Research article

No impact of filaggrin deficiency on the efficacy of epicutaneous immunotherapy in a murine model

  • Received: 17 November 2016 Accepted: 08 March 2017 Published: 15 March 2017
  • Background: Epicutaneous immunotherapy (EPIT®), currently investigated in the treatment of food allergy, needs the integrity of the skin to warrant safety and efficacy. Mutations in the gene encoding the key epidermal protein filaggrin (FLG) are risk factors for peanut allergy and disrupt the skin intergrity. We investigated the association between FLG deficiency and peanut EPIT® efficacy in a murine model. Methods: FLG mutant mice deficient in filaggrin (FLG-/-) or wild-type (WT) mice were sensitized with peanut protein extract (peanut protein) and cholera toxin. Sensitized mice received a patch per week during 8 weeks for EPIT®, using Viaskin®, and were then submitted to sustained peanut oral exposure. We assessed blood humoral and cellular responses and evaluated eosinophil infiltration in the gut mucosa. The different steps of allergen capture and transportation following deposition on the skin was also analyzed in sensitized mice. Results: Sensitization of mice was confirmed by a significant increase of specific Th2 biaised immunological responses. In sensitized mice, EPIT® significantly reduced IgE levels, splenocytes secretion of Th2 cytokines and recruitment of eosinophils in esophagus, compared to sensitized mice without epicutaneous immunotherapy. The allergen applied onto the skin of FLG-/- mice did not undergo passive skin passage or systemic delivery. Instead, the allergen was captured by skin CD205high DCs, which migrated to afferent lymph nodes, as already described in WT mice. Conclusions: EPIT® was efficient and safe in FLG-/- mice, suggesting that in Humans EPIT® keeps efficacy and safety in the presence of loss of function of FLG.

    Citation: Lucie Mondoulet, Sophie Wavrin, Vincent Dioszeghy, Véronique Dhelft, Emilie Puteaux, Mélanie Ligouis, Camille Plaquet, Christophe Dupont, Pierre-Henri Benhamou. No impact of filaggrin deficiency on the efficacy of epicutaneous immunotherapy in a murine model[J]. AIMS Allergy and Immunology, 2017, 1(1): 1-14. doi: 10.3934/Allergy.2017.1.1

    Related Papers:

    [1] Masahiro Seike . Histamine suppresses T helper 17 responses mediated by transforming growth factor-β1 in murine chronic allergic contact dermatitis. AIMS Allergy and Immunology, 2018, 2(4): 180-189. doi: 10.3934/Allergy.2018.4.180
    [2] Fatma Akpinar, Ayla Balci, Gulcan Ozomay, Ayca Sozen, Esra Kotan, Gulendam Kocak, Feyzullah Cetinkaya . Baby-skin care habits from different socio-economic groups and its impact on the development of atopic dermatitis. AIMS Allergy and Immunology, 2018, 2(1): 1-9. doi: 10.3934/Allergy.2018.1.1
    [3] Ross M. Tanis, Piper A. Wedman-Robida, Alena P. Chumanevich, John W. Fuseler, Carole A. Oskeritzian . The mast cell/S1P axis is not linked to pre-lesional male skin remodeling in a mouse model of eczema. AIMS Allergy and Immunology, 2021, 5(3): 160-174. doi: 10.3934/Allergy.2021012
    [4] Mujibullah Sheikh, Pranita S. Jirvankar . Innovative immunotherapeutic strategies for gastric cancer: A comprehensive review. AIMS Allergy and Immunology, 2025, 9(1): 27-55. doi: 10.3934/Allergy.2025003
    [5] McKenna S. Vininski, Sunanda Rajput, Nicholas J. Hobbs, Joseph J. Dolence . Understanding sex differences in the allergic immune response to food. AIMS Allergy and Immunology, 2022, 6(3): 90-105. doi: 10.3934/Allergy.2022009
    [6] Mohamed El Yaqoubi, Mouad Lahyaoui, Yousra Seqqat, Taoufiq Saffaj, Bouchaib Ihssane, Nabil Saffaj, Rachid Mamouni, Fouad Ouazzani Chahdi, Fahad M Alshabrmi, Alaa Abdulaziz Alnahari, Saad M. Howladar, Ammar AL-Farga, Youssef Kandri Rodi . Pyridopyrazine derivatives as highly selective histamine H4 receptor antagonist for the treatment of atopic dermatitis: QSAR modeling and molecular docking studies. AIMS Allergy and Immunology, 2024, 8(4): 303-323. doi: 10.3934/Allergy.2024019
    [7] Mansur Aliyu, Ali Akbar Saboor-Yaraghi, Sadegh Khodavaisy, Behrouz Robat-Jazi, Muhammad Ibrahim Getso . Glucuronoxylomannan: the salient polysaccharide in cryptococcal immunity. AIMS Allergy and Immunology, 2022, 6(2): 71-89. doi: 10.3934/Allergy.2022008
    [8] Gianna Moscato, Gianni Pala . Occupational allergy to food-derived allergens. AIMS Allergy and Immunology, 2017, 1(1): 21-30. doi: 10.3934/Allergy.2017.1.21
    [9] Rabia F Khan, Shehar B Bhatti, Zaigham Abbas . Formulating preservatives to enhance stability of crude extracts of food allergens used for food allergy management and immunotherapy. AIMS Allergy and Immunology, 2023, 7(1): 45-59. doi: 10.3934/Allergy.2023004
    [10] Seda Çevik, Uğur Altaş, Zeynep Meva Altaş, Mehmet Yaşar Özkars . Investigation of clinical characteristics of children with food allergy and factors associated with tolerance development. AIMS Allergy and Immunology, 2025, 9(2): 98-107. doi: 10.3934/Allergy.2025007
  • Background: Epicutaneous immunotherapy (EPIT®), currently investigated in the treatment of food allergy, needs the integrity of the skin to warrant safety and efficacy. Mutations in the gene encoding the key epidermal protein filaggrin (FLG) are risk factors for peanut allergy and disrupt the skin intergrity. We investigated the association between FLG deficiency and peanut EPIT® efficacy in a murine model. Methods: FLG mutant mice deficient in filaggrin (FLG-/-) or wild-type (WT) mice were sensitized with peanut protein extract (peanut protein) and cholera toxin. Sensitized mice received a patch per week during 8 weeks for EPIT®, using Viaskin®, and were then submitted to sustained peanut oral exposure. We assessed blood humoral and cellular responses and evaluated eosinophil infiltration in the gut mucosa. The different steps of allergen capture and transportation following deposition on the skin was also analyzed in sensitized mice. Results: Sensitization of mice was confirmed by a significant increase of specific Th2 biaised immunological responses. In sensitized mice, EPIT® significantly reduced IgE levels, splenocytes secretion of Th2 cytokines and recruitment of eosinophils in esophagus, compared to sensitized mice without epicutaneous immunotherapy. The allergen applied onto the skin of FLG-/- mice did not undergo passive skin passage or systemic delivery. Instead, the allergen was captured by skin CD205high DCs, which migrated to afferent lymph nodes, as already described in WT mice. Conclusions: EPIT® was efficient and safe in FLG-/- mice, suggesting that in Humans EPIT® keeps efficacy and safety in the presence of loss of function of FLG.


