CD4+ 和 CD8+ T 细胞发出的不同脂质信号有助于 1 型糖尿病的发展

IF 5.7 2区 医学 Q1 IMMUNOLOGY Frontiers in Immunology Pub Date : 2024-09-18 DOI:10.3389/fimmu.2024.1444639
Tayleur D. White, Abdulaziz Almutairi, Ying Gai-Tusing, Daniel J. Stephenson, Benjamin D. Stephenson, Charles E. Chalfant, Xiaoyong Lei, Brian Lu, Bruce D. Hammock, Teresa P. DiLorenzo, Sasanka Ramanadham
{"title":"CD4+ 和 CD8+ T 细胞发出的不同脂质信号有助于 1 型糖尿病的发展","authors":"Tayleur D. White, Abdulaziz Almutairi, Ying Gai-Tusing, Daniel J. Stephenson, Benjamin D. Stephenson, Charles E. Chalfant, Xiaoyong Lei, Brian Lu, Bruce D. Hammock, Teresa P. DiLorenzo, Sasanka Ramanadham","doi":"10.3389/fimmu.2024.1444639","DOIUrl":null,"url":null,"abstract":"IntroductionWe reported that Ca<jats:sup>2+</jats:sup>-independent phospholipase A<jats:sub>2</jats:sub>β (iPLA<jats:sub>2</jats:sub>β)–derived lipids (iDLs) contribute to type 1 diabetes (T1D) onset. As CD4<jats:sup>+</jats:sup> and CD8<jats:sup>+</jats:sup> T cells are critical in promoting β-cell death, we tested the hypothesis that iDL signaling from these cells participates in T1D development.MethodsCD4<jats:sup>+</jats:sup> and CD8<jats:sup>+</jats:sup> T cells from wild-type non-obese diabetic (<jats:italic>NOD</jats:italic>) and <jats:italic>NOD</jats:italic>.<jats:italic>iPLA<jats:sub>2</jats:sub>β<jats:sup>+/-</jats:sup></jats:italic> (NOD<jats:italic>.HET</jats:italic>) mice were administered in different combinations to immunodeficient NOD.<jats:italic>scid</jats:italic>.ResultsIn mice receiving only <jats:italic>NOD</jats:italic> T cells, T1D onset was rapid (5 weeks), incidence 100% by 20 weeks, and islets absent. In contrast, onset was delayed 1 week and incidence reduced 40%–50% in mice receiving combinations that included NOD<jats:italic>.HET</jats:italic> T cells. Consistently, islets from these non-diabetic mice were devoid of infiltrate and contained insulin-positive β-cells. Reduced iPLA<jats:sub>2</jats:sub>β led to decreased production of proinflammatory lipids from CD4<jats:sup>+</jats:sup> T cells including prostaglandins and dihydroxyeicosatrienoic acids (DHETs), products of soluble epoxide hydrolase (sEH), and inhibition of their signaling decreased (by 82%) IFNγ<jats:sup>+</jats:sup>CD4<jats:sup>+</jats:sup> cells abundance. However, only DHETs production was reduced from CD8<jats:sup>+</jats:sup> T cells and was accompanied by decreases in <jats:italic>sEH</jats:italic> and <jats:italic>granzyme B</jats:italic>.DiscussionThese findings suggest that differential select iDL signaling in CD4<jats:sup>+</jats:sup> and CD8<jats:sup>+</jats:sup> T cells contributes to T1D development, and that therapeutics targeting such signaling might be considered to counter T1D.","PeriodicalId":12622,"journal":{"name":"Frontiers in Immunology","volume":null,"pages":null},"PeriodicalIF":5.7000,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Differential lipid signaling from CD4+ and CD8+ T cells contributes to type 1 diabetes development\",\"authors\":\"Tayleur D. White, Abdulaziz Almutairi, Ying Gai-Tusing, Daniel J. Stephenson, Benjamin D. Stephenson, Charles E. Chalfant, Xiaoyong Lei, Brian Lu, Bruce D. Hammock, Teresa P. DiLorenzo, Sasanka Ramanadham\",\"doi\":\"10.3389/fimmu.2024.1444639\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"IntroductionWe reported that Ca<jats:sup>2+</jats:sup>-independent phospholipase A<jats:sub>2</jats:sub>β (iPLA<jats:sub>2</jats:sub>β)–derived lipids (iDLs) contribute to type 1 diabetes (T1D) onset. As CD4<jats:sup>+</jats:sup> and CD8<jats:sup>+</jats:sup> T cells are critical in promoting β-cell death, we tested the hypothesis that iDL signaling from these cells participates in T1D development.MethodsCD4<jats:sup>+</jats:sup> and CD8<jats:sup>+</jats:sup> T cells from wild-type non-obese diabetic (<jats:italic>NOD</jats:italic>) and <jats:italic>NOD</jats:italic>.<jats:italic>iPLA<jats:sub>2</jats:sub>β<jats:sup>+/-</jats:sup></jats:italic> (NOD<jats:italic>.HET</jats:italic>) mice were administered in different combinations to immunodeficient NOD.<jats:italic>scid</jats:italic>.ResultsIn mice receiving only <jats:italic>NOD</jats:italic> T cells, T1D onset was rapid (5 weeks), incidence 100% by 20 weeks, and islets absent. In contrast, onset was delayed 1 week and incidence reduced 40%–50% in mice receiving combinations that included NOD<jats:italic>.HET</jats:italic> T cells. Consistently, islets from these non-diabetic mice were devoid of infiltrate and contained insulin-positive β-cells. Reduced iPLA<jats:sub>2</jats:sub>β led to decreased production of proinflammatory lipids from CD4<jats:sup>+</jats:sup> T cells including prostaglandins and dihydroxyeicosatrienoic acids (DHETs), products of soluble epoxide hydrolase (sEH), and inhibition of their signaling decreased (by 82%) IFNγ<jats:sup>+</jats:sup>CD4<jats:sup>+</jats:sup> cells abundance. However, only DHETs production was reduced from CD8<jats:sup>+</jats:sup> T cells and was accompanied by decreases in <jats:italic>sEH</jats:italic> and <jats:italic>granzyme B</jats:italic>.DiscussionThese findings suggest that differential select iDL signaling in CD4<jats:sup>+</jats:sup> and CD8<jats:sup>+</jats:sup> T cells contributes to T1D development, and that therapeutics targeting such signaling might be considered to counter T1D.