Journal Home > Volume 12 , Issue 3

Increasing incidences showed that food allergies have attracted more and more attention from researchers. BALB/c mice were sensitized with wheat gluten combined with aluminum hydroxide adjuvant via intraperitoneal injection, transdermal sensitization, and oral gavage sensitization route. Results showed that all the three sensitization methods could induce allergic symptoms; increase the serum antibody (total immunoglobulin E (IgE), specific IgE, IgG, IgA) and histamine content; promote the secretion of Th2 cytokines (interleukin (IL)-4, IL-5, IL-13) and inflammatory factors (IL-6, IL-17A, IL-10); and inhibit the production of Th1 cytokines (IFN-γ, IL-2). However, the allergic symptoms of mice sensitized by intraperitoneal injection were the most obvious among the three models. The level of serum antibodies in intraperitoneal injection group was significantly higher than control. Subsequently, 16S rRNA sequencing technology was used to analyze the intestinal flora of mice. The results showed that the abundance of Firmicutes in the wheat protein sensitized group was lower than that in the normal group, while the abundance of Bacteroidetes was higher, and Lactobacillus was the difference marker in normal group. Bacterial species diversity analysis showed that the species richness and diversity of intestinal flora in mice were decreased, the difference between mice induced by intraperitoneal injection and normal control group mice was the most significant. Taken together, these results show that among three sensitization methods used to build a mouse model with aluminum hydroxide as adjuvant, intraperitoneal injection is the comparably best way to build a mouse sensitization mode.


menu
Abstract
Full text
Outline
About this article

Assessment of immune responses and intestinal flora in BALB/c mice model of wheat food allergy via different sensitization methods

Show Author's information Chenglong LiuChen ChenXingyu YanShimin GuXin JiaWenhui FuXiao MengWentong Xue( )
College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China

Peer review under responsibility of KeAi Communications Co., Ltd.

Abstract

Increasing incidences showed that food allergies have attracted more and more attention from researchers. BALB/c mice were sensitized with wheat gluten combined with aluminum hydroxide adjuvant via intraperitoneal injection, transdermal sensitization, and oral gavage sensitization route. Results showed that all the three sensitization methods could induce allergic symptoms; increase the serum antibody (total immunoglobulin E (IgE), specific IgE, IgG, IgA) and histamine content; promote the secretion of Th2 cytokines (interleukin (IL)-4, IL-5, IL-13) and inflammatory factors (IL-6, IL-17A, IL-10); and inhibit the production of Th1 cytokines (IFN-γ, IL-2). However, the allergic symptoms of mice sensitized by intraperitoneal injection were the most obvious among the three models. The level of serum antibodies in intraperitoneal injection group was significantly higher than control. Subsequently, 16S rRNA sequencing technology was used to analyze the intestinal flora of mice. The results showed that the abundance of Firmicutes in the wheat protein sensitized group was lower than that in the normal group, while the abundance of Bacteroidetes was higher, and Lactobacillus was the difference marker in normal group. Bacterial species diversity analysis showed that the species richness and diversity of intestinal flora in mice were decreased, the difference between mice induced by intraperitoneal injection and normal control group mice was the most significant. Taken together, these results show that among three sensitization methods used to build a mouse model with aluminum hydroxide as adjuvant, intraperitoneal injection is the comparably best way to build a mouse sensitization mode.

Keywords: Intestinal flora, Allergy, Wheat gluten, BALB/c mice, Sensitization methods

References(42)

[1]

A. Aquino, C.A. Conte-Junior, A systematic review of food allergy: nanobiosensor and food allergen detection, Biosensors 10 (2020) 194. https://doi.org/10.3390/bios10120194.

[2]

G. Ricci, L. Andreozzi, F. Cipriani, et al., Wheat allergy in children: a comprehensive update, Medicina 55 (2019) 1-11. https://doi.org/10.3390/medicina55070400.

