在α-gal综合征小鼠模型中,使用包裹半乳糖-α-1,3-半乳糖寡糖的可生物降解纳米颗粒进行免疫治疗,可增强对α-gal致敏的免疫耐受能力

IF 3.3 Q2 ALLERGY Frontiers in allergy Pub Date : 2024-08-09 DOI:10.3389/falgy.2024.1437523
Michael N. Saunders, Claudia M. Rival, Mahua Mandal, Kayla Cramton, Laila M Rad, K. Janczak, Laura A Williams, Amogh R. Angadi, J. O’Konek, L. Shea, Loren D. Erickson
{"title":"在α-gal综合征小鼠模型中,使用包裹半乳糖-α-1,3-半乳糖寡糖的可生物降解纳米颗粒进行免疫治疗,可增强对α-gal致敏的免疫耐受能力","authors":"Michael N. Saunders, Claudia M. Rival, Mahua Mandal, Kayla Cramton, Laila M Rad, K. Janczak, Laura A Williams, Amogh R. Angadi, J. O’Konek, L. Shea, Loren D. Erickson","doi":"10.3389/falgy.2024.1437523","DOIUrl":null,"url":null,"abstract":"IgE antibodies against the mammalian oligosaccharide allergen galactose-α-1,3-galactose (αGal) can result in a severe allergic disease known as alpha-gal syndrome (AGS). This syndrome, acquired by tick bites that cause αGal sensitization, leads to allergic reactions after ingestion of non-primate mammalian meat and mammalian-derived products that contain αGal. Allergen-specific immunotherapies for this tickborne allergic syndrome are understudied, as are the immune mechanisms of allergic desensitization that induce clinical tolerance to αGal. Here, we reveal that prophylactic administration of αGal glycoprotein-containing nanoparticles to mice prior to tick protein-induced αGal IgE sensitization blunts the production of Th2 cytokines IL-4, IL-5, and IL-13 in an αGal-dependent manner. Furthermore, these effects correlated with suppressed production of αGal-specific IgE and hypersensitivity reactions, as measured by reduced basophil activation and histamine release and the systemic release of mast cell protease-1 (MCPT-1). Therapeutic administration of two doses of αGal-containing nanoparticles to mice sensitized to αGal had partial efficacy by reducing the Th2 cytokine production, αGal-specific IgE production, and MCPT-1 release without reducing basophil activation or histamine release. These data identify nanoparticles carrying encapsulated αGal glycoprotein as a potential strategy for augmenting αGal-specific immune tolerance and reveal diverse mechanisms by which αGal nanoparticles modify immune responses for established αGal-specific IgE-mediated allergic reactions.","PeriodicalId":73062,"journal":{"name":"Frontiers in allergy","volume":null,"pages":null},"PeriodicalIF":3.3000,"publicationDate":"2024-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Immunotherapy with biodegradable nanoparticles encapsulating the oligosaccharide galactose-alpha-1,3-galactose enhance immune tolerance against alpha-gal sensitization in a murine model of alpha-gal syndrome\",\"authors\":\"Michael N. Saunders, Claudia M. Rival, Mahua Mandal, Kayla Cramton, Laila M Rad, K. Janczak, Laura A Williams, Amogh R. Angadi, J. O’Konek, L. Shea, Loren D. Erickson\",\"doi\":\"10.3389/falgy.2024.1437523\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"IgE antibodies against the mammalian oligosaccharide allergen galactose-α-1,3-galactose (αGal) can result in a severe allergic disease known as alpha-gal syndrome (AGS). This syndrome, acquired by tick bites that cause αGal sensitization, leads to allergic reactions after ingestion of non-primate mammalian meat and mammalian-derived products that contain αGal. Allergen-specific immunotherapies for this tickborne allergic syndrome are understudied, as are the immune mechanisms of allergic desensitization that induce clinical tolerance to αGal. Here, we reveal that prophylactic administration of αGal glycoprotein-containing nanoparticles to mice prior to tick protein-induced αGal IgE sensitization blunts the production of Th2 cytokines IL-4, IL-5, and IL-13 in an αGal-dependent manner. Furthermore, these effects correlated with suppressed production of αGal-specific IgE and hypersensitivity reactions, as measured by reduced basophil activation and histamine release and the systemic release of mast cell protease-1 (MCPT-1). Therapeutic administration of two doses of αGal-containing nanoparticles to mice sensitized to αGal had partial efficacy by reducing the Th2 cytokine production, αGal-specific IgE production, and MCPT-1 release without reducing basophil activation or histamine release. These data identify nanoparticles carrying encapsulated αGal glycoprotein as a potential strategy for augmenting αGal-specific immune tolerance and reveal diverse mechanisms by which αGal nanoparticles modify immune responses for established αGal-specific IgE-mediated allergic reactions.\",\"PeriodicalId\":73062,\"journal\":{\"name\":\"Frontiers in allergy\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":3.3000,\"publicationDate\":\"2024-08-09\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Frontiers in allergy\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.3389/falgy.2024.1437523\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q2\",\"JCRName\":\"ALLERGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Frontiers in allergy","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.3389/falgy.2024.1437523","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"ALLERGY","Score":null,"Total":0}
引用次数: 0