    Abbreviations:

    AD  atopic dermatitis

    DCs  dendritic cells

    EPIT   epicutaneous immunotherapy

    FLG-/-  filaggrin deficient

    Ig (E, G1, G2a)  immunoglobulin type E, G1, G2a

    RT-qPCR   reverse transcriptase-quantitatve-polymerase chain reaction

    WT    wild-type


    1. Introduction

    Mutations in the gene encoding filaggrin (FLG), an essential component of the epithelial barrier, are a significant predisposing factor for atopic dermatitis (AD) [1,2] and other allergies, including peanut allergy [3], as well as for the coexistence of atopic asthma or allergic rhinitis and AD [3,4,5]. Interestingly, the association between FLG deficiency and peanut allergy is not fully considered to be associated with coexistent AD. Filaggrin has been demonstrated to play a role in stratum corneum (SC) integrity as a structural protein that organizes keratin filaments. It is of note that lipid composition was aberrant in FLG-/- SC, whereas free fatty acid levels remained normal, and levels of ceramide and cholesterol were increased [6]. It is possible that aberrant lipid composition, as well as SC fragility, might have direct or indirect effects on the penetration of certain materials by affecting SC hydrophobicity. These data suggest that FLG deficiency constitutes a skin barrier defect that facilitates enhanced exposure of peanut allergen to antigen-presenting cells and subsequent sensitization, even in the absence of AD [1,2,4].

    Epicutaneous immunotherapy (EPIT®) uses a novel Viaskin® patch technology containing peanut proteins (DBV Technologies, Paris, France) [7] and should be performed on intactskin, in order to ensure the efficacy and safety of the treatment by avoiding a massive free passage of the allergen into the blood stream but also to induce a tolerogenic immune profile [7,9]. EPIT® has proven to be effective in peanut-sensitized mice [8,9,10,11,12] and is safe and well tolerated by subjects with peanut allergy [13]. The efficacy has been investigated in a large DBPC phase Ⅱ dose-finding trial (VIPES) in Europe and North America, and is currently being investigated in a pivotal Phase Ⅲ program for the treatment of peanut allergy in children [14].

    In view of the strong association of FLG mutations with peanut allergy (20% of cases) and the importance of skin integrity for EPIT® safety and tolerance induction, we soughta to investigate the association between FLG deficiency and peanut EPIT® efficacy.


    2. Materials and Methods


    2.1. Animals

    All experiments were performed in the animal facility of the Faculty of Pharmacy (Ch tenay-Malabry, France) according to the European Community rules on animal care and with permission 92-305 from the French Veterinary Services and with approval of French Ethic Committee number 26 (authorization 2012-091). For our experiments, we used a transgenic mice produced by Pr. Chambon [unpublished data]. His team generated FLG mutant mice in C57BL/6 background that were completely deficient in FLG (FLG-/-) and then we proceed to a backcross in a BALB/c background. The total loss of FLG expression was confirmed at mRNA level by reverse transcription quantitative polymerase chain reaction (RT-qPCR) (Figure 1a) and at protein level by immunohistochemistry in the skin (Figures 1b and 1c), upon methods published by Presland et al. [15] and Stout et al. [16], respectively. Atopic dermatitis (AD) was assessed by macroscopic observation in young FLG mutant pups and measurement of transepidermal water loss (TEWL) [Unpublished data]. In adult mice, TEWL is similar to Wild Type (WT) mice (respectively, mean 23.7 ± 0.6 vs 24.9 ± 0.6 g/m2/hr).