[3]

A. Cianferoni, Wheat allergy: diagnosis and management, J. Asthma Allergy 9 (2016) 13-25. https://doi.org/10.2147/JAA.S81550.

[4]
WHO/IUIS, Allergen nomenclature home page[DB/OL]. http://www.allergen.org/index.php.
[5]

P.R. Shewry, Wheat, J. Exp. Bot. 60 (2009) 1537-1553. https://doi.org/10.1093/jxb/erp058.

[6]

F.R. Greer, S.H. Sicherer, A.W. Burks, Effects of early nutritional interventions on the development of atopic disease in infants and children: the role of maternal dietary restriction, breastfeeding, timing of introduction of complementary foods, and hydrolyzed formula, Pediatrics 121 (2008) 183-191. https://doi.org/10.1542/peds.2007-3022.

[7]

B.I. Nwaru, M. Erkkola, S. Ahonen, et al., Age at the introduction of solid foods during the first year and allergic sensitization at age 5 years, Pediatrics 125 (2010) 50. https://doi.org/10.1542/peds.2009-0813.

[8]

M.D. Martinis, M.M. Sirufo, A. Viscido, et al., Food allergies and ageing, Int. J. Mol. Sci. 20 (2019) 1-11. https://doi.org/10.3390/ijms20225580.

[9]

A. Nowak-Wegrzyn, H. Szajewska, G. Lack, et al., Food allergy and the gut, Nat. Rev. Gastro. Hepat. 14 (2017): 241-257. https://doi.org/10.1038/nrgastro.2016.187.

[10]

O.I. Iweala, C.R. Nagler, The microbiome and food allergy, Annu. Rev. Immunol. 37 (2019) 377-403. https://doi.org/10.1146/annurev-immunol-042718-041621.

[11]

A.T. Stefka, T. Feehley, P. Tripathi, et al., Commensal bacteria protect against food allergen sensitization, PNAS 111 (2014) 13145-13150. https://doi.org/10.1073/pnas.1412008111.

[12]

M. Chieppa, M. Rescigno, A. Huang, et al., Dynamic imaging of dendritic cell extension into the small bowel lumen in response to epithelial cell TLR engagement, J. Exp. Med. 203 (2007) 2841-2852. https://doi.org/10.1084/jem.20061884.

[13]

R. Aitoro, L. Paparo, A. Amoroso, et al., Gut microbiota as a target for preventive and therapeutic intervention against food allergy, Nutrients 9 (2017) 672-684. https://doi.org/10.3390/nu9070672.

[14]

A.R. Ballegaard, C.B. Madsen, K.L. Bogh, et al., An animal model for wheat allergy skin sensitisation: a comparative study in naive versus tolerant brown norway rats, Int. Arch. Allergy Imm. 178 (2019) 106-118. https://doi.org/10.1159/000493802.

[15]

H. Akiyama, R. Teshima, J.I. Sakushima, et al., Examination of oral sensitization with ovalbumin in Brown Norway rats and three strains of mice, Immunol. Lett. 78 (2001) 1-5. https://doi.org/10.1016/S0165-2478(01)00229-2.

[16]

Y. Jin, S. Ebaugh, A. Martens, et al., A mouse model of anaphylaxis and atopic dermatitis to salt-soluble wheat protein extract, Int. Arch. Allergy Imm. 174 (2017) 7-16. https://doi.org/10.1159/000479386.

[17]

M. Vijaykrishnaraj, B.V. Mohan Kumar, P. Prabhasankar, et al., Antigen-specific gut inflammation and systemic immune responses induced by prolonging wheat gluten sensitization in BALB/c murine model, J. Proteome Res. 16 (2017) 3514-3528. https://doi.org/10.1021/acs.jproteome.7b00199.

[18]

L.L. Fu, M. Xie, C. Wang, et al., Allergenicity of shrimp tropomyosin from different sensitization approaches on BALB/c mice, Food Sci. 39 (2018) 166-175. http://www.corc.org.cn/handle/1471x/5564023.

