insulitis

脊柱炎
  • 文章类型: Journal Article
    背景:1型糖尿病(T1D)是一种自身免疫性疾病,其特征在于免疫系统对胰腺中产生胰岛素的β细胞的特异性破坏,包括协调攻击的CD4细胞和直接破坏β细胞的CD8细胞,导致葡萄糖稳态的丧失。
    方法:这份全面的文件深入研究了免疫系统和β细胞之间复杂的相互作用,旨在阐明在T1D中驱动其破坏的机制。对遗传倾向的见解,环境触发因素,自身免疫反应为理解β细胞的自身免疫攻击提供了基础。从病毒感染作为β细胞炎症反应的潜在触发因素的作用,一个复杂的谜题开始展开。这一探索强调了β细胞在打破免疫耐受中的重要性以及导致其靶向破坏的因素。此外,它研究了自噬的潜在作用以及细胞因子信号对β细胞功能和存活的影响.
    结论:这篇综述代表了目前关于T1D的研究结果,为保护β细胞群的新治疗方法提供了有价值的观点。恢复免疫耐受,并最终阻止或停止T1D的进展。通过解开免疫系统和β细胞之间复杂的动力学,我们更接近于对T1D发病机制的全面了解,为更有效的治疗和最终治愈铺平道路。
    BACKGROUND: Type 1 diabetes (T1D) is an autoimmune disease characterized by the specific destruction of insulin-producing beta cells in the pancreas by the immune system, including CD4 cells which orchestrate the attack and CD8 cells which directly destroy the beta cells, resulting in the loss of glucose homeostasis.
    METHODS: This comprehensive document delves into the complex interplay between the immune system and beta cells, aiming to shed light on the mechanisms driving their destruction in T1D. Insights into the genetic predisposition, environmental triggers, and autoimmune responses provide a foundation for understanding the autoimmune attack on beta cells. From the role of viral infections as potential triggers to the inflammatory response of beta cells, an intricate puzzle starts to unfold. This exploration highlights the importance of beta cells in breaking immune tolerance and the factors contributing to their targeted destruction. Furthermore, it examines the potential role of autophagy and the impact of cytokine signaling on beta cell function and survival.
    CONCLUSIONS: This review collectively represents current research findings on T1D which offers valuable perspectives on novel therapeutic approaches for preserving beta cell mass, restoring immune tolerance, and ultimately preventing or halting the progression of T1D. By unraveling the complex dynamics between the immune system and beta cells, we inch closer to a comprehensive understanding of T1D pathogenesis, paving the way for more effective treatments and ultimately a cure.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    研究胰岛炎症-胰岛炎的挑战之一是它是一种短暂的现象。传统的胰岛炎进展报告是基于累积,胰岛附近白细胞密度的供体平均值,阻碍疾病进展的胰岛内和胰岛间异质性。这里,我们的目的是了解为什么胰岛炎是不均匀的,通常在胰岛一侧形成胰岛周围炎病变。为了实现这一点,我们证明了网络理论在胰岛素炎期间分离胰岛内多细胞相互作用中的适用性。具体来说,我们问了这样一个问题:“胰岛中首先与免疫细胞相互作用的区域有什么独特之处?”这项研究利用非肥胖糖尿病小鼠模型的1型糖尿病,并检查了之间的相互作用α-,β-,T细胞,骨髓细胞,胰岛炎进展过程中胰岛中的巨噬细胞。基于个体胰岛中的T/β细胞比率跟踪疾病演变。在早期阶段,我们发现免疫细胞优先与胰岛的富含α细胞的区域相互作用。在胰岛外周,与没有α细胞邻居的细胞相比,发现α连接的β细胞被靶向得更多。此外,网络分析显示T-髓样增加,和T巨噬细胞与所有β细胞的相互作用。