\",\"PeriodicalId\":12622,\"journal\":{\"name\":\"Frontiers in Immunology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":5.7000,\"publicationDate\":\"2024-09-18\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Frontiers in Immunology\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.3389/fimmu.2024.1444639\",\"RegionNum\":2,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"IMMUNOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Frontiers in Immunology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.3389/fimmu.2024.1444639","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"IMMUNOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

引言我们曾报道过 Ca2+ 非依赖性磷脂酶 A2β (iPLA2β) 衍生的脂质(iDLs)会导致 1 型糖尿病(T1D)发病。由于 CD4+ 和 CD8+ T 细胞在促进 β 细胞死亡方面起着关键作用,我们测试了来自这些细胞的 iDL 信号参与 T1D 发病的假设。结果在只接受 NOD T 细胞的小鼠中,T1D 发病迅速(5 周),到 20 周时发病率为 100%,且无胰岛。相比之下,接受包含 NOD.HET T 细胞的组合的小鼠发病时间推迟了 1 周,发病率降低了 40%-50%。同样,这些非糖尿病小鼠的胰岛没有浸润,并含有胰岛素阳性的β细胞。iPLA2β的减少导致CD4+ T细胞产生的促炎脂质减少,包括前列腺素和二羟基二十碳三烯酸(DHETs),它们是可溶性环氧化物水解酶(sEH)的产物,抑制它们的信号传导可使IFNγ+CD4+细胞数量减少(82%)。这些研究结果表明,CD4+和CD8+ T细胞中不同的选择性iDL信号传导是T1D发病的原因之一,针对这种信号传导的疗法可用于治疗T1D。
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
Differential lipid signaling from CD4+ and CD8+ T cells contributes to type 1 diabetes development
IntroductionWe reported that Ca2+-independent phospholipase A2β (iPLA2β)–derived lipids (iDLs) contribute to type 1 diabetes (T1D) onset. As CD4+ and CD8+ T cells are critical in promoting β-cell death, we tested the hypothesis that iDL signaling from these cells participates in T1D development.MethodsCD4+ and CD8+ T cells from wild-type non-obese diabetic (NOD) and NOD.iPLA2β+/- (NOD.HET) mice were administered in different combinations to immunodeficient NOD.scid.ResultsIn mice receiving only NOD T cells, T1D onset was rapid (5 weeks), incidence 100% by 20 weeks, and islets absent. In contrast, onset was delayed 1 week and incidence reduced 40%–50% in mice receiving combinations that included NOD.HET T cells. Consistently, islets from these non-diabetic mice were devoid of infiltrate and contained insulin-positive β-cells. Reduced iPLA2β led to decreased production of proinflammatory lipids from CD4+ T cells including prostaglandins and dihydroxyeicosatrienoic acids (DHETs), products of soluble epoxide hydrolase (sEH), and inhibition of their signaling decreased (by 82%) IFNγ+CD4+ cells abundance. However, only DHETs production was reduced from CD8+ T cells and was accompanied by decreases in sEH and granzyme B.DiscussionThese findings suggest that differential select iDL signaling in CD4+ and CD8+ T cells contributes to T1D development, and that therapeutics targeting such signaling might be considered to counter T1D.
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
9.80
自引率
11.00%
发文量
7153
审稿时长
14 weeks
期刊介绍: Frontiers in Immunology is a leading journal in its field, publishing rigorously peer-reviewed research across basic, translational and clinical immunology. This multidisciplinary open-access journal is at the forefront of disseminating and communicating scientific knowledge and impactful discoveries to researchers, academics, clinicians and the public worldwide. Frontiers in Immunology is the official Journal of the International Union of Immunological Societies (IUIS). Encompassing the entire field of Immunology, this journal welcomes papers that investigate basic mechanisms of immune system development and function, with a particular emphasis given to the description of the clinical and immunological phenotype of human immune disorders, and on the definition of their molecular basis.
期刊最新文献
Cell-bound complement activation products in antiphospholipid antibody-positive patients without other systemic autoimmune rheumatic diseases Identifying immune checkpoints on dysregulated T-cells as prognostic biomarkers for multiple myeloma patients with COVID-19 Identification of telomere-related lncRNAs and immunological analysis in ovarian cancer From single-cell to spatial transcriptomics: decoding the glioma stem cell niche and its clinical implications Case report: Artificial thymic organoids facilitate clinical decisions for a patient with a TP63 variant and severe persistent T cell lymphopenia
×
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