摘要

针对哺乳动物寡糖过敏原半乳糖-α-1,3-半乳糖(αGal)的 IgE 抗体可导致严重的过敏性疾病,即α-Gal 综合征(AGS)。针对这种蜱传过敏综合征的过敏原特异性免疫疗法以及诱导对αGal产生临床耐受的过敏脱敏免疫机制研究不足。在这里,我们揭示了在蜱蛋白诱导的αGal IgE致敏之前给小鼠服用含αGal糖蛋白的预防性纳米颗粒,能以αGal依赖的方式抑制Th2细胞因子IL-4、IL-5和IL-13的产生。此外,这些效应还与 αGal 特异性 IgE 和超敏反应的产生受到抑制有关,具体表现为嗜碱性粒细胞活化和组胺释放的减少以及肥大细胞蛋白酶-1 (MCPT-1) 的全身释放。给对αGal过敏的小鼠服用两种剂量的含αGal纳米颗粒,可减少Th2细胞因子的产生、αGal特异性IgE的产生和MCPT-1的释放,而不减少嗜碱性粒细胞的活化或组胺的释放,因此具有部分疗效。这些数据确定了携带封装的αGal糖蛋白的纳米颗粒是增强αGal特异性免疫耐受的一种潜在策略,并揭示了αGal纳米颗粒改变已建立的αGal特异性IgE介导的过敏反应的免疫反应的不同机制。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
Immunotherapy with biodegradable nanoparticles encapsulating the oligosaccharide galactose-alpha-1,3-galactose enhance immune tolerance against alpha-gal sensitization in a murine model of alpha-gal syndrome
IgE antibodies against the mammalian oligosaccharide allergen galactose-α-1,3-galactose (αGal) can result in a severe allergic disease known as alpha-gal syndrome (AGS). This syndrome, acquired by tick bites that cause αGal sensitization, leads to allergic reactions after ingestion of non-primate mammalian meat and mammalian-derived products that contain αGal. Allergen-specific immunotherapies for this tickborne allergic syndrome are understudied, as are the immune mechanisms of allergic desensitization that induce clinical tolerance to αGal. Here, we reveal that prophylactic administration of αGal glycoprotein-containing nanoparticles to mice prior to tick protein-induced αGal IgE sensitization blunts the production of Th2 cytokines IL-4, IL-5, and IL-13 in an αGal-dependent manner. Furthermore, these effects correlated with suppressed production of αGal-specific IgE and hypersensitivity reactions, as measured by reduced basophil activation and histamine release and the systemic release of mast cell protease-1 (MCPT-1). Therapeutic administration of two doses of αGal-containing nanoparticles to mice sensitized to αGal had partial efficacy by reducing the Th2 cytokine production, αGal-specific IgE production, and MCPT-1 release without reducing basophil activation or histamine release. These data identify nanoparticles carrying encapsulated αGal glycoprotein as a potential strategy for augmenting αGal-specific immune tolerance and reveal diverse mechanisms by which αGal nanoparticles modify immune responses for established αGal-specific IgE-mediated allergic reactions.
求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
2.80
自引率
0.00%
发文量
0
审稿时长
12 weeks
期刊最新文献
Early life exposures of childhood asthma and allergies-an epidemiologic perspective. Editorial: The safety, efficacy, and effectiveness of allergen-specific oral immunotherapy. A retrospective analysis of the clinical efficacy in patients treated with Alternaria alternata and Dermatophagoides farinae immunotherapy. The effect of the housing crisis in the Alabama Black Belt on respiratory health. Changes in epinephrine dispensings and allergy hospitalisations in Sweden in the years following the removal of autoinjector co-payments.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1