    One of the challenges in studying islet inflammation-insulitis-is that it is a transient phenomenon. Traditional reporting of the insulitis progression is based on cumulative, donor-averaged values of leucocyte density in the vicinity of pancreatic islets, that hinder intra- and inter-islet heterogeneity of disease progression. Here, we aimed to understand why insulitis is non-uniform, often with peri-insulitis lesions formed on one side of an islet. To achieve this, we demonstrated the applicability of network theory in detangling intra-islet multi-cellular interactions during insulitis. Specifically, we asked the question \"What is unique about regions of the islet that interact with immune cells first\". This study utilized the non-obese diabetic mouse model of type one diabetes and examined the interplay among α-, β-, T-cells, myeloid cells, and macrophages in pancreatic islets during the progression of insulitis. Disease evolution was tracked based on the T/β cell ratio in individual islets. In the early stage, we found that immune cells are preferentially interacting with α-cell-rich regions of an islet. At the islet periphery α-linked β-cells were found to be targeted significantly more compared to those without α-cell neighbors. Additionally, network analysis revealed increased T-myeloid, and T-macrophage interactions with all β-cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    目标:血管炎,自身免疫性1型糖尿病之前的炎症标志,导致功能β细胞的最终丧失。然而,即使面对持续的胰岛炎,功能β细胞也可以持续存在。尽管免疫抑制治疗取得了进展,维持功能β细胞以防止胰岛炎进展和高血糖仍然是一个挑战。大麻素1型受体(CB1R),存在于免疫细胞和β细胞中,调节炎症和β细胞功能。这里,我们开创了一个反映人类胰岛炎的离体模型,以研究CB1R在这一过程中的作用。
    方法:从1型糖尿病发病的男性和女性个体和非糖尿病个体的外周血单核细胞(PBMC)中分离CD4+T淋巴细胞,提取RNA并通过实时PCR分析mRNA表达。从数据挖掘获得来自患有1型糖尿病的供体的单个β细胞表达。将来自男性和女性尸体供体的患者来源的人胰岛在溶解的细胞外基质凝胶中3D培养,与相同的供体PBMC共培养,并与细胞因子(IL-1β,TNF-α,IFN-γ)在存在媒介物或增加浓度的CB1R阻断剂JD-5037的情况下持续24-48小时。使用CRISPR/Cas9技术消除CNR1(编码CB1R)的表达。活力,通过活细胞探测和实时PCR测定细胞内应激和信号传导。在灌注系统中确定了以葡萄糖刺激的胰岛素分泌测量的胰岛功能。通过显微镜监测免疫细胞向胰岛中的浸润。将7周龄的非肥胖糖尿病小鼠用JD-5037治疗1周,然后安乐死。通过流式细胞术对胰岛中浸润的免疫细胞进行分析。
    结果:CNR1表达在1型糖尿病发病个体的循环CD4+T细胞中上调(比健康个体高6.9倍),在1型糖尿病供体的分选胰岛β细胞中上调(比健康个体高3.6倍)。外周限制性CB1R反向激动剂JD-5037可阻止人和小鼠胰岛炎的发生。机械上,CB1R阻断阻止了胰岛NO的产生,并改善了未折叠蛋白反应的ATF6臂。因此,人胰岛细胞/趋化因子表达降低,导致持续的胰岛细胞活力和功能。
    结论:这些结果表明,CB1R可能是1型糖尿病的一个有趣的靶标,同时突出了胰岛炎的调节机制。此外,这些发现可能适用于2型糖尿病,胰岛炎症也是一种病理生理因素。
    方法:分选的人β细胞的转录组学分析来自基因表达综合数据库,加入编号GSE121863,可在https://www上获得。ncbi.nlm.nih.gov/geo/query/acc。cgi?acc=GSM3448161。
    OBJECTIVE: Insulitis, a hallmark of inflammation preceding autoimmune type 1 diabetes, leads to the eventual loss of functional beta cells. However, functional beta cells can persist even in the face of continuous insulitis. Despite advances in immunosuppressive treatments, maintaining functional beta cells to prevent insulitis progression and hyperglycaemia remains a challenge. The cannabinoid type 1 receptor (CB1R), present in immune cells and beta cells, regulates inflammation and beta cell function. Here, we pioneer an ex vivo model mirroring human insulitis to investigate the role of CB1R in this process.