[19]

A. Peri, D. Vojvodi, B. Vukomanovi-Durdevid, Influence of allergy on clinical, immunological and histological characteristics of nasal polyposis, B-ENT 8 (2012) 25-32. https://doi.org/10.1177/0194599811412927.

[20]

C. Quast, E. Pruesse, P. Yilmaz, et al., The SILVA ribosomal RNA gene database project: improved data processing and web-based tools, Nucleic Acids Res. 41 (2012) D590-D596. https://doi.org/10.1093/nar/gks1219.

[21]

H. Broeck, A. America, M. Smulders, et al., A modified extraction protocol enables detection and quantification of celiac disease-related gluten proteins from wheat, J. Chromatogr. B 877 (2009) 975-982. https://doi.org/10.1016/j.jchromb.2009.02.035.

[22]

L. Tordesillas, M.C. Berin, H.A. Sampson. Immunology of food allergy, Immunity 47 (2017) 32-50. https://doi.org/10.1016/j.immuni.2017.07.004.

[23]

P. Rupa, Y. Mine, Oral immunotherapy with immunodominant T-cell epitope peptides alleviates allergic reactions in a BALB/c mouse model of egg allergy, Allergy 67 (2015) 74-82. https://doi.org/10.1111/j.1398-9995.2011.02724.x.

[24]

C. Chen, L. Lu, N. Sun, et al., Development of a Balb/c mouse model for food allergy: comparison of allergy-related responses to peanut agglutinin, β-lactoglobulin and potato acid phosphatase, Toxicol. Res. 6 (2017) 251-261. https://doi.org/10.1039/c6tx00371k.

[25]

E. Schiavi, B. Barletta, C. Butteroni, et al., Oral therapeutic administration of a probiotic mixture suppresses established Th2 responses and systemic anaphylaxis in a murine model of food allergy, Allergy 66 (2010) 499-508. https://doi.org/10.1111/j.1398-9995.2010.02501.x.

[26]

Y. Suzuki, S. Liu, T. Ogasawara, et al., A novel MRGPRX2-targeting antagonistic DNA aptamer inhibits histamine release and prevents mast cell-mediated anaphylaxis, Eur. J. Pharmacol. 878 (2020) 173104-173113. https://doi.org/10.1016/j.ejphar.2020.173104.

[27]

R. He, M.K. Oyoshi, H. Jin, et al., Epicutaneous antigen exposure induces a Th17 response that drives airway inflammation after inhalation challenge, PNAS 104 (2007) 15817-15822. https://doi.org/10.1073/pnas.0706942104.

[28]

R.H. Wilson, G.S. Whitehead, H. Nakano, et al., Allergic sensitization through the airway primes Th17-dependent neutrophilia and airway hyperresponsiveness, Am J. Respir. Crit. Care. Med. 180 (2009) 720-730. https://doi.org/10.1164/rccm.200904-0573OC.

[29]

A.K. Verma, S. Kumar, A. Sharma, et al., Allergic manifestation by black gram (Vigna mungo) proteins in allergic patients, Balb/c mice and RBL-2H3 cells, Int. Immunopharmacol. 23 (2014) 92-103. https://doi.org/10.1016/j.intimp.2014.08.016.

[30]

L.S. Ceballos, M.S. Sampelayo, F.G. Extremera, et al., Antigenic stimulation with goat and cow milk by oral and parenteral route in guinea pigs, Food Agr. Immunol. 21 (2010) 1-13. https://doi.org/10.1080/09540100903365860.

[31]

X. Hua, J.J. Goedert, A. Pu, et al., Allergy associations with the adult fecal microbiota: analysis of the American gut project, Ebiomedicine 3 (2016) 172-179. https://doi.org/10.1016/j.ebiom.2015.11.038.