    Figure 1. Assessment of Filaggrin deficiency in the skin of mice. Reduced levels of profilaggrin mRNA in the dorsal skin of FLG-deficient mice. (a) RT-qPCR was performed on total RNA isolated from WT and FLG-deficient (FLG-/-) mouse skins using primer pairs specific for profilaggrin (forward: 5'GAATCCATATTTACAGCAAA GCACCTTG3' and reverse: 5'GGTATGTCCAATGTGATTGCACGATTG3'). SDHA and GAPDH represent housekeeping genes for the relative normalization. Immunohistology of dorsal skin of WT and FLG deficient mice. Skin tissues were stained with polyclonal FLG antibody (PRB-471P, Covance) followed by an anti-rabbit secondary antibody labelled with APC, then analyzed by microscopy (×20). (b) normal and (c) FLG-deficient dorsal skin from mice. Note the complete absence of FLG expression in the epidermis of FLG deficient mice.

    2.2. Treatment (Figure 2a)

    Figure 2. Study design. (a) Epicutaneous immunotherapy evaluated in Flg deficient mice; (b) Passive passage of major peanut allergen in bloodstream after peanut Viaskin® application; (c) Allergen (OVA*) delivery by Viaskin® patch, capture and migration of DCs.

    After oral sensitization by gavages with peanut protein and cholera toxin, WT or FLG-/- mice were epicutaneously treated using a Viaskin® loaded with 100 μg peanut protein (EPIT) or a placebo Viaskin® (Sham) followed by sustained oral exposure to peanuts for 10 consecutive days as already published [8]. The application of the Viaskin® was done on the back of mice, 24 h after the removal of hairs as previously described [8,9]. Blood was taken on days 42 (the end of sensitization) and 102 (the end of EPIT and substained peanut oral exposure) to determine peanut-specific IgE and IgG2a levels by in house indirect ELISA [10,11]. After termination by cervical dislocation, spleen and esophagus were collected on day 102 for cellular responses and cell infiltration analysis. Splenocytes were restimulated in vitro in presence of peanut protein (100 μg/mL) for 72 h and cytokine levels were measured using Bioplex® cytokine assay (Biorad, Marnes-la-Coquette, France) according to the manufacturer's instructions. Eosinophil infiltration was assessed by histology using Hematoxylin Erythrosine Safran (HES) staining and eosinophils were blindly counted by a skilled European College of Veterinary Certified Pathologist. Results were expressed as number of eosinophils per mm2. Negative control animals were naive WT or FLG-/- mice without sensitization and immunotherapy protocol.


    2.3. Passive passage of allergen in the bloodstream (Figure 2b)

    In order to evaluate the free passage to the bloodstream following epicutaneous administration of peanut protein, six-week-old sensitized WT or FLG-/- mice received a single application of Viaskin® loaded with 500 μg of peanut protein for 48 h (Viaskin-500). Epicutaneous application on tape-stripped skin (ie. stratum corneum removed by using iterative application on the same area of the skin of 6 adhesive tapes) and subcutaneous injections (200 μl containing 500 μg of peanut protein) were done as positive controls of bloodstream passage. Blood was collected by retro-orbital bleeding at different time points (0, 2, 8, 24, 48 h) to quantify Ara h 1 in serum using a commercial ELISA kit (Indoor Biotechnologies) as previously described [12]. The limit of quantification was determined as 7.8 ng/mL. Results were expressed in ng/mL as means ± SD for each group (8 mice per group).


    2.4. Allergen delivery, capture and migration of DCs (Figure 2c)

    The allergen delivery through the skin was assessed by microscopy 2 h and 24 h after application of a Viaskin® loaded with 100 μg of Alexa 488-conjugated OVA (OVA*) on intact skin of WT and FLG-/- mice. Skin below the OVA*-Viaskin was harvested for fluorescence microscopy. The immune uptake of OVA and migration of DCs were measured after sensitization using OVA*-Viaskin® applied onto intact skin of WT or FLG-/- mice (n = 5 per group). Positive control of allergen passive passage through the skin was obtained by applying the OVA* onto tape-stripped skin of WT mice. Negative control animals received an empty Viaskin® (naive WT mice). Inguinal LNs were harvested after 2 h to evaluate the absence of passive passage across the skin into lymphatics and 24 h for cell isolation and flow cytometry analysis of Alexa 488-positive CD11c+ DCs to determine the cell-mediated passage into LNs.


    2.5. Phenotype of migrating DCs

    To characterize the phenotype of migrating DCs in LNs of WT or FLG-/- mice, the OVA*-Viaskin® was applied onto the back of sensitized mice (n = 5 per group) for 24 h. Control received OVA*-Viaskin® on stripped skin (stripped WT mice). Inguinal LNs were harvested and the expression of DC marker CD205high and CD205low was analyzed on Alexa 488-positive CD11c+ DCs.