    METHODS: CD4+ T lymphocytes were isolated from peripheral blood mononuclear cells (PBMCs) from male and female individuals at the onset of type 1 diabetes and from non-diabetic individuals, RNA was extracted and mRNA expression was analysed by real-time PCR. Single beta cell expression from donors with type 1 diabetes was obtained from data mining. Patient-derived human islets from male and female cadaveric donors were 3D-cultured in solubilised extracellular matrix gel in co-culture with the same donor PBMCs, and incubated with cytokines (IL-1β, TNF-α, IFN-γ) for 24-48 h in the presence of vehicle or increasing concentrations of the CB1R blocker JD-5037. Expression of CNR1 (encoding for CB1R) was ablated using CRISPR/Cas9 technology. Viability, intracellular stress and signalling were assayed by live-cell probing and real-time PCR. The islet function measured as glucose-stimulated insulin secretion was determined in a perifusion system. Infiltration of immune cells into the islets was monitored by microscopy. Non-obese diabetic mice aged 7 weeks were treated for 1 week with JD-5037, then euthanised. Profiling of immune cells infiltrated in the islets was performed by flow cytometry.
    RESULTS: CNR1 expression was upregulated in circulating CD4+ T cells from individuals at type 1 diabetes onset (6.9-fold higher vs healthy individuals) and in sorted islet beta cells from donors with type 1 diabetes (3.6-fold higher vs healthy counterparts). The peripherally restricted CB1R inverse agonist JD-5037 arrested the initiation of insulitis in humans and mice. Mechanistically, CB1R blockade prevented islet NO production and ameliorated the ATF6 arm of the unfolded protein response. Consequently, cyto/chemokine expression decreased in human islets, leading to sustained islet cell viability and function.
    CONCLUSIONS: These results suggest that CB1R could be an interesting target for type 1 diabetes while highlighting the regulatory mechanisms of insulitis. Moreover, these findings may apply to type 2 diabetes where islet inflammation is also a pathophysiological factor.
    METHODS: Transcriptomic analysis of sorted human beta cells are from Gene Expression Omnibus database, accession no. GSE121863, available at https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSM3448161 .
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    1型糖尿病(T1D)是一种自身免疫性疾病,其中胰岛β细胞受到免疫系统的攻击,导致胰岛素缺乏和高血糖。与T1D相关的最高非同义单核苷酸多态性(SNP)之一是干扰素诱导的含解旋酶C结构域的蛋白1(IFIH1),它编码抗病毒细胞溶质RNA传感器。该SNP导致丙氨酸在氨基酸946(IFIH1A946T)被替换为苏氨酸,并增加了几种自身免疫性疾病的风险。包括T1D。我们假设IFIH1A946T风险变体,(IFIH1R)将通过刺激导致免疫细胞改变的I型干扰素(IFNI)信号传导来促进T1D发病机理。为了测试这个,我们在非肥胖糖尿病(NOD)小鼠背景下开发了Ifih1R基因敲入小鼠,自发的T1D模型。我们的结果表明,与非风险Ifih1(Ifih1NR)小鼠相比,Ifih1R的糖尿病发病率和胰岛炎略有增加,并且Ifih1R雌性的糖尿病发作显着加速。与Ifih1NR相比,Ifih1R小鼠表现出显着增强的干扰素刺激基因(ISG)特征,表明IFNI信号传导增加。Ifih1R小鼠表现出浆细胞的频率增加以及CD8T细胞的频率和活化的组织依赖性变化。我们的结果表明,IFIH1R可能通过改变免疫细胞的频率和激活来促进T1D的发病。这些发现提高了我们对rs1990760变体和T1D之间联系的认识。Further,这些数据首次证明了Ifih1R在NOD小鼠中的作用,这对于T1D的治疗方法的开发将是重要的。
    Type 1 diabetes (T1D) is an autoimmune disease in which pancreatic islet β-cells are attacked by the immune system, resulting in insulin deficiency and hyperglycemia. One of the top non-synonymous single-nucleotide polymorphisms (SNP) associated with T1D is in the interferon-induced helicase C domain-containing protein 1 (IFIH1), which encodes an anti-viral cytosolic RNA sensor. This SNP results in an alanine to threonine substitution at amino acid 946 (IFIH1A946T) and confers an increased risk for several autoimmune diseases, including T1D. We hypothesized that the IFIH1A946T risk variant, (IFIH1R) would promote T1D pathogenesis by stimulating