Oral myeloid cells uptake allergoids coupled to mannan driving Th1/Treg responses upon sublingual delivery in mice

I Soria, J López-Relaño, M Viñuela, J-I Tudela, A Angelina, C Benito-Villalvilla, C M Díez-Rivero, B Cases, A I Manzano, E Fernández-Caldas, M Casanovas, O Palomares, J L Subiza, I Soria, J López-Relaño, M Viñuela, J-I Tudela, A Angelina, C Benito-Villalvilla, C M Díez-Rivero, B Cases, A I Manzano, E Fernández-Caldas, M Casanovas, O Palomares, J L Subiza

Abstract

Background: Polymerized allergoids coupled to nonoxidized mannan (PM-allergoids) may represent novel vaccines targeting dendritic cells (DCs). PM-allergoids are better captured by DCs than native allergens and favor Th1/Treg cell responses upon subcutaneous injection. Herein we have studied in mice the in vivo immunogenicity of PM-allergoids administered sublingually in comparison with native allergens.

Methods: Three immunization protocols (4-8 weeks long) were used in Balb/c mice. Serum antibody levels were tested by ELISA. Cell responses (proliferation, cytokines, and Tregs) were assayed by flow cytometry in spleen and lymph nodes (LNs). Allergen uptake was measured by flow cytometry in myeloid sublingual cells.

Results: A quick antibody response and higher IgG2a/IgE ratio were observed with PM-allergoids. Moreover, stronger specific proliferative responses were seen in both submandibular LNs and spleen cells assayed in vitro. This was accompanied by a higher IFNγ/IL-4 ratio with a quick IL-10 production by submandibular LN cells. An increase in CD4+ CD25high FOXP3+ Treg cells was detected in LNs and spleen of mice treated with PM-allergoids. These allergoids were better captured than native allergens by antigen-presenting (CD45+ MHC-II+ ) cells obtained from the sublingual mucosa, including DCs (CD11b+ ) and macrophages (CD64+ ). Importantly, all the differential effects induced by PM-allergoids were abolished when using oxidized instead of nonoxidized PM-allergoids.

Conclusion: Our results demonstrate for the first time that PM-allergoids administered through the sublingual route promote the generation of Th1 and FOXP3+ Treg cells in a greater extent than native allergens by mechanisms that might well involve their better uptake by oral antigen-presenting cells.

Keywords: allergoid; glycoconjugate; immunotherapy; mannan; sublingual.

© 2018 The Authors. Allergy Published by John Wiley & Sons Ltd.