    2.6. Ancillary methods

    Reverse-transcription quantitative PCR (RT-qPCR): Total RNA from skin was extracted using the RNeasy Mini Kit (Qiagen, France) following the manufacturer's instructions. cDNA was synthetized by reverse transcription (SuperScript Ⅱ RNase H reverse transcription reagents, Invitrogen, France) containing 500 ng of RNA from the experimental samples. Quantitative PCR analyses in real time were performed with CFX (Biorad, France) for quantification. The thermal cycling conditions were: 95 ℃ for 5 min and then 45 cycles at 95 ℃ (5 s), 55 ℃ (5 s) and 72 ℃ (10 s). This was followed by the standard denaturation curve. The murine primers sequence for Flg is 5' GAATCCATATATTTACAGCAAAGCACCTTG3' (forward) and 5'GGTATGTCCAATGTGATTGCACGACGATTG3' (reverse). Expression of results was normalized to housekeeping genes (GAPDH and SDHA).

    Fluorescence microscopy: Skin tissue was harvested in Shandon cryomatrix (Thermo, France) and frozen in liquid nitrogen immediately after removing the Viaskin®. Cryostat sections (7 μm) mounted with Vectashield DAPI (Vactor lab, UK) were analyzed using a DMR microscope (Leica, France). Immunostaining for Flg was done with polyclonal antibodies (PRB-417P, Covance, UK) followed by an Alexa Fluor 555 anti-rabbit secondary antibody (A10042, Life Technologies, USA).

    Flow cytometry: cell suspensions wereincubated with anti-mouse CD16/CD32 Fc Block (BD Biosciences) and then stained with CD11c, CD3, MHC Ⅱ and B220 (BD Biosciences) and CD205 (eBioscience, UK) and their corresponding isotype control were also used. Flow cytometry was performed on FACSCalibur and analyzed using CellQuest software.


    2.7. Statistical analysis

    All statistical analysis were performed using the GraphPad Prism Software 6 (San Diego, CA, USA). Results are expressed as mean ± standard deviation (SD). For histological analyses, cytokine responses and humoral responses, statistical analyses were determined by Kruskall Wallis and post-Dunn test when comparing all groups.


    3. Results


    3.1. Absence of allergen passive passage into bloodstream after EPIT application in FLG-deficient mice

    We first evaluated whether a passive passage of Ara h 1 to the bloodstream could appear following application of a Viaskin® patch loaded with peanut protein on sensitized mice. When Viaskin-500 was applied on intact skin of sensitized FLG-/- or WT mice, no Ara h 1 was detected in the serum from 0 to 48 h (with a limit of quantification of the ELISA at 7.8 ng/ml). In contrast, when Viaskin-500 was applied on stripped skin, Ara h 1 was detected in the serum 2 h (39.5 ± 21.2 ng/mL) and 8 h (10.8 ± 5.4 ng/mL) after the application, in FLG-/- as well as in WT mice. In the positive control group (ie. mice treated by subcutaneous injection of 500 μg of peanut protein), a high quantity of Ara h 1 was detected from 2 h to 48 h, with a peak at 8 h (147.5 ± 20.6 ng/mL).


    3.2. No modification of antigen capture and migration of DCs in LNs after EPIT application in filaggrin deficient mice

    When the OVA*-Viaskin® was applied onto intact skin of WT and FLG-/- for 2 h and 24 h to evaluate the delivery of allergens through intact skin after Viaskin application, fluorescence was mainly observed in epidermis (Figure 3a-d).

    Figure 3. Study of allergen uptake in the skin. a-d, Fluorescence microscopy after 2 h and 24 h application of OVA-A-488-Viaskin patch respectively on (a and c) normal and (b and d) FLG-deficient dorsal skin from mice (original magnification ×20; green: OVA; blue: DAPI and red: MHC-Ⅱ). e, Passive passage of allergens to LNs (left part of the graph, 2 h) and allergen uptake in the skin and migration of A488+ cells into LNs (left part of the graph, 24 h). OVA*-Viaskin® were applied onto intact skin of either WT or FLG-/- mice and the presence of Alexa 488-positive resident CD11c+ DCs in inguinal LNs was analyzed by flow cytometry 2 h later. Positive control was obtained by applying the OVA*-Viaskin® onto stripped skin of WT mice (n = 5). Negative control received a placeboViaskin® (naive WT mice, n = 5). Data represent mean ± SD and are representative of three independent experiments. $: stripped vs naive, p < 0.01; *WT or FLG vs stripped, p < 0.01; ≠ FLG vs WT, ns.Phenotype of migrating DCs (right part of the graph, 24 h). Cells, isolated from the inguinal LNs of WT or FLG-/- mice that received OVA*-Viaskin® for 24 h, were gated on Alexa 488-positive CD11c+ DCs, and expression of CD205 was analyzed. Control received OVA*-Viaskin® on stripped skin (stripped WT mice). Data represent mean ± SD and are representative of three independent experiments. * p < 0.01, ** p < 0.001.

    Two hours after application of OVA*-Viaskin® applied on intact skin of sensitized FLG-/- or WT mice, the proportion of Alexa 488-positive resident CD11c+ DCs in LNs was not different from the background fluorescence in negative control naive WT mice, indicating that no passive passage of OVA was observed in LNs and likely the immune uptake of OVA applied by the Viaskin® and migration of DCs would be the same. In contrast, the proportion of Alexa 488-positive resident CD11c+ DCs in LNs increased markedly when OVA*-Viaskin® was applied onto stripped skin (positive control, p < 0.01) (Figure 3e, Passive passage of allergens to LNs) in FLG-/- and WT mice.