[32]

R.B. Canani, N. Sangwan, A.T. Stefka, et al., Lactobacillus rhamnosus GG-supplemented formula expands butyrate-producing bacterial strains in food allergic infants, Isme Journal 10 (2015) 742-750. https://doi.org/10.1038/ismej.2015.151.

[33]

C. Shi, T. Pan, M. Cao, et al., Suppression of Th2 immune responses by the sulfated polysaccharide from Porphyra haitanensis in tropomyosin-sensitized mice, Int. Immunopharmacol. 24 (2015) 211-218. https://doi.org/10.1016/j.intimp.2014.11.019.

[34]

Q. Liu, Y. Zhang, Z. Shu, et al., Sulfated oligosaccharide of Gracilaria lemaneiformis protect against food allergic response in mice by up-regulating immunosuppression, Carbohyd. Polym. 230 (2019) 115567-115579. https://doi.org/10.1016/j.carbpol.2019.115567.

[35]

S. Bunyavanich, N. Shen, A. Grishin, et al., Early-life gut microbiome composition and milk allergy resolution, J. Allergy Clin. Immun. 138 (2016) 1122-1130. https://doi.org/10.1016/j.jaci.2016.03.041.

[36]

B. Rodriguez, G. Prioult, R. Bibiloni, et al., Germ-free status and altered caecal subdominant microbiota are associated with a high susceptibility to cow'smilk allergy in mice, FEMS Microbiol. Ecol. 76 (2011) 133-144. https://doi.org/10.1111/j.1574-6941.2010.01035.x.

[37]

R. Aitoro, R. Simeoli, A. Amoroso, et al., Extensively hydrolyzed casein formula alone or with L.rhamnosus GG reduces β-lactoglobulin sensitization in mice, Pediatr. Allergy Immu. 28 (2017) 230-237. https://doi.org/10.1111/pai.12687.

[38]

R. Sultana, A.J. Mcbain, C.A. O'Neill, Lysates of Lactobacillus and bifidobacterium augment tight junction barrier function in human primary epidermal keratinocytes in a strain-dependent manner, Appl. Environ. Microb. 79 (2013) 4887-4894. https://doi.org/10.1128/AEM.00982-13.

[39]

M.N. Rivas, O.T. Burton, P. Wise, et al., A microbiota signature associated with experimental food allergy promotes allergic sensitization and anaphylaxis, J. Allergy Clin. Immunol. 131 (2013) 201-221. https://doi.org/ 10.1016/j.jaci.2012.10.026.

[40]

Y.J. Huang, B.J. Marsland, S. Bunyavanich, et al., The microbiome in allergic disease: current understanding and future opportunities-2017 PRACTALL document of the American Academy of Allergy, Asthma & Immunology and the European Academy of Allergy and Clinical Immunology, J. Allergy Clin. Immunol. 139 (2017) 1099-1110. https://doi.org/10.1016/j.jaci.2017.02.007.

[41]

A.R. Panzer, S.V. Lynch, Influence and effect of the human microbiome in allergy and asthma, Curr. Opin. Rheumatol. 27 (2015) 373-380. https://doi.org/10.1097/BOR.0000000000000191.

[42]

R.B. Canani, R. Nocerino, G. Terrin, et al., Effect of Lactobacillus GG on tolerance acquisition in infants with cow's milk allergy: a randomized trial, J. Allergy Clin. Immunol. 129 (2012) 580-582. https://doi.org/10.1016/j.jaci.2011.10.004.

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 10 June 2021
Revised: 12 August 2021
Accepted: 21 December 2021
Published: 15 October 2022
Issue date: May 2023

Copyright

© 2023 Beijing Academy of Food Sciences. Publishing services by Elsevier B.V. on behalf of KeAi Communications Co., Ltd.

Acknowledgements

Acknowledgements

This review is supported by the National Key Research and Development Program of China (2019YFC1605000) and the National Natural Science Foundation (1872904).

Rights and permissions

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return