Figures

Figure 1
Figure 1
Sublingual immunizations with Phleum pratense pollen allergens and assayed immunological parameters. (A) Schematic diagrams of the three different protocols used. The immunogens were as follows: N (native allergen); PM (PM‐allergoids); PM‐OX (PM‐allergoids further oxidized); PBS (phosphate‐buffered saline as a negative control). (B) After the last immunization, all animals were killed. Blood was collected for the measurement of specific antibodies in serum. Spleen and submandibular LN cells were isolated and cultured in vitro for assessing cytokines production and cell proliferation. In some experiments, FOXP3+ cells were determined in freshly isolated spleen and submandibular LNs
Figure 2
Figure 2
Serum antibody response in mice after sublingual immunization with Phleum pratense pollen allergens. The immunogens were as follows: N (native allergen); PM (PM‐allergoids); PM‐OX (PM‐allergoids further oxidized); PBS (phosphate‐buffered saline as a negative control). (A) Serum levels of IgG1, IgG2a, and IgE were measured by ELISA against native allergens (Phleum pratense) in mice immunized with different protocols. (B) IgG2a/IgE ratios of antibody levels from each individual mouse immunized with different protocols. Results are the mean ± SEM of 6 mice. Statistical differences were analyzed with Student's t test. *P < .05, **P < .01, ***P < .001
Figure 3
Figure 3
Proliferative T‐cell response in submandibular LNs and spleen from mice after sublingual immunization with Phleum pratense pollen allergens. The immunogens were as follows: N (native allergen); PM (PM‐allergoids); PM‐OX (PM‐allergoids further oxidized); PBS (phosphate‐buffered saline as a negative control). Proliferation was measured by flow cytometry (CFSE assay) in response to the native allergens (Phleum pratense) after 5‐day culture. The results obtained with submandibular LN (A) or spleen (B) cells, from mice immunized with different protocols, are the mean ± SEM of 6 mice per group. Statistical differences were analyzed with Student's t test. *P < .05, **P < .01, ***P < .001
Figure 4
Figure 4
Cytokine production in submandibular LNs and spleen from mice after sublingual immunization with Phleum pratense pollen allergens. The immunogens were as follows: N (native allergen); PM (PM‐allergoids); PM‐OX (PM‐allergoids further oxidized); PBS (phosphate‐buffered saline as a negative control). Cytokines were measured in the culture supernatant by flow cytometry (CBA) in response to the native allergens (Phleum pratense) after 48‐h culture. The results obtained with submandibular LN (A) or spleen (B) cells, from mice immunized with different protocols, are the mean ± SEM of 6 mice per group. N.d. (nondetermined). Statistical differences were analyzed with Student's t test. *< .05, **< .01, ***< .001
Figure 5
Figure 5
FOXP3+ Treg cell induction in submandibular LNs and spleen from mice after sublingual immunization with Phleum pratense pollen allergens. The immunogens were as follows: N (native allergen); PM (PM‐allergoids); PM‐OX (PM‐allergoids further oxidized). Percentage of CD4+ CD25high FOXP3+ Treg cells measured by flow cytometry in submandibular LNs (A) or spleen (B) from mice immunized with different protocols. A representative flow cytometry dot plot is also displayed for each assayed condition. Results are the mean ± SEM of 6 mice. Statistical differences were analyzed with Student's t test. *< .05
Figure 6
Figure 6
Allergen uptake by antigen‐presenting (CD45+ MHC‐II +) cells isolated from the mouse sublingual mucosa. (A) Phenotypic characterization of the different cell subsets by flow cytometry: CD45+ MHC‐II + cells were gated and analyzed for CD64, CD11b, and CD207 cell surface markers. (B) Uptake of fluorescence‐labeled Phleum pratense allergen preparations after 30 min of incubation with CD45+/MHC‐II + cells. N (native allergen); PM (PM‐allergoids); PM‐OX (PM‐allergoids further oxidized). Representative histograms for each assayed condition are also displayed. (C) Percentage of CD64+ and CD11b+ cells within the CD45+ MHC‐II + cells fraction that uptake PM‐allergoids or N allergens. A representative example out of two independent experiments is shown. Data are the mean ± SEM of 4 independent experiments in A and B. Statistical differences were analyzed with Student's t test. **P < .01, ***P < .001