    After 24 h application of OVA*-Viaskin® on intact skin, aiming to evaluate the antigen capture and processing prior to entering the LNs, fluorescence was detected in cells from all skin layers in both FLG-/- and WT mice. MHC-Ⅱ staining indicated that OVA was specifically captured by CD11c+ DCs (data not shown). The migration of these cells from the skin was confirmed by the appearance of Alexa 488-positive CD11c+ DCs in LNs of both FLG-/- and WT mice after 24 h. This was also seen when OVA* was applied onto stripped skin. In contrast, Alexa 488-positive CD11c+ DCs remained low in negative control naive WT mice (Figure 3e, Allergen uptake in the skin and migration of A488+ cells into LNs).


    3.3. No difference of phenotype of migrating DCs after EPIT application in filaggrin deficient mice

    Migrating DCs in sensitized WT mice consisted of two populations according to CD205 staining intensity, such as previously described: CD205low DCs, which expressed low levels of CD86 and CD83 and CD205high, CD86high, and CD83high migrating DCs [6]. We observed no difference of migrating DCs between FLG-/- and WT mice, i.e CD205high DCs (Figure 3, Phenotype of migrating DCs). In contrast, migrating DCs of stripped WT mice were CD205low.


    3.4. Efficacy of EPIT in FLG-deficient mice

    We then investigated the association between FLG deficiency and EPIT efficacy. On day 42, peanut protein sensitized FLG-/- mice, as well as peanut protein sensitized WT, a significant increase in specific IgE (Table 1) together with IL-5, IL-13 and IFN-γ secretion of in vitro restimulated splenocytes (data not shown), compared to naive FLG-/- or WT mice. Sensitization of FLG-/- mice was thus as efficient as that of WT mice using a standard procedure.

    Table 1. Humoral, cellular responses and cell infiltration after oral exposure to peanuts.
    Humoral responses Cellular responses Cell Infiltration
    Specific IgE (μg/mL) Specific IgG1 Specific IgG2a (μg/mL) IL-5 (ng/mL) IL-13 (ng/mL) IFN-g (ng/mL) Eosinophil infiltration in esophagus/mm2
    (μg/mL)
    D42 D102 D42 D102 D42 D102 D102 D102 D102 D102
    WT EPIT 0.29 ± 0.04 0.17 ± 0.01* 231 ± 66 351 ± 88 1.5 ± 0.06 4.3 ± 0.18** 137.8 ± 81.6* 204.8 ± 71.6* 40.3 ± 12.1 14.5 ± 2.8**
    Sham 0.25 ± 0.03 0.23 ± 0.02 225 ± 85 226 ± 49 1.1 ± 0.13 1.2 ± 0.23 408.5 ± 144.1 596.5 ± 167.6 30.5 ± 21.3 49.7 ± 8.4
    naive <LOD <LOD <LOD <LOD <LOD <LOD <LOD <LOD <LOD 11.3 ± 1.7
    FLG EPIT 0.28 ± 0.02 0.19 ± 0.03* 298 ± 22 315 ± 56* 0.95 ± 0.14 3.8 ± 1.1** 215.4 ± 44.2* 492.3 ± 83.8* 87.1 ± 29.3 16.9 ± 2.7**
    -/- Sham 0.27 ± 0.03 0.26 ± 0.08 323 ± 52 176 ± 63 1.1 ± 0.16 1.2 ± 0.51 718.8 ± 140.6 957.8 ± 208.6 68.9 ± 36.9 50.8 ± 14.5
    naive <LOD <LOD <LOD <LOD <LOD <LOD <LOD <LOD <LOD 11.2 ± 3.2
    LOD: IgE = 0, 024 μg/mL, IgG1 = à 0, 004 μg/mL et IgG2a = 0, 004 μg/mL.
    IgE and IgG2a responses to peanut protein. Blood was harvested at the end of sensitization (D42) and at the end of EPIT and sustained peanut oral exposure (D102) of naive, sham-treated and EPIT WT or FLG-/- mice. IgE and IgG2a antibodies reactive to peanut protein were measured by ELISA. Data are expressed as means ± SD for each group of 8 mice. * p < 0.05, naive vs sham at D42 for WT or FLG-/- mice.* p < 0.05, ** p < 0.01 EPIT vs sham at D102 for WT or FLG-/- mice.
    Cytokine production by peanut protein-restimulated splenocytes. Spleen was harvested at the end of EPIT of naive, sham-treated and EPIT WT or FLG-/- mice. Cytokines production in splenocytes in presence of peanut protein for 72 h were measured by the Bioplex cytokine assay. Data are represented as means ± SD for each group of 8 mice. * p < 0.05, EPIT vs Sham at D102 for WT or FLG-/- mice.
    Eosinophil infiltration. Esophagus was collected for histological analysis of eosinophil infiltration using HES staining at D102.Results are expressed as number of eosinophils per mm2 and data are represented as means ± SD for each group of 8 mice. ** p < 0.01.
     | Show Table
    DownLoad: CSV

    After 8 weeks of EPIT in FLG-/- and in WT mice, specific IgE significantly decreased (p < 0.05) while specific IgG2a significantly increased compared to sham and negative control groups (p < 0.01) as previously described [10]. Peanut stimulated splenocytes from naive FLG-/- or WT mice did not secrete cytokines. Peanut stimulated splenocytes from sham FLG-/- and WT mice reactivated in vitro with peanut for 72 h secreted high levels of IL-5 (p < 0.05), IL-13 (p < 0.05) and IFN-γ (p < 0.05) compared to naive mice. EPIT significantly decreased Th2 cytokines secretion, both in EPIT FLG-/- and WT mice, respectively, IL-5 (p < 0.05) and IL-13 (p < 0.05). No cytokine secretion was detected by restimulation in medium alone (data not shown).