type I interferon (IFN I) signaling leading to immune cell alterations. To test this, we developed Ifih1R knock-in mice on the non-obese diabetic (NOD) mouse background, a spontaneous T1D model. Our results revealed a modest increase in diabetes incidence and insulitis in Ifih1R compared to non-risk Ifih1 (Ifih1NR) mice and a significant acceleration of diabetes onset in Ifih1R females. Ifih1R mice exhibited a significantly enhanced interferon stimulated gene (ISG) signature compared to Ifih1NR, indicative of increased IFN I signaling. Ifih1R mice exhibited an increased frequency of plasma cells as well as tissue-dependent changes in the frequency and activation of CD8+ T cells. Our results indicate that IFIH1R may contribute to T1D pathogenesis by altering the frequency and activation of immune cells. These findings advance our knowledge on the connection between the rs1990760 variant and T1D. Further, these data are the first to demonstrate effects of Ifih1R in NOD mice, which will be important to consider for the development of therapeutics for T1D.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    目的:1型糖尿病(T1D)的发生是由于自身反应性免疫细胞的胰岛浸润导致β细胞的破坏,并且β细胞功能障碍参与了这一过程。我们先前报道了大麻素1受体(CB1)在小鼠中的遗传缺失和药理拮抗作用可改善胰岛素的合成和分泌,上调葡萄糖传感机械,通过减少细胞凋亡有利于β细胞存活,并增强β细胞增殖。此外,β细胞特异性CB1缺失保护高脂高糖饮食小鼠抵抗胰岛炎症和β细胞功能障碍。因此,我们假设它将减轻导致T1D的沉淀事件中β细胞的功能障碍.
    方法:我们从非肥胖糖尿病(NOD;NODRIPCre+Cnr1fl/fl)小鼠的β细胞中基因删除了CB1。我们评估了雌性NODRIPCreCnr1fl/fl小鼠及其NODRIPCre-Cnr1fl/fl和NODRIPCreCnr1Wt/Wt幼仔在26周内的高血糖发作。我们还检查了胰岛形态,胰岛受免疫细胞浸润和β细胞功能和增殖。
    结果:NOD小鼠中CB1的β细胞特异性缺失通过保持β细胞功能和质量显着降低了高血糖的发生率。与野生型同窝动物相比,缺失还可以防止NODRIPCreCnr1fl/fl小鼠中的β细胞凋亡和侵袭性胰岛炎。NODRIPCre+Cnr1fl/fl胰岛维持正常形态,没有β细胞去分化或出现胰岛外β细胞的迹象,表明自身免疫保护是β细胞CB1遗传缺失固有的。胰腺淋巴结Treg细胞在NODRIPCreCnr1fl/flvsNODRIPCre-Cnr1fl/fl中明显更高。
    结论:这些数据共同证明了在T1D活跃期保护β细胞免受代谢应激如何改善破坏性胰岛炎,并为CB1作为T1D的潜在药理靶点提供了证据。
    OBJECTIVE: Type 1 diabetes (T1D) occurs because of islet infiltration by autoreactive immune cells leading to destruction of beta cells and it is becoming evident that beta cell dysfunction partakes in this process. We previously reported that genetic deletion and pharmacological antagonism of the cannabinoid 1 receptor (CB1) in mice improves insulin synthesis and secretion, upregulates glucose sensing machinery, favors beta cell survival by reducing apoptosis, and enhances beta cell proliferation. Moreover, beta cell specific deletion of CB1 protected mice fed a high fat high sugar diet against islet inflammation and beta cell dysfunction. Therefore, we hypothesized that it would mitigate the dysfunction of beta cells in the precipitating events leading to T1D.
    METHODS: We genetically deleted CB1 specifically from beta cells in non-obese diabetic (NOD; NOD RIP Cre+ Cnr1fl/fl) mice. We evaluated female NOD RIP Cre+ Cnr1fl/fl mice and their NOD RIP Cre-Cnr1fl/fl and NOD RIP Cre+ Cnr1Wt/Wt littermates for onset of hyperglycemia over 26 weeks. We also examined islet morphology, islet infiltration by immune cells and beta cell function and proliferation.
    RESULTS: Beta cell specific deletion of CB1 in NOD mice significantly reduced the incidence of hyperglycemia by preserving beta cell function and mass. Deletion also prevented beta cell apoptosis and aggressive insulitis in NOD RIP Cre+ Cnr1fl/fl mice compared to wild-type littermates. NOD RIP Cre+ Cnr1fl/fl islets maintained normal morphology with no evidence of beta cell dedifferentiation or appearance of extra islet beta cells, indicating that protection from autoimmunity is inherent to genetic deletion of beta cell CB1. Pancreatic lymph node Treg cells were significantly higher in NOD RIP Cre+ Cnr1fl/flvs NOD RIP Cre-Cnr1fl/fl.
    CONCLUSIONS: Collectively these data demonstrate how protection of beta cells from metabolic stress during the active phase of T1D can ameliorate destructive insulitis and provides evidence for CB1 as a potential pharmacologic target in T1D.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    1型糖尿病(T1DM)的发展涉及复杂的遗传相互作用,环境,和免疫因素。通过调节蛋白酶和受体的活性,金属蛋白酶组织抑制因子3(TIMP3)在限制促炎细胞因子的表达和功能中发挥作用,这与T1DM的进展有关。本研究旨在研究骨髓细胞中TIMP3过表达对T1DM发展的影响。
    多次低剂量链脲佐菌素(MLDS)治疗12周后,在CD68启动子下的骨髓细胞中特异性过度表达TIMP3的糖尿病小鼠(MacT3小鼠)显示出改善的胰岛素分泌,胰岛形态和血管化,抗氧化防御系统,以及线粒体生物合成和功能的调节因子。为了了解这种保护的起源,MLSD治疗后11天,在胰腺组织中评估胰岛炎的严重程度和炎症参数,显示胰岛炎和促炎细胞因子肿瘤坏死因子-α的水平显着降低,白细胞介素-1β,和干扰素-γ在MacT3小鼠中。
    结果表明,TIMP3参与维护胰岛结构和功能,至少在某种程度上,通过调节与胰岛炎相关的促炎细胞因子的产生,可能代表了T1DM的一种新的治疗策略。
    Type 1 diabetes mellitus (T1DM) development involves a complex interplay of genetic, environmental, and immunological factors. By modulating the activity of proteases and receptors, the protein tissue inhibitor of metalloproteinase 3 (TIMP3) plays a role in limiting the expression and function of pro-inflammatory cytokines, which have been implicated in the advancement of T1DM. This study was aimed at examining the effect of TIMP3 overexpression in myeloid cells on the development of T1DM.
    Twelve weeks after multiple low doses of streptozotocin (MLDS) treatment, diabetic mice overexpressing TIMP3 specifically in myeloid cells under the CD68 promoter (MacT3 mice) showed improved insulin secretion, islet morphology and vascularization, antioxidant defense system, and regulatory factors of mitochondrial biosynthesis and function. To get mechanistic insights into the origin of this protection, the severity of insulitis and inflammatory parameters were evaluated in pancreatic tissues 11 days after MLSD treatment, showing significantly reduced insulitis and levels of the pro-inflammatory cytokine tumor necrosis factor-α, interleukin -1β, and interferon -γ in MacT3 mice.
    The results indicate that TIMP3 is involved in maintaining islet architecture and functions, at least in part, through modulation of pro-inflammatory cytokine production associated with insulitis and may represent a novel therapeutic strategy for T1DM.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:花生四烯酸(AA)被认为与营养素代谢有关,炎症和免疫力,表明它可能在自身免疫性疾病中起作用。我们先前的研究表明,DPP-4抑制剂(DPP-4i)可能在1型糖尿病中调节AA-相对信号。
    目的:研究AA对非肥胖糖尿病(NOD)小鼠自身免疫性糖尿病及其与DPP-4i的交互作用的影响。
    方法:将NOD小鼠随机等分为3组:AA组,AA+DPP-4i组和对照组。糖尿病的发病率,血糖,监测胰岛炎和细胞因子谱.实验结束时,胰腺组织用H&E染色使用MescoScaleDiscovery多重测定试剂盒检查血清细胞因子谱。
    结果:尽管AA或AA加DPP-4i治疗对糖尿病发病率和体重没有影响,AA治疗可降低血糖,保留胰岛形态并减轻NOD小鼠胰岛炎症细胞浸润,伴随着IL-10,IL-1β的血清水平升高,IL-6、IL-5、KC/GRO和TNF-α水平降低血清IL-2水平。
    结论:我们观察到AA治疗通过降低高血糖来缓解NOD小鼠的自身免疫性糖尿病,减轻胰岛炎和改善细胞因子谱。DPP-4i可能通过串扰减轻AA的影响。我们提供了AA治疗缓解NOD小鼠1型糖尿病的证据,这可能为1型糖尿病提供新的治疗选择。
    BACKGROUND: Arachidonic acid (AA) is considered to link nutrient metabolism, to inflammation and immunity, suggesting it may have a role in autoimmune diseases. Our previous study suggests that DPP-4 inhibitors (DPP-4i) might regulate AA - relative signaling in type 1 diabetes.