References

    1. Roberts G, Pfaar O, Akdis CA, et al. EAACI Guidelines on Allergen Immunotherapy: Allergic Rhinoconjunctivitis. Allergy 2017;
    1. Passalacqua G, Canonica GW, Bagnasco D. Benefit of SLIT and SCIT for Allergic Rhinitis and Asthma. Curr Allergy Asthma Rep 2016;16:88.
    1. Pajno GB, Caminiti L, Crisafulli G, et al. Adherence to sublingual immunotherapy in preschool children. Pediatr Allergy Immunol 2012;23:688‐689.
    1. Di Bona D, Plaia A, Leto‐Barone MS, La Piana S, Di Lorenzo G. Efficacy of subcutaneous and sublingual immunotherapy with grass allergens for seasonal allergic rhinitis: a meta‐analysis‐based comparison. J Allergy Clin Immunol. 2012;130:1097‐1107.
    1. Scadding GW, Calderon MA, Shamji MH, et al. Effect of 2 Years of Treatment With Sublingual Grass Pollen Immunotherapy on Nasal Response to Allergen Challenge at 3 Years Among Patients With Moderate to Severe Seasonal Allergic Rhinitis: The GRASS Randomized Clinical Trial. JAMA 2017;317:615‐625.
    1. Allam JP, Novak N. Local immunological mechanisms of sublingual immunotherapy. Curr Opin Allergy Clin Immunol 2011;11:571‐578.
    1. Akdis M, Akdis CA. Mechanisms of allergen‐specific immunotherapy: multiple suppressor factors at work in immune tolerance to allergens. J Allergy Clin Immunol. 2014;133:621‐631.
    1. Bohle B, Kinaciyan T, Gerstmayr M, Radakovics A, Jahn‐Schmid B, Ebner C. Sublingual immunotherapy induces IL‐10‐producing T regulatory cells, allergen‐specific T‐cell tolerance, and immune deviation. J Allergy Clin Immunol. 2007;120:707‐713.
    1. Canonica GW, Cox L, Pawankar R, et al. Sublingual immunotherapy: World Allergy Organization position paper 2013 update. World Allergy Organ J. 2014;7:51.
    1. Palomares O, Akdis M, Martín‐Fontecha M, Akdis CA. Mechanisms of immune regulation in allergic diseases: the role of regulatory T and B cells. Immunol Rev 2017;278:219‐236.
    1. Allam JP, Novak N. Immunological mechanisms of sublingual immunotherapy. Curr Opin Allergy Clin Immunol 2014;14:564‐569.
    1. Larenas‐Linnemann D, Cox LS. Immunotherapy and Allergy Diagnostics Committee of the American Academy of Allergy, Asthma and Immunology. European allergen extract units and potency: review of available information. Ann Allergy Asthma Immunol 2008;100:137‐145.
    1. Sun JB, Cuburu N, Blomquist M, Li BL, Czerkinsky C, Holmgren J. Sublingual tolerance induction with antigen conjugated to cholera toxin B subunit induces Foxp3 + CD25 + CD4 + regulatory T cells and suppresses delayed‐type hypersensitivity reactions. Scand J Immunol 2006;64:251‐259.
    1. Razafindratsita A, Saint‐Lu N, Mascarell L, et al. Improvement of sublingual immunotherapy efficacy with a mucoadhesive allergen formulation. J Allergy Clin Immunol. 2007;120:278‐285.
    1. Salari F, Varasteh AR, Vahedi F, Hashemi M, Sankian M. Down‐regulation of Th2 immune responses by sublingual administration of poly (lactic‐co‐glycolic) acid (PLGA)‐encapsulated allergen in BALB/c mice. Int Immunopharmacol 2015;29:672‐678.
    1. Sirvent S, Soria I, Cirauqui C, et al. Novel vaccines targeting dendritic cells by coupling allergoids to nonoxidized mannan enhance allergen uptake and induce functional regulatory T cells through programmed death ligand 1. J Allergy Clin Immunol. 2016;138:558‐567.
    1. Soria I, Alvarez J, Manzano AI, et al. Mite allergoids coupled to nonoxidized mannan from Saccharomyces cerevisae efficiently target canine dendritic cells for novel allergy immunotherapy in veterinary medicine. Vet Immunol Immunopathol 2017;190:65‐72.
    1. Manzano AI, Javier Cañada F, Cases B, et al. Structural studies of novel glycoconjugates from polymerized allergens (allergoids) and mannans as allergy vaccines. Glycoconj J 2016;33:93‐101.
    1. Schülke S, Vieths S. Dendritic cell targeting with C‐type lectins for improvement of allergen immunotherapy. J Allergy Clin Immunol. 2016;138:568‐570.
    1. Tanaka Y, Nagashima H, Bando K, et al. Oral CD103(‐)CD11b(+) classical dendritic cells present sublingual antigen and induce Foxp3(+) regulatory T cells in draining lymph nodes. Mucosal Immunol 2017;10:79‐90.
    1. Mascarell L, Saint‐Lu N, Moussu H, et al. Oral macrophage‐like cells play a key role in tolerance induction following sublingual immunotherapy of asthmatic mice. Mucosal Immunol 2011;4:638‐647.