    To investigate the potential protection of EPIT in FLG-/- mice against allergen exposure, all treated and non treated mice were subjected to peanut oral exposure which induced inflammation and eosinophil infiltration in the esophagus. The eosinophil infiltration in the esophagus of EPIT treated FLG-/- and WT mice was significantly lower than in sham treated FLG-/- and WT mice (p < 0.01) and reached similar values compared to naive mice.


    4. Discussion

    Several immunotherapeutic options are being investigated and EPIT® appears promising to treat peanut allergic patients in Phase Ⅱ clinical trials [14]. Filaggrin mutations have been implicated in the development of peanut allergy, with filaggrin-deficient peanut allergic subjects representing 20% of cases [3]. Since disruption of the skin barrier might interfere with EPIT® [12], we took advantage of preclinical models of peanut-sensitized mice to test whether FLG loss of function might alter the efficacy of EPIT® [8,9,10,11,12] and showed that the absence of FLG does not reduce the desensitizing capacity of EPIT® in this model.

    Filaggrin is a major structural protein in the stratum corneum layer of the epidermis and is produced as the precursor profilaggrin. Mutations in the FLG gene are the most significant known genetic risk factor for the development of AD [1,2]. More interestingly, Brown et al. [3] identified that FLG mutations confer a risk for peanut allergy, even in the absence of AD. These data suggest a barrier defect that facilitates enhanced exposure of peanut allergen to antigen-presenting cells and subsequent sensitization in FLG-deficient human subjects, even in the absence of AD.

    We first confirmed that oral sensitization with peanut protein and cholera toxin in the selected animal model, ie. FLG-null (FLG-/-) mice, was similar to WT mice using a standard procedures [10]: the significant increase of peanut protein-specific Th2 biased immunological responses is is in accordance with the observation that general immunity through intraperitoneal sensitization with OVA was comparable between ft/ma and control mice [18].

    This mouse model among other ones has been selected based on the proposed rationale. Flaky tail/matted (ft/ma) mice display a matted hair phenotype due to a loss-of-function mutation in Tmem79 [18] and have spontaneous dermatitis with increased IgE levels under specific pathogen-free conditions [15]. They also express truncated profilaggrin and significantly reduced mature FLG [18] suggesting that FLG deficiency might explain the propensity of ft/ma mice to spontaneously have dermatitis in analogy with the situation in human subjects affected with AD [19,20]. Therefore, they have been widely used over the years to investigate pathogenic mechanisms of AD in the context of FLG deficiency [21,22]. However, two recently published studies report that the Tmem79 mutation, rather than the deletion of FLG, was found to be associated with the spontaneous dermatitis phenotype in flaky tail mice [19,22]. Furthermore, these mice do not show complete loss of FLG [23]. In this context, we used genetically engineered FLG-null mice that were characterized by a total loss of FLG expression at mRNA and protein levels. These FLG-/- mice have altered stratum corneum barrier function but do not develop spontaneous dermatitis under specific pathogen-free conditions as already described [24]. Skin lesions are possible in young mutant pups (up to 3 weeks of age) and in older ones (after one year of age).

    In contrast, ft/ma mice are predisposed to develop an allergen-specific immune response after epicutaneous sensitization with the clinically relevant allergen OVA: the topical application of OVA to ft/ma mice resulted in cutaneous inflammation and enhanced cutaneous allergen priming with development of systemic Th2 immune responses [17,21]. In FLG-/- generated by Kawasaki et al. [23], antigens also penetrated the stratum corneum more efficiently, leading to enhanced responses in hapten-induced contact hypersensitivity and higher serum levels of anti-OVA IgG1 and IgE [23]. In the same way, application of allergen on disrupted skin of naive mice induced sensitization [24,25,26]. These data provide experimental evidence that antigen transfer through a defective epidermal barrier is a key mechanism underlying increased IgE sensitization and initiation of cutaneous inflammation. This has been illustrated in humans by Lack et al. [27], showing that the exposure of the skin to peanut proteins may facilitate the sensitization process in very young children, when the normal architecture of the skin is altered by local or generalized eczema.

    On the other hand, EPIT® needs the integrity of the superficial layers of the stratum corneum to ensure safety and to induce a tolerogenic immune profile. Indeed, application of a Viaskin® on intact skin, in contrast with stripped skin, enhances the skin hydration and results in allergen diffusion through the superficial layers of the epidermis by preserving its architecture [7,9]. We used the model of peanut specific EPIT® in sensitized WT mice, with well characterized decrease of specific IgE, increase of specific IgG2a, lower production of Th2 cytokines and decrease of local eosinophil recruitment using various allergens (peanut, pollen, ovalbumin, house dust mites) [8,9,10,11,12]. FLG-/- mice showed similar EPIT® efficacy, thus strongly suggesting that EPIT® can be used effectively to treat allergic patient with FLG deficiency.

