class='head no_bottom_margin' id='sec1title'>Int'/> On-going Mechanical Damage from Mastication Drives Homeostatic Th17 Cell Responses at the Oral Barrier
首页> 美国卫生研究院文献>Elsevier Sponsored Documents >On-going Mechanical Damage from Mastication Drives Homeostatic Th17 Cell Responses at the Oral Barrier
【2h】

On-going Mechanical Damage from Mastication Drives Homeostatic Th17 Cell Responses at the Oral Barrier

机译:咀嚼引起的持续机械损伤在口腔壁障处驱动稳态Th17细胞反应

代理获取
本网站仅为用户提供外文OA文献查询和代理获取服务,本网站没有原文。下单后我们将采用程序或人工为您竭诚获取高质量的原文,但由于OA文献来源多样且变更频繁,仍可能出现获取不到、文献不完整或与标题不符等情况,如果获取不到我们将提供退款服务。请知悉。

摘要

class="head no_bottom_margin" id="sec1title">IntroductionBarrier-resident immune populations integrate local cues to generate responses that preserve barrier integrity, maintain host-commensal interactions, and aid in fighting infection (, ). In recent years our understanding of barrier-tailoring of immune responses has dramatically expanded. This is particularly true in the gastrointestinal (GI) tract and skin, where tissue-specific and microbial-derived signals have been shown to shape the immune surveillance network and immune responsiveness (, , ). Yet, little is known regarding the development of tissue-specific immunity at the gingiva, an essential oral barrier that supports the dentition, harbors a complex commensal microbiome, and is a site where food antigens are first encountered prior to GI tract entry. Indeed, how effective immunity and regulation are balanced at this oral barrier is poorly understood. Expanding our understanding of the basic mechanisms controlling immunity at this barrier is important because the breakdown of controlled immune responses at the gingiva leads to periodontitis, a common inflammatory disease of humans. Additionally, periodontitis has been linked to the potentiation of a plethora of inflammatory conditions, such as cardiovascular disease and rheumatoid arthritis (). Therefore, understanding the mediators of health and disease at the gingiva may have broad-reaching implications for systemic inflammation.T helper 17 (Th17) cells are key mediators of barrier immunity, participating in immune surveillance and maintenance of barrier integrity (). Importantly, this T cell subset has been implicated in mediating protective immunity as well as pathogenic inflammation at the oral barrier. The development of Th17-cell-mediated responses at barriers such as the skin and GI tract is linked to tissue-specific factors, particularly colonization by site-specific commensals (, ). However, in the gingiva the factors controlling tissue-specific immunity remain ill defined, and as such it is not known how Th17 cells are induced in this environment. The critical role of Th17 cells in mediating protection at the oral barrier is evident in patients with genetic defects in Th17 cell differentiation and function; these patients present with severe and recurrent oral fungal infections (, ). However, exaggerated Th17 cell responses at the gingiva are detrimental and have been shown to promote inflammatory bone loss and tissue damage in periodontitis (, ). How Th17 cells are induced in the gingiva and subsequently become deregulated in periodontitis is poorly understood. Therefore, elucidating the factors involved in the induction and regulation of Th17 cells in this environment will shed light on the tissue-specific cues that regulate immunity in the gingiva.Here we delineated the mechanisms controlling accumulation of Th17 cells in the gingiva. Our data show that the gingival interleukin 17 (IL-17)-producing CD4+ T cell population increased with age. Exploring this increase in Th17 cells in older mice, we found that the mechanisms controlling CD4+ T cell effector function in the gingiva were different to those operating at other barrier sites. Our data demonstrate that gingival Th17 cells were not dependent on colonization by commensal bacteria, as the Th17 cell population was unchanged in germ-free mice. However, gingival Th17 cells were dependent upon IL-6-mediated signals. We identified that mechanical damage, which induces IL-6 and occurs physiologically in the oral cavity through mastication and abrasion, promoted accumulation of gingival Th17 cells. Thus, damage, as opposed to commensal colonization, helps define the immune tone of the gingiva, clearly demonstrating that unique rules shape gingival immune-homeostasis.
机译:<!-fig ft0-> <!-fig @ position =“ anchor” mode =文章f4-> <!-fig mode =“ anchred” f5-> <!-fig / graphic | fig / alternatives / graphic mode =“ anchored” m1-> class =“ head no_bottom_margin” id =“ sec1title”>简介居住在障碍地区的免疫种群整合了局部线索,以产生能保持障碍完整性,维持宿主的反应互动,并有助于抵抗感染(,)。近年来,我们对免疫应答的屏障定制的理解已大大扩展。在胃肠道和皮肤中尤其如此,那里的组织特异性信号和微生物衍生信号已显示出可以塑造免疫监视网络和免疫反应性(“”)。然而,关于牙龈组织特异性免疫的发展还知之甚少,牙龈是支持牙列的必不可少的口腔屏障,具有复杂的共生微生物组,是在胃肠道进入之前首先遇到食物抗原的场所。的确,人们对如何在这种口服屏障上平衡有效的免疫力和调节能力了解得很少。扩大我们对在此障碍处控制免疫的基本机制的理解很重要,因为在牙龈处受控免疫反应的破坏会导致牙周炎,这是人类常见的炎症性疾病。此外,牙周炎与多种炎症性疾病的增强有关,例如心血管疾病和类风湿性关节炎()。因此,了解牙龈健康和疾病的介质可能对全身炎症有广泛的影响。辅助性17(Th17)细胞是屏障免疫的关键介质,参与免疫监视和屏障完整性的维持()。重要的是,该T细胞亚群与介导保护性免疫以及口腔屏障的致病性炎症有关。 Th17细胞介导的在诸如皮肤和胃肠道等屏障的反应的发展与组织特异性因子有关,尤其是通过位点特异性共生作用(,)定植。然而,在牙龈中,控制组织特异性免疫的因素仍然不清楚,因此,尚不知道在这种环境中如何诱导Th17细胞。在Th17细胞分化和功能存在遗传缺陷的患者中,Th17细胞在介导口腔屏障保护中的关键作用显而易见。这些患者出现严重且反复出现的口腔真菌感染(,)。然而,在牙龈处过度的Th17细胞反应是有害的,并已显示出可促进牙周炎中炎症性骨丢失和组织损伤(,)。人们对如何在牙龈中诱导Th17细胞并随后使其失调的了解甚少。因此,阐明在这种环境下诱导和调节Th17细胞的相关因素将阐明调节牙龈免疫力的组织特异性线索。在此,我们描述了控制Th17细胞在牙龈中积累的机制。我们的数据显示,随着年龄增长,产生牙龈白介素17(IL-17)的CD4 + T细胞数量增加。探索老年小鼠Th17细胞的这种增加,我们发现控制牙龈中CD4 + T细胞效应子功能的机制与在其他屏障位点的控制机制不同。我们的数据表明,牙龈Th17细胞不依赖共生细菌的定殖,因为在无菌小鼠中Th17细胞的数量没有变化。但是,牙龈Th17细胞依赖于IL-6介导的信号。我们确定机械损伤,它诱导IL-6并通过咀嚼和磨蚀在口腔中发生生理作用,从而促进了牙龈Th17细胞的蓄积。因此,与共生定植相反,损伤有助于确定牙龈的免疫力,清楚地表明独特的规则塑造了牙龈的免疫稳态。

相似文献

  • 外文文献
  • 中文文献
  • 专利
代理获取

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号