin or alternative pathway. C5a is one important mediator of the system that is generated in response to canonical and non-canonical C5 cleavage by circulating or cell-derived proteases. In addition to its function as a chemoattractant for neutrophils and other myeloid effectors, C5a and its sister molecule C3a have concerted roles in cell homeostasis and surveillance. Through activation of their cognate G protein coupled receptors, C3a and C5a regulate multiple intracellular pathways within the mitochondria and the lysosomal compartments that harbor multiple enzymes critical for protein, carbohydrate and lipid metabolism. Genetic mutations of such lysosomal enzymes or their receptors can result in the compartmental accumulation of specific classes of substrates in this organelle summarized as lysosomal storage diseases (LSD). A frequent LSD is Gaucher disease (GD), caused by autosomal recessively inherited mutations in GBA1, resulting in functional defects of the encoded enzyme, acid β-glucosidase (glucocerebrosidase, GCase). Such mutations promote excessive accumulation of β-glucosylceramide (GC or GL1) in innate and adaptive immune cells frequently associated with chronic inflammation. Recently, we uncovered an unexpected link between the C5a and C5a receptor 1 (C5aR1) axis and the accumulation of GL1 in experimental and clinical GD. Here, we will review the pathways of complement activation in GD, its role as a mediator of the inflammatory response, and its impact on glucosphingolipid metabolism. Further, we will discuss the potential role of the C5a/C5aR1 axis in GL1-specific autoantibody formation and as a novel therapeutic target in GD.
Collapse
Affiliation(s)
- Manoj K Pandey
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA; The Department of Pediatrics of the University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA.
| | - Gregory A Grabowski
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA; The Department of Pediatrics of the University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Jörg Köhl
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA; The Department of Pediatrics of the University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA; Institute for Systemic Inflammation Research, University of Lübeck, 23562, Lübeck, Germany.
| |
Collapse
|
22
|
Ma Y, Dawicki W, Zhang X, Gordon JR. Contributions of direct versus indirect mechanisms for regulatory dendritic cell suppression of asthmatic allergen-specific IgG1 antibody responses. PLoS One 2018; 13:e0190414. [PMID: 29293622 PMCID: PMC5749789 DOI: 10.1371/journal.pone.0190414] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 12/14/2017] [Indexed: 12/23/2022] Open
Abstract
IL-10-differentiated dendritic cells (DC10) can reverse the asthma phenotype in mice, but how they suppress the asthmatic B cell response is unclear. Herein we assessed the mechanism(s) by which DC10 and DC10-induced Treg affect IgG1 production in asthma. We observed a rapid decline in lung-resident OVA-specific IgG1-secreting B cells on cessation of airway allergen challenge, and intraperitoneal DC10 therapy did not amplify that (p>0.05). It did however increase the loss of IgG1-B cells from the bone marrow (by 45+/-7.2%; p≤0.01) and spleen (by 65+/-17.8%; p≤0.05) over 2 wk. Delivery of OVA-loaded DC10 directly into the airways of asthmatic mice decreased the lung IgG1 B cell response assessed 2 dy later by 33+/-9.7% (p≤0.01), while their co-culture with asthmatic lung cell suspensions reduced the numbers of IgG1-secreting cells by 56.5+/-9.7% (p≤0.01). This effect was dependent on the DC10 carrying intact allergen on their cell surface; DC10 that had phagocytosed and fully processed their allergen were unable to suppress B cell responses, although they did suppress asthmatic Th2 cell responses. We had shown that therapeutic delivery of DC10-induced Treg can effectively suppress asthmatic T and B cell (IgE and IgG1) responses; herein CD4+ cells or Treg from the lungs of DC10-treated OVA-asthmatic mice suppressed in vitro B cell IgG1 production by 52.2+/-8.7% (p≤0.001) or 44.6+/-12.2% (p≤0.05), respectively, but delivery of DC10-induced Treg directly into the airways of asthmatic mice had no discernible impact over 2 dy on the numbers of lung IgG1-secreting cells (p≥0.05). In summary, DC10 treatment down-regulates OVA-specific B cell responses of asthmatic mice. While DC10 that carry intact allergen on their cell surface can dampen this response, DC10-induced Treg are critical for full realization of this outcome. This suggests that infectious tolerance is an essential element in regulatory DC control of the B cell response in allergic asthma.
Collapse
Affiliation(s)
- Yanna Ma
- Division of Respirology, Critical Care and Sleep Medicine, Department of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Wojciech Dawicki
- Division of Respirology, Critical Care and Sleep Medicine, Department of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Xiaobei Zhang
- Division of Respirology, Critical Care and Sleep Medicine, Department of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - John R. Gordon
- Division of Respirology, Critical Care and Sleep Medicine, Department of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- * E-mail:
| |
Collapse
|
23
|
Chaisri U, Tungtrongchitr A, Indrawattana N, Meechan P, Phurttikul W, Tasaniyananda N, Saelim N, Chaicumpa W, Sookrung N. Immunotherapeutic efficacy of liposome-encapsulated refined allergen vaccines against Dermatophagoides pteronyssinus allergy. PLoS One 2017; 12:e0188627. [PMID: 29182623 PMCID: PMC5705073 DOI: 10.1371/journal.pone.0188627] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 11/10/2017] [Indexed: 12/17/2022] Open
Abstract
Allergen specific immunotherapy (AIT) can modulate the allergic response causing a long-term symptom subsidence/abolishment which leads to reduced drug use and prevention of new sensitization. AIT of house dust mite allergy (HDM) using the mite crude extract (CE) as the therapeutic agent is not only less effective than the AIT for many other allergens, but also frequently causes adverse effects during the treatment course. In this study, mouse model of Dermatophagoides pteronyssinus (Dp) allergy was invented for testing therapeutic efficacies of intranasally administered liposome (L) encapsulated vaccines made of single Dp major allergens (L-Der p 1, L-Der p 2), combined allergens (L-Der p 1 and Der p 2), and crude Dp extract (L-CE). The allergen sparing intranasal route was chosen as it is known that the effective cells induced at the nasal-associated lymphoid tissue can exert their activities at the lower respiratory tissue due to the common mucosal traffic. Liposome was chosen as the vaccine delivery vehicle and adjuvant as the micelles could reduce toxicity of the entrapped cargo. The Dp-CE allergic mice received eight doses of individual vaccines/placebo on alternate days. All vaccine formulations caused reduction of the Th2 response of the Dp allergic mice. However, only the vaccines made of single refined allergens induced expressions of immunosuppressive cytokines (TGF-β, IL-35 and/or IL-10) which are the imperative signatures of successful AIT. The data emphasize the superior therapeutic efficacy of single refined major allergen vaccines than the crude allergenic extract vaccine.
Collapse
Affiliation(s)
- Urai Chaisri
- Department of Tropical Pathology, Faculty of Tropical Medicine, Bangkok, Thailand
| | - Anchalee Tungtrongchitr
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nitaya Indrawattana
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Bangkok, Thailand
| | - Panisara Meechan
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Watchara Phurttikul
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Natt Tasaniyananda
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nawannaporn Saelim
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wanpen Chaicumpa
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nitat Sookrung
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
24
|
Nolin JD, Lai Y, Ogden HL, Manicone AM, Murphy RC, An D, Frevert CW, Ghomashchi F, Naika GS, Gelb MH, Gauvreau GM, Piliponsky AM, Altemeier WA, Hallstrand TS. Secreted PLA2 group X orchestrates innate and adaptive immune responses to inhaled allergen. JCI Insight 2017; 2:94929. [PMID: 29093264 PMCID: PMC5752296 DOI: 10.1172/jci.insight.94929] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 09/26/2017] [Indexed: 01/15/2023] Open
Abstract
Phospholipase A2 (PLA2) enzymes regulate the formation of eicosanoids and lysophospholipids that contribute to allergic airway inflammation. Secreted PLA2 group X (sPLA2-X) was recently found to be increased in the airways of asthmatics and is highly expressed in airway epithelial cells and macrophages. In the current study, we show that allergen exposure increases sPLA2-X in humans and in mice, and that global deletion of Pla2g10 results in a marked reduction in airway hyperresponsiveness (AHR), eosinophil and T cell trafficking to the airways, airway occlusion, generation of type-2 cytokines by antigen-stimulated leukocytes, and antigen-specific immunoglobulins. Further, we found that Pla2g10-/- mice had reduced IL-33 levels in BALF, fewer type-2 innate lymphoid cells (ILC2s) in the lung, less IL-33-induced IL-13 expression in mast cells, and a marked reduction in both the number of newly recruited macrophages and the M2 polarization of these macrophages in the lung. These results indicate that sPLA2-X serves as a central regulator of both innate and adaptive immune response to proteolytic allergen.
Collapse
Affiliation(s)
- James D. Nolin
- Department of Medicine, Division of Pulmonary and Critical Care
| | - Ying Lai
- Department of Medicine, Division of Pulmonary and Critical Care
| | | | | | - Ryan C. Murphy
- Department of Medicine, Division of Pulmonary and Critical Care
| | - Dowon An
- Department of Medicine, Division of Pulmonary and Critical Care
| | - Charles W. Frevert
- Department of Medicine, Division of Pulmonary and Critical Care
- Department of Comparative Medicine
| | | | | | - Michael H. Gelb
- Department of Chemistry, and
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
| | - Gail M. Gauvreau
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Adrian M. Piliponsky
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington, USA
| | | | | |
Collapse
|
25
|
Humoral immune responses during asthma and influenza co-morbidity in mice. Immunobiology 2017; 222:1064-1073. [PMID: 28889999 DOI: 10.1016/j.imbio.2017.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 06/29/2017] [Accepted: 08/31/2017] [Indexed: 12/27/2022]
Abstract
Humoral immunity serve dual functions of direct pathogen neutralization and enhancement of leukocyte function. Antibody classes are determined by antigen triggers, and the resulting antibodies can contribute to disease pathogenesis and host defense. Although asthma and influenza are immunologically distinct diseases, since we have found that allergic asthma exacerbation promotes antiviral host responses to influenza A virus, we hypothesized that humoral immunity may contribute to allergic host protection during influenza. C57BL/6J mice sensitized and challenged with Aspergillus fumigatus (or not) were infected with pandemic influenza A/CA/04/2009 virus. Negative control groups included naïve mice, and mice with only 'asthma' or influenza. Concentrations of antibodies were quantified by ELISA, and in situ localization of IgA- and IgE-positive cells in the lungs was determined by immunohistochemistry. The number and phenotype of B cells in spleens and mediastinal lymph nodes were determined by flow cytometry at predetermined timepoints after virus infection until viral clearance. Mucosal and systemic antibodies remained elevated in mice with asthma and influenza with prominent production of IgE and IgA compared to influenza-only controls. B cell expansion was prominent in the mediastinal lymph nodes of allergic mice during influenza where most cells produced IgG1 and IgA. Although allergy-skewed B cell responses dominated in mice with allergic airways inflammation during influenza virus infection, virus-specific antibodies were also induced. Future studies are required to identify the mechanisms involved with B cell activation and function in allergic hosts facing respiratory viral infections.
Collapse
|
26
|
Th2 related markers in milk allergic inflammatory mice model, versus OVA. J Genet Eng Biotechnol 2017; 15:453-461. [PMID: 30647686 PMCID: PMC6296581 DOI: 10.1016/j.jgeb.2017.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 05/03/2017] [Accepted: 07/02/2017] [Indexed: 01/01/2023]
Abstract
Experimental studies on allergic asthma are limited by the high cost of the administrated allergens. In this study we tested the allergic potency of low fat milk as a cheap substitute to the widely used standard allergen, ovalbumin (OVA). BALB/c female mice (4 weeks old) were sensitized intraperitoneally with low fat milk/or OVA followed by intranasal challenge with the two allergens on days 28 and 29. At day 31, serum, bronchoalveolar lavage fluid (BALF), and lungs were harvested. Mice of the low fat milk model showed infiltration of eosinophils, macrophages, lymphocytes, and neutrophils in BALF comparable to that of the OVA model. Both allergic protocols led to the production of similar numbers of Th2 cells and induced comparable expression of Th2 cytokine (IL-13) as evident by real time PCR for IL-13 and GATA3 (Th2 transcription factor) and confirmed by immunofluorescence for Th2 surface markers (T1/ST2). In addition, both mouse models had similar elevated levels of allergen specific antibody, IgG1 and IgE. Notably, HE, PAS, and LUNA stained lung sections from low fat milk treated mice had higher average pathological scores as compared to OVA treated mice. In conclusion, this study suggests that the low fat milk-induced inflammation showed hallmarks of allergic airway inflammatory model such as eosinophilic influx in BALF, increased numbers of Th2 cells, augmented expression of IL-13, elevated levels of circulatory IgG1 and IgE, signs of robust pulmonary inflammation, and most importantly it is a cheap and promising model for studying acute allergic airway inflammation and acute asthma.
Collapse
|
27
|
Guo TL, Meng AH. In Utero exposure to genistein enhanced intranasal house dust mite allergen-induced respiratory sensitization in young adult B6C3F1 mice. Toxicol Lett 2016; 253:17-26. [PMID: 27113705 DOI: 10.1016/j.toxlet.2016.04.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 03/30/2016] [Accepted: 04/20/2016] [Indexed: 02/07/2023]
Abstract
Despite many hypothesized benefits of dietary isoflavone genistein (GEN) deriving from soy-based products, questions surrounding GEN's developmental immunotoxic effects are increasing. To understand how in utero GEN exposure may modulate postnatal respiratory sensitization, we conducted a time course study using a common household allergen (house dust mites: HDM; 10μg/mouse) following intranasal instillation, a physiological route of allergen exposure. GEN was administered to dams by gavage from gestational day 14 to parturition at a physiologically relevant dose (20mg/kg bw). Female and male offspring were sensitized with HDM allergens beginning about one month prior to sacrifice followed by challenges with three weekly doses of HDM extracts, and they were euthanized at day 3 following the final HDM exposure at four different time points (postnatal day (PND) 80, 120, 160, and 200). In utero GEN combined with postnatal HDM exposures (GEN+HDM) increased total IgE production in both young female and male B6C3F1 offspring (e.g., PND 80 in females and PND 120 in males). Increased antigen-specific IgG1, IgG2a and IgG2b levels were also observed at various time points in both female and male offspring. In addition, increases in macrophage number in bronchoalveolar lavage fluid of both female and male GEN+HDM offspring at PND 80 and PND 120, respectively, were observed when compared to the vehicle group. For T cells, an increase over the vehicle in female GEN+HDM offspring was observed at PND 80. Due to similar patterns of increases, it seems likely that GEN+HDM-induced increases in total IgE and macrophages are related. Overall, in utero GEN plus later-life HDM exposures exert increases in total IgE and HDM-specific IgG production as well as macrophage recruitments to the lung in young adult mice.
Collapse
Affiliation(s)
- Tai L Guo
- Department of Veterinary Biosciences and Diagnostic Imaging, College of Veterinary Medicine, University of Georgia, Athens, GA 30602-7382, United States.
| | - Andrew H Meng
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298-0613, United States
| |
Collapse
|
28
|
Tasaniyananda N, Chaisri U, Tungtrongchitr A, Chaicumpa W, Sookrung N. Mouse Model of Cat Allergic Rhinitis and Intranasal Liposome-Adjuvanted Refined Fel d 1 Vaccine. PLoS One 2016; 11:e0150463. [PMID: 26954254 PMCID: PMC4783078 DOI: 10.1371/journal.pone.0150463] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/15/2016] [Indexed: 12/19/2022] Open
Abstract
Cats (Felis domesticus) are rich source of airborne allergens that prevailed in the environment and sensitized a number of people to allergy. In this study, a mouse model of allergic rhinitis caused by the cat allergens was developed for the first time and the model was used for testing therapeutic efficacy of a novel intranasal liposome-entrapped vaccines made of native Fel d 1 (major cat allergen) in comparison with the vaccine made of crude cat hair extract (cCE). BALB/c mice were sensitized with cCE mixed with alum intraperitoneally and intranasally. The allergic mice were treated with eight doses of either liposome (L)-entrapped native Fel d 1 (L-nFD1), L-cCE), or placebo on every alternate day. Vaccine efficacy evaluation was performed one day after provoking the treated mice with aerosolic cCE. All allergenized mice developed histological features of allergic rhinitis with rises of serum specific-IgE and Th2 cytokine gene expression. Serum IgE and intranasal mucus production of allergic mice reduced significantly after vaccination in comparison with the placebo mice. The vaccines also caused a shift of the Th2 response (reduction of Th2 cytokine expressions) towards the non-pathogenic responses: Th1 (down-regulation of the Th1 suppressive cytokine gene, IL-35) and Treg (up-regulation of IL-10 and TGF-β). In conclusions, a mouse model of allergic rhinitis to cat allergens was successfully developed. The intranasal, liposome-adjuvanted vaccines, especially the refined single allergen formulation, assuaged the allergic manifestations in the modeled mice. The prototype vaccine is worthwhile testing further for clinical use in the pet allergic patients.
Collapse
Affiliation(s)
- Natt Tasaniyananda
- Graduate Program in Immunology, Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Urai Chaisri
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Anchalee Tungtrongchitr
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Wanpen Chaicumpa
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Nitat Sookrung
- Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- * E-mail:
| |
Collapse
|
29
|
Coltherd JC, Rodgers DT, Lawrie RE, Al-Riyami L, Suckling CJ, Harnett W, Harnett MM. The parasitic worm-derived immunomodulator, ES-62 and its drug-like small molecule analogues exhibit therapeutic potential in a model of chronic asthma. Sci Rep 2016; 6:19224. [PMID: 26763929 PMCID: PMC4725896 DOI: 10.1038/srep19224] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/09/2015] [Indexed: 12/21/2022] Open
Abstract
Chronic asthma is associated with persistent lung inflammation and long-term remodelling of the airways that have proved refractory to conventional treatments such as steroids, despite their efficacy in controlling acute airway contraction and bronchial inflammation. As its recent dramatic increase in industrialised countries has not been mirrored in developing regions, it has been suggested that helminth infection may protect humans against developing asthma. Consistent with this, ES-62, an immunomodulator secreted by the parasitic worm Acanthocheilonema viteae, can prevent pathology associated with chronic asthma (cellular infiltration of the lungs, particularly neutrophils and mast cells, mucus hyper-production and airway thickening) in an experimental mouse model. Importantly, ES-62 can act even after airway remodelling has been established, arresting pathogenesis and ameliorating the inflammatory flares resulting from repeated exposure to allergen that are a debilitating feature of severe chronic asthma. Moreover, two chemical analogues of ES-62, 11a and 12b mimic its therapeutic actions in restoring levels of regulatory B cells and suppressing neutrophil and mast cell responses. These studies therefore provide a platform for developing ES-62-based drugs, with compounds 11a and 12b representing the first step in the development of a novel class of drugs to combat the hitherto intractable disorder of chronic asthma.
Collapse
Affiliation(s)
- J C Coltherd
- Institute of Infection, Immunity and Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, 120 University Place, G12 8TA, Glasgow, United Kingdom
| | - D T Rodgers
- Institute of Infection, Immunity and Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, 120 University Place, G12 8TA, Glasgow, United Kingdom
| | - R E Lawrie
- Institute of Infection, Immunity and Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, 120 University Place, G12 8TA, Glasgow, United Kingdom
| | - L Al-Riyami
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, G4 0RE, Glasgow, United Kingdom
| | - C J Suckling
- Department of Pure and Applied Chemistry, University of Strathclyde, G1 1XL, Glasgow, United Kingdom
| | - W Harnett
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, G4 0RE, Glasgow, United Kingdom
| | - M M Harnett
- Institute of Infection, Immunity and Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, 120 University Place, G12 8TA, Glasgow, United Kingdom
| |
Collapse
|
30
|
Tjota MY, Hrusch CL, Blaine KM, Williams JW, Barrett NA, Sperling AI. Signaling through FcRγ-associated receptors on dendritic cells drives IL-33-dependent TH2-type responses. J Allergy Clin Immunol 2014; 134:706-713.e8. [PMID: 25088053 PMCID: PMC4149927 DOI: 10.1016/j.jaci.2014.06.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/25/2014] [Accepted: 06/09/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND Although allergic sensitization can be generated against various allergens, it is unknown how such a diversity of antigens is able to promote TH2-mediated inflammation leading to atopy. Our previous studies demonstrated that allergen-specific IgG immune complexes (ICs) and house dust mite (HDM) extract both induced dendritic cells (DCs) to drive TH2-mediated inflammation, but the mechanism by which these diverse stimuli produce similar responses is unknown. OBJECTIVE We sought to identify the DC signaling pathways used by TH2 stimuli to promote TH2-mediated inflammation. METHODS C57BL/6, FcγRIII(-/-), FcRγ(-/-), and ST2(-/-) mice were sensitized and challenged with HDM, and inflammation was assessed based on results of flow cytometry and histology and cytokine production. Bone marrow-derived DCs from these strains were used in signaling and adoptive transfer experiments. RESULTS Our findings indicate that 2 distinct TH2 stimuli, ICs and HDM, use the FcRγ-associated receptors FcγRIII and Dectin-2, respectively, to promote TH2-mediated lung inflammation. In this study we demonstrate that both ICs and HDM induce expression of IL-33, a critical mediator in asthma pathogenesis and the differentiation of TH2 cells, in DCs. Upregulation of IL-33 in DCs is dependent on FcRγ, Toll-like receptor 4, and phosphoinositide 3-kinase. Exogenous IL-33 is sufficient to restore the development of TH2 responses in FcRγ-deficient mice. Finally, adoptive transfer of allergen-pulsed FcRγ(+/-) bone-marrow derived DCs restores the development of TH2-type inflammation in FcRγ-deficient mice, demonstrating the necessity of this signaling pathway in DCs for allergen-induced inflammation. CONCLUSION These data identify a mechanism whereby TH2 stimuli signal through FcRγ-associated receptors on DCs to upregulate IL-33 production and induce TH2-mediated allergic airway inflammation.
Collapse
Affiliation(s)
- Melissa Y Tjota
- Committee on Immunology, University of Chicago, Chicago, Ill; Medical Scientist Training Program, University of Chicago, Chicago, Ill
| | - Cara L Hrusch
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, Ill
| | - Kelly M Blaine
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, Ill
| | - Jesse W Williams
- Committee on Molecular Pathogenesis and Molecular Medicine, University of Chicago, Chicago, Ill
| | - Nora A Barrett
- Department of Medicine, Division of Rheumatology, Immunology, and Allergy, Harvard Medical School, Boston, Mass
| | - Anne I Sperling
- Committee on Immunology, University of Chicago, Chicago, Ill; Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, Ill; Committee on Molecular Pathogenesis and Molecular Medicine, University of Chicago, Chicago, Ill.
| |
Collapse
|
31
|
Abstract
Fc receptors play a central role in maintaining the homeostatic balance in the immune system. Our knowledge of the structure and function of these receptors and their naturally occurring polymorphisms, including single nucleotide polymorphisms and/or copy number variations, continues to expand. Through studies of their impact on human biology and clinical phenotype, the contributions of these variants to the pathogenesis, progression, and/or treatment outcome of many diseases that involve immunoglobulin have become evident. They affect susceptibility to bacterial and viral pathogens, constitute as risk factors for IgG or IgE mediated inflammatory diseases, and impact the development of many autoimmune conditions. In this chapter, we will provide an overview of these genetic variations in classical FcγRs, FcRLs, and other Fc receptors, as well as challenges in achieving an accurate and comprehensive understanding of the FcR polymorphisms and genomic architecture.
Collapse
Affiliation(s)
- Marc Daeron
- grid.428999.70000000123536535Institut Pasteur, Paris, France
| | - Falk Nimmerjahn
- grid.5330.50000000121073311Department of Biology, Institute of Genetics, University of Erlangen-Nürnberg, Erlangen, Germany
| | | |
Collapse
|
32
|
Molinari G, Colombo G, Celenza C. Respiratory allergies: a general overview of remedies, delivery systems, and the need to progress. ISRN ALLERGY 2014; 2014:326980. [PMID: 25006500 PMCID: PMC3972928 DOI: 10.1155/2014/326980] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 12/24/2013] [Indexed: 01/10/2023]
Abstract
The spread of respiratory allergies is increasing in parallel with the alarm of the scientific community. Evidently, our knowledge of the onset mechanisms of these diseases and, as a consequence, of the available remedies is inadequate. This review provides a brief, general description of current therapeutic resources and the state of research with regard to both drugs and medical devices in order to highlight their limits and the urgent need for progress. Increasing the amount of basic biochemical research will improve our knowledge of such onset mechanisms and the potential efficacy of therapeutic preparations.
Collapse
Affiliation(s)
- Giuliano Molinari
- Biochemistry Consulting Service, Giuliano Molinari, 20017 Rho, Milan, Italy
| | - Giselda Colombo
- Allergy and Immunology Unit, San Raffaele Hospital, 20132 Milan, Italy
| | - Cinzia Celenza
- Quality Assurance Service, Sandoz S.P.A., 21040 Origgio, Varese, Italy
| |
Collapse
|
33
|
Molecular basis for downregulation of C5a-mediated inflammation by IgG1 immune complexes in allergy and asthma. Curr Allergy Asthma Rep 2014; 13:596-606. [PMID: 24013944 DOI: 10.1007/s11882-013-0387-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Allergy and asthma are triggered primarily by the binding of allergen-specific immunoglobulin E (IgE)-allergen complexes to their receptors, recognition of the allergens by antigen-presenting cells, and allergen presentation to the T cells. These events lead to mucus secretions, runny nose, itchy eyes, sneezing, airway hyperresponsiveness, and nasal congestion. Complement 5a (C5a) has emerged as a central molecule that mediates these allergic reactions. Many allergens and allergen-specific IgG immune complexes (IgG-ICs) cause complement activation and C5a generation. C5a interaction with its receptor (C5aR) leads to the infiltration and activation of several immunologic cell types and the secretion of pathogenic inflammatory and proinflammatory mediators. However, IgG1-IC binding to the IgG inhibitory Fc gamma receptor (FcγRIIB) suppresses C5aR-mediated inflammatory signaling and, hence, may reduce the inflammatory immune responses through this FcγRIIB-mediated pathway. Reviews of the IgG1-IC interactions with C5a-mediated inflammatory immune responses suggest that IgG1-IC-C5a inhibitory therapy may reduce inflammation in allergic diseases.
Collapse
|
34
|
Tjota MY, Williams JW, Lu T, Clay BS, Byrd T, Hrusch CL, Decker DC, de Araujo CA, Bryce PJ, Sperling AI. IL-33-dependent induction of allergic lung inflammation by FcγRIII signaling. J Clin Invest 2013; 123:2287-97. [PMID: 23585480 DOI: 10.1172/jci63802] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 02/21/2013] [Indexed: 11/17/2022] Open
Abstract
Atopic asthma is a chronic inflammatory disease of the lungs generally marked by excessive Th2 inflammation. The role of allergen-specific IgG in asthma is still controversial; however, a receptor of IgG-immune complexes (IgG-ICs), FcγRIII, has been shown to promote Th2 responses through an unknown mechanism. Herein, we demonstrate that allergen-specific IgG-ICs, formed upon reexposure to allergen, promoted Th2 responses in two different models of IC-mediated inflammation that were independent of a preformed T cell memory response. Development of Th2-type airway inflammation was shown to be both FcγRIII and TLR4 dependent, and T cells were necessary and sufficient for this process to occur, even in the absence of type 2 innate lymphoid cells. We sought to identify downstream targets of FcγRIII signaling that could contribute to this process and demonstrated that bone marrow-derived DCs, alveolar macrophages, and respiratory DCs significantly upregulated IL-33 when activated through FcγRIII and TLR4. Importantly, IC-induced Th2 inflammation was dependent on the ST2/IL-33 pathway. Our results suggest that allergen-specific IgG can enhance secondary responses by ligating FcγRIII on antigen-presenting cells to augment development of Th2-mediated responses in the lungs via an IL-33-dependent mechanism.
Collapse
Affiliation(s)
- Melissa Y Tjota
- Committee on Immunology, University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|