    We next demonstrated that FLG-/- mice show no difference with WT mice for free passage of allergen (i.e absence of passive passage of Ara h1 to bloodstream) following EPIT® application of peanut protein by Viaskin®. This contrasts with skin stripping, which leads to passive and increased passage of allergen through the skin and the lymphatics [9] resulting in systemic spread in vivo, completely modifying the biodistribution of allergen. FLG deficiency, although altering the skin barrier, thus appears different from skin stripping, which alters the activation of Langerhans cells, triggers mechanical injuries, activates keratinocytes and upregulates thymic stromal lymphopoietin expression by keratinocytes and mRNA expression of inflammatory cytokines. All these processes, observed after skin stripping, are involved in the polarization of skin DCs to elicit a Th2 response. In FLG-/- mice, the allergen is captured by CD205high DCs in the superficial layers of the stratum corneum of the epidermis, which migrate through the dermis into afferent LNs, as it was previously shown in WT mice [8].These results suggest that FLG-/- mice are not different than WT mice in terms of (ⅰ) absence of passive passage of allergen, (ⅱ) antigen uptake by DCs and (ⅲ) migration of DCs to draining LNs.


    5. Conclusion

    This study demonstrates for the first time the efficacy of EPIT® in FLG-deficient mice and suggests that EPIT® may be a safe and effective treatment for allergic patients with a FLG deficiency.


    Acknowledgments

    Acknowledgements are addressed to Dr. Mei Li and Pr. Pierre Chambon, from the IGBMC Institute, Strasbourg, France, for providing the FLG-deficient mice and their support to design the present mouse experiment. The authors would like to thank the staff of the animal facility of Paris XI University, School of Pharmacy, for the animal care.


    Declarations


    Ethics approval

    All experiments on mice were performed according to the European Community rules on animal care and with permission 92-305 from the French Veterinary Services and with approval of french Ethic Committee number 26 (authorization 2012-091).


    Consent for publication

    All co-authors gave their consents for data publication..


    Availability of data and material

    The datasets supporting the conclusions of this article are included within the article. For more detailed information, please contact author for data requests.


    Competing interests and funding

    This study was supported by DBV Technologies, the developer and owner of Viaskin®. Lucie Mondoulet, Sophie Wavrin, Vincent Dioszeghy, Emilie Puteaux, Mélanie Ligouis, Véronique Dhelft and Camille Plaquet are DBV Technologies employees.

    Christophe Dupont and Pierre-Henri Benhamou received honoraria and/or compensation in regards to the study, as investigators, coordinators or experts, in relation with the time spent on the study.

    The funders had a role in study design, decision to publish, and preparation of the manuscript. This does not alter our adherence to AIMS policies on sharing data and materials.