    1. Subiza JL, Cañada J, Soria I, et al. Immunogenic complex for vaccination and method for the production thereof. WO2014162036A1.
    1. Mistrello G, Brenna O, Roncarolo D, Zanoni D, Gentili M, Falagiani P. Monomeric chemically modified allergens: immunologic and physicochemical characterization. Allergy 1996;51:8‐15.
    1. Kweon MN. Sublingual mucosa: a new vaccination route for systemic and mucosal immunity. Cytokine 2011;54:1‐5.
    1. Novak N. Targeting dendritic cells in allergen immunotherapy. Immunol Allergy Clin North Am. 2006;26:307‐319.
    1. Nagai Y, Shiraishi D, Tanaka Y, et al. Transportation of sublingual antigens across sublingual ductal epithelial cells to the ductal antigen‐presenting cells in mice. Clin Exp Allergy 2015;45:677‐686.
    1. Mascarell L, Lombardi V, Louise A, et al. Oral dendritic cells mediate antigen‐specific tolerance by stimulating TH1 and regulatory CD4 + T cells. J Allergy Clin Immunol. 2008;122:603‐609.
    1. Benito‐Villalvilla C, Cirauqui C, Diez‐Rivero CM, Casanovas M, Subiza JL, Palomares O. MV140, a sublingual polyvalent bacterial preparation to treat recurrent urinary tract infections, licenses human dendritic cells for generating Th1, Th17, and IL‐10 responses via Syk and MyD88. Mucosal Immunol 2017;10:924‐935.
    1. Cirauqui C, Benito‐Villalvilla C, Sánchez‐Ramón S, et al. Human dendritic cells activated with MV130 induce Th1, Th17 and IL‐10 responses via RIPK2 and MyD88 signalling pathways. Eur J Immunol 2018;48:180‐193.
    1. Gautier EL, Shay T, Miller J, et al. Gene‐expression profiles and transcriptional regulatory pathways that underlie the identity and diversity of mouse tissue macrophages. Nat Immunol 2012;13:1118‐1128.
    1. Misharin AV, Morales‐Nebreda L, Mutlu GM, Budinger GR, Perlman H. Flow cytometric analysis of macrophages and dendritic cell subsets in the mouse lung. Am J Respir Cell Mol Biol 2013;49:503‐510.
    1. Takahara K, Omatsu Y, Yashima Y, et al. Identification and expression of mouse Langerin (CD207) in dendritic cells. Int Immunol 2002;14:433‐444.
    1. Stambach NS, Taylor ME. Characterization of carbohydrate recognition by langerin, a C‐type lectin of Langerhans cells. Glycobiology 2003;13:401‐410.
    1. Allam JP, Würtzen PA, Reinartz M, et al. Phl p 5 resorption in human oral mucosa leads to dose‐dependent and time‐dependent allergen binding by oral mucosal Langerhans cells, attenuates their maturation, and enhances their migratory and TGF‐beta1 and IL‐10‐producing properties. J Allergy Clin Immunol. 2010;126:638‐645.
    1. Hovav AH. Dendritic cells of the oral mucosa. Mucosal Immunol 2014;7:27‐37.
    1. Rodriguez AR, Yu JJ, Murthy AK, et al. Mast cell/IL‐4 control of Francisella tularensis replication and host cell death is associated with increased ATP production and phagosomal acidification. Mucosal Immunol 2011;4:217‐226.
    1. Royer PJ, Emara M, Yang C, et al. The mannose receptor mediates the uptake of diverse native allergens by dendritic cells and determines allergen‐induced T cell polarization through modulation of IDO activity. J Immunol. 2010;185:1522‐1531.
    1. Morel PA, Butterfield LH. Dendritic cell control of immune responses. Front Immunol 2015;6:42.
    1. Berings M, Karaaslan C, Altunbulakli C, et al. Advances and Highlights in Allergen Immunotherapy: on the way to sustained clinical and immunologic tolerance. J Allergy Clin Immunol. 2017;140:1250‐1267.
    1. Scadding GW, Shamji MH, Jacobson MR, et al. Sublingual grass pollen immunotherapy is associated with increases in sublingual Foxp3‐expressing cells and elevated allergen‐specific immunoglobulin G4, immunoglobulin A and serum inhibitory activity for immunoglobulin E‐facilitated allergen binding to B cells. Clin Exp Allergy 2010;40:598‐606.
    1. Aloulou M, Carr EJ, Gador M, et al. Follicular regulatory T cells can be specific for the immunizing antigen and derive from naive T cells. Nat Commun 2016;7:10579.
    1. Allam JP, Stojanovski G, Friedrichs N, et al. Distribution of Langerhans cells and mast cells within the human oral mucosa: new application sites of allergens in sublingual immunotherapy? Allergy 2008;63:720‐727.
    1. Das M, Galeotti C, Stephen‐Victor E, Karnam A, Kaveri SV, Bayry J. Human basophils may not undergo modulation by DC‐SIGN and mannose receptor‐targeting immunotherapies due to absence of receptors. J Allergy Clin Immunol. 2017;139:1403‐1404.

Source: PubMed

3
Suscribir