    OBJECTIVE: To examine the effect of AA on autoimmune diabetes and its cross-talk with DPP-4i in The Non-Obese Diabetic (NOD) mice.
    METHODS: The NOD mice were divided randomly and equally into three groups: AA group, AA plus DPP-4i group and control group. The incidence of diabetes, blood glucose, insulitis and cytokine profiles were monitored. At the end of the experiment, pancreatic tissues were stained by H&E. Serum cytokine profiles were examined using a Mesco Scale Discovery multiplexed-assay kit.
    RESULTS: Even though AA or AA plus DPP-4i treatment has no effect on incidence of diabetes and weight, AA treatment reduces blood glucose, preserves islet morphology and alleviates inflammatory cell infiltration into pancreatic islets in NOD mice, accompanying with increased serum levels of IL-10, IL-1 β, IL-6, IL-5, KC/GRO and TNF-α and decreased serum levels of IL-2.
    CONCLUSIONS: We observed that AA treatment alleviates autoimmune diabetes in NOD mice by reducing hyperglycemia, alleviating insulitis and improving cytokine profiles. DPP-4i might alleviate the effect of AA by cross-talk. We provide evidence of AA treatment to alleviate type 1 diabetes in NOD mice, which may provide a novel therapeutic option for type 1 diabetes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Systematic Review
    目的:在西方国家,儿童1型糖尿病的发病率显著增加。因此,确定所涉及的环境决定因素最终可能导致疾病预防。这里,我们旨在系统回顾(PROSPEROID:CRD42022362522)目前的证据,即妊娠期母体饮食因素与小鼠和人类后代发生1型糖尿病和/或胰岛自身免疫(IA)的风险之间存在关联.
    方法:根据PRISMA指南,本系统综述搜索了PubMed和Scopus(n=343)不同的MeSH术语组合,比如1型糖尿病,饮食,胰岛自身免疫,产前,营养,面筋,麦醇溶蛋白,维生素,牛奶,和纤维。
    结果:我们发现,目前文献中研究最多的饮食因素是面筋,膳食高级糖基化终产物(DAGEs),维生素D,脂肪酸,和铁。有关产前暴露于无麸质环境的结果表明,对IA的发育具有一贯的保护作用。产前暴露于维生素D和某些脂肪酸似乎可以防止IA的发展,而子宫内铁和脂肪暴露与IA风险增加相关。
    结论:我们得出的结论是,对于大多数调查的因素,没有建立明确的关联,因为文献代表了异构的数据池,尽管胎儿暴露于某些母体饮食成分,比如面筋,显示与IA风险增加的一致性关联。我们建议,队列和临床环境中的人类前瞻性饮食干预研究对于更好地评估怀孕期间母体饮食的关键和保护性产前暴露至关重要。
    OBJECTIVE: The incidence of type 1 diabetes mellitus in children is considerably increasing in western countries. Thus, identification of the environmental determinants involved could ultimately lead to disease prevention. Here, we aimed to systematically review (PROSPERO ID: CRD42022362522) the current evidence of the association between maternal dietary factors during gestation and the risk of developing type 1 diabetes and/or islet autoimmunity (IA) in murine and human offspring.
    METHODS: In accordance with PRISMA guidelines, the present systematic review searched PubMed and Scopus (n = 343) for different combinations of MeSH terms, such as type 1 diabetes, diet, islet autoimmunity, prenatal, nutrient, gluten, gliadin, vitamin, milk, and fibers.
    RESULTS: We found that the most investigated dietary factors in the present literature were gluten, dietary advanced glycosylated end products (dAGEs), vitamin D, fatty acids, and iron. The results concerning prenatal exposure to a gluten-free environment showed a consistently protective effect on the development of IA. Prenatal exposures to vitamin D and certain fatty acids appeared to protect against the development of IA, whereas in utero iron and fat exposures correlated with increased risks of IA.
    CONCLUSIONS: We conclude that a definite association is not established for most factors investigated as the literature represents a heterogeneous pool of data, although fetal exposures to some maternal dietary components, such as gluten, show consistent associations with increased risks of IA. We suggest that human prospective dietary intervention studies in both cohort and clinical settings are crucial to better evaluate critical and protective prenatal exposures from the maternal diet during pregnancy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    CXCL10是IFNγ诱导的趋化因子,与1型糖尿病的发病机理有关。被胰岛吸引的T细胞产生IFNγ,但目前尚不清楚是什么吸引了胰岛中第一个产生IFNγ的T细胞。在健康的非糖尿病易感(C57BL/6)小鼠的胰岛中,施用病原体后的肠道菌群失调诱导CXCL10表达,并依赖于TLR4信号传导,在非肥胖糖尿病(NOD)小鼠中,肠道菌群失调也在胰腺淋巴结IGRP反应性胰岛特异性T细胞中诱导CXCR3趋化因子受体。低度内毒素血症的典型数量,细菌脂多糖在野生型和RAG1或IFNG受体缺陷但不是I型IFN受体缺陷的NOD小鼠的分离胰岛中诱导CXCL10的产生,分离脂多糖诱导的T细胞和IFNγ产生CXCL10。虽然主要依赖骨髓细胞,β细胞对脂多糖也表现出先天免疫信号通路的激活和Cxcl10表达,表明它们对菌群失调的独立敏感性。因此,响应于低水平的脂多糖的CXCL10诱导可能允许胰腺淋巴结中印迹的胰岛特异性T细胞独立于IFN-g进入健康的胰岛。因此,通过与生态失调相关的低度内毒素血症,将肠道生态失调与早期胰岛自身免疫联系起来。
    CXCL10 is an IFNγ-inducible chemokine implicated in the pathogenesis of type 1 diabetes. T-cells attracted to pancreatic islets produce IFNγ, but it is unclear what attracts the first IFNγ -producing T-cells in islets. Gut dysbiosis following administration of pathobionts induced CXCL10 expression in pancreatic islets of healthy non-diabetes-prone (C57BL/6) mice and depended on TLR4-signaling, and in non-obese diabetic (NOD) mice, gut dysbiosis induced also CXCR3 chemokine receptor in IGRP-reactive islet-specific T-cells in pancreatic lymph node. In amounts typical to low-grade endotoxemia, bacterial lipopolysaccharide induced CXCL10 production in isolated islets of wild type and RAG1 or IFNG-receptor-deficient but not type-I-IFN-receptor-deficient NOD mice, dissociating lipopolysaccharide-induced CXCL10 production from T-cells and IFNγ. Although mostly myeloid-cell dependent, also β-cells showed activation of innate immune signaling pathways and Cxcl10 expression in response to lipopolysaccharide indicating their independent sensitivity to dysbiosis. Thus, CXCL10 induction in response to low levels of lipopolysaccharide may allow islet-specific T-cells imprinted in pancreatic lymph node to enter in healthy islets independently of IFN-g, and thus link gut dysbiosis to early islet-autoimmunity via dysbiosis-associated low-grade endotoxemia.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    1型糖尿病(T1D)被广泛认为是由产生胰岛素的β细胞的自身免疫破坏引起的。几十年来,这个概念一直是试图破译疾病的发病机制和预防/逆转疾病的中心原则。最近,这个和许多其他疾病相关的概念受到越来越多的质疑,特别是考虑到从人类T1D胰腺分析中获得的知识。也许最关键的是研究结果表明,一组细胞成分-免疫,内分泌,和外分泌在起源-机械上聚结以促进T1D。这篇综述考虑了这些新兴的概念,从基础科学到临床研究,并确定了几个关键的剩余知识空白。
    Type 1 diabetes (T1D) is widely considered to result from the autoimmune destruction of insulin-producing β cells. This concept has been a central tenet for decades of attempts seeking to decipher the disorder\'s pathogenesis and prevent/reverse the disease. Recently, this and many other disease-related notions have come under increasing question, particularly given knowledge gained from analyses of human T1D pancreas. Perhaps most crucial are findings suggesting that a collective of cellular constituents-immune, endocrine, and exocrine in origin-mechanistically coalesce to facilitate T1D. This review considers these emerging concepts, from basic science to clinical research, and identifies several key remaining knowledge voids.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号