    [1] Irvine AD, McLean WH, Leung DY (2011) Filaggrin mutations associated with skin and allergic diseases. N Engl J Med 365: 1315–1327. doi: 10.1056/NEJMra1011040
    [2] Palmer CN, Irvine AD, Terron-Kwiatkowski A, et al. (2006) Common loss-of-function variants of the epidermal barrier protein filaggrin are a major predisposing factor for atopic dermatitis. Nat Genet 38: 441–446. doi: 10.1038/ng1767
    [3] Brown SJ, Asai Y, Cordell HJ, et al. (2011) Loss-of-function variants in the filaggrin gene are a significant risk factor for peanut allergy. J Allergy Clin Immunol 127: 661–667. doi: 10.1016/j.jaci.2011.01.031
    [4] Marenholz I, Nickel R, Ruschendorf F, et al. (2006) Filaggrin loss-of-function mutations predispose to phenotypes involved in the atopic march. J Allergy Clin Immunol 118: 866–871. doi: 10.1016/j.jaci.2006.07.026
    [5] Weidinger S, Rodriguez E, Stahl C, et al. (2007) Filaggrin mutations strongly predispose to early-onset and extrinsic atopic dermatitis. J Invest Dermatol 127: 724–726. doi: 10.1038/sj.jid.5700630
    [6] Kawasaki H, Nagao K, Kubo A, et al. (2012) Altered stratum corneum barrier and enhanced percutaneous immune responses in filaggrin-null mice. J Allergy Clin Immunol 129: 1538–1546.
    [7] Dupont C, Kalach N, Soulaines P, et al (2010) Cow's milk epicutaneous immunotherapy in children: a pilot trial of safety, acceptability, and impact on allergic reactivity. J Allergy Clin Immunol 125: 1165–1167. doi: 10.1016/j.jaci.2010.02.029
    [8] Mondoulet L, Dioszeghy V, Larcher T, et al. (2012) Epicutaneous immunotherapy (EPIT) blocks the allergic esophago-gastro-enteropathy induced by sustained oral exposure to peanuts in sensitized mice. PLoS One 7: e31967. doi: 10.1371/journal.pone.0031967
    [9] Dioszeghy V, Mondoulet L, Dhelft V, et al. (2011) Epicutaneous immunotherapy results in rapid allergen uptake by dendritic cells through intact sSkin and downregulates the allergen-specific response in sensitized mice. J Immunol 186: 5629–5637.
    [10] Mondoulet L, Dioszeghy V, Ligouis M, et al. (2010) Epicutaneous immunotherapy on intact skin using a new delivery system in a murine model of allergy. Clin Exp Allergy 40: 659–667.
    [11] Mondoulet L, Dioszeghy V, Vanoirbeek J, et al. (2011) Epicutaneous immunotherapy using a new epicutaneous delivery system in mice sensitized to peanut. Int Arch Allergy Immunol 154: 299–309. doi: 10.1159/000321822
    [12] Mondoulet L, Dioszeghy V, Puteaux E, et al. (2012) Intact skin and not stripped skin is crucial for the safety and efficacy of peanut epicutaneous immunotherapy (EPIT) in mice. Clin Transl Allergy 2: 22–34.
    [13] Jones SM, Agbotounou WK, Fleischer DM, et al. (2016) Safety of epicutaneous immunotherapy for the treatment of peanut allergy: A phase 1 study using the Viaskin patch. J Allergy Clin Immunol 137: 1258–1261.
    [14] Sampson HA, Agbotounou W, Thebaut C, et al. (2015) Epicutaneous immunotherapy (EPIT) is effective and safe to treat peanut allergy: a multi-national double-blind placebo-controlled randomized phase IIb trial. J Allergy Clin Immunol 135: AB390.
    [15] Presland RB, Boggess D, Lewis SP, et al. (2000) Loss of normal profilaggrin and filaggrin in flaky tail (ft/ft) mice: an animal model for the filaggrin-deficient skin disease ichthyosis vulgaris. J Invest Dermatol 115: 1072–1081. doi: 10.1046/j.1523-1747.2000.00178.x
    [16] Stout TE, McFarland T, Mitchell JC, et al. (2014) Recombinant filaggrin is internalized and processed to correct filaggrin deficiency. J Invest Dermatol 134: 423–429.
    [17] Fallon PG, Sasaki T, Sandilands A, et al. (2009) A homozygous frameshift mutation in the mouse Flg gene facilitates enhanced percutaneous allergen priming. Nat Genet 41: 602–608. doi: 10.1038/ng.358
    [18] Sasaki T, Shiohama A, Kubo A, et al. (2013) A homozygous nonsense mutation in the gene for Tmem79, a component for the lamellar granule secretory system, produces spontaneous eczema in an experimental model of atopic dermatitis. J Allergy Clin Immunol 132: 1111–1120. doi: 10.1016/j.jaci.2013.08.027
    [19] McAleer MA, Irvine AD (2013) The multifunctional role of filaggrin in allergic skin disease. J Allergy Clin Immunol 131: 280–291. doi: 10.1016/j.jaci.2012.12.668
    [20] Moniaga CS, Kabashima K (2011) Filaggrin in atopic dermatitis: flaky tail mice as a novel model for developing drug targets in atopic dermatitis. Inflamm Allergy Drug Targets 10: 477–485. doi: 10.2174/187152811798104881
    [21] Moniaga CS, Egawa G, Kawasaki H (2010) Flaky tail mouse denotes human atopic dermatitis in the steady state and by topical application with Dermatophagoides pteronyssinus extract. Am J Pathol 176: 2385–2393. doi: 10.2353/ajpath.2010.090957
    [22] Saunders SP, Goh CS, Brown SJ, et al. (2013) Tmem79/Matt is the matted mouse gene and is a predisposing gene for atopic dermatitis in human subjects. J Allergy Clin Immunol 132: 1121–1129. doi: 10.1016/j.jaci.2013.08.046
    [23] Kawasaki H, Nagao K, Kubo A, et al. (2012) Altered stratum corneum barrier and enhanced percutaneous immune responses in filaggrin-null mice. J Allergy Clin Immunol 129: 1538–1546.
    [24] Oyoshi MK, Murphy GF, Geha RS (2009) Filaggrin-deficient mice exhibit TH17-dominated skin inflammation and permissiveness to epicutaneous sensitization with protein antigen. J Allergy Clin Immunol 124: 485–493.
    [25] Spergel JM, Mizoguchi E, Brewer JP, et al. (1998) Epicutaneous sensitization with protein antigen induces localized allergic dermatitis and hyperresponsiveness to methacholine after single exposure to aerosolized antigen in mice. J Clin Invest 101: 1614–1622. doi: 10.1172/JCI1647
    [26] Strid J, Callard R, Strobel S (2006) Epicutaneous immunization converts subsequent and established antigen-specific T helper type 1 (Th1) to Th2-type responses. Immunology 119: 27–35. doi: 10.1111/j.1365-2567.2006.02401.x
    [27] Lack G, Fox D, Northstone K, et al. (2003) Factors associated with the development of peanut allergy in childhood. N Engl J Med 348: 977–985. doi: 10.1056/NEJMoa013536
  • This article has been cited by:

    1. Hugh A. Sampson, The Riddle of Response to Cutaneous Allergen Exposure in Patients with Atopic Dermatitis, 2024, 10811206, 10.1016/j.anai.2024.05.005
    2. Jiayan Zhang, Guofang Li, Qiuyang Guo, Yijun Yang, Jinxiang Yang, Xiaobo Feng, Zhirong Yao, Allergens in Atopic Dermatitis, 2025, 68, 1559-0267, 10.1007/s12016-025-09024-7
  • Reader Comments
  • © 2017 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(6118) PDF downloads(1191) Cited by(2)

Article outline

Figures and Tables

Figures(3)  /  Tables(1)

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog