Gut inflammation

肠道炎症
  • 文章类型: Journal Article
    慢性炎症在流行病学上与胃肠道疾病的发病机制有关。包括炎症性肠病(IBD)和结直肠癌(CRC)。然而,我们对控制肠道炎症的分子机制的理解仍然不足,阻碍IBD和CRC靶向治疗的发展。在这项研究中,我们发现C15ORF48/miR-147是肠道炎症的负调节因子,通过调节上皮细胞的代谢。C15ORF48/miR-147编码两种分子产物,C15ORF48蛋白和miR-147-3pmicroRNA,主要在肠上皮中表达。C15ORF48/miR-147消融导致小鼠肠道菌群失调并加剧化学诱导的结肠炎。C15ORF48和miR-147-3p共同通过沉默线粒体复合物IV(CIV)的亚基NDUFA4来抑制结肠细胞代谢和炎症。有趣的是,C15ORF48蛋白,NDUFA4的结构同系物,包含一个独特的C末端α-螺旋结构域,对于将NDUFA4从mCIV置换及其随后的降解至关重要。NDUFA4沉默阻碍NF-κB信号激活并因此减弱炎症反应。总的来说,我们的发现已经确立了C15ORF48/miR-147-NDUFA4分子轴作为肠道稳态不可或缺的调节因子,桥接线粒体代谢和炎症。
    Chronic inflammation is epidemiologically linked to the pathogenesis of gastrointestinal diseases, including inflammatory bowel disease (IBD) and colorectal cancer (CRC). However, our understanding of the molecular mechanisms controlling gut inflammation remains insufficient, hindering the development of targeted therapies for IBD and CRC. In this study, we uncovered C15ORF48/miR-147 as a negative regulator of gut inflammation, operating through the modulation of epithelial cell metabolism. C15ORF48/miR-147 encodes two molecular products, C15ORF48 protein and miR-147-3p microRNA, which are predominantly expressed in the intestinal epithelium. C15ORF48/miR-147 ablation leads to gut dysbiosis and exacerbates chemically induced colitis in mice. C15ORF48 and miR-147-3p work together to suppress colonocyte metabolism and inflammation by silencing NDUFA4, a subunit of mitochondrial complex IV (CIV). Interestingly, the C15ORF48 protein, a structural paralog of NDUFA4, contains a unique C-terminal α-helical domain crucial for displacing NDUFA4 from CIV and its subsequent degradation. NDUFA4 silencing hinders NF-κB signaling activation and consequently attenuates inflammatory responses. Collectively, our findings have established the C15ORF48/miR-147-NDUFA4 molecular axis as an indispensable regulator of gut homeostasis, bridging mitochondrial metabolism and inflammation.
    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
    暂无摘要。
    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
    肥大细胞(MC)是组织驻留的免疫细胞,分布在所有组织中,并策略性地位于血液和淋巴管和神经附近。由于广泛的受体表达,MC充当组织哨兵,能够检测细菌和寄生虫的存在,并对不同的环境刺激做出反应。MC起源于骨髓(BM)祖细胞,在微环境因素的影响下进入循环并在外周器官成熟,从而区分为异质组织特异性亚群。尽管MC激活传统上与IgE介导的过敏反应有关,最近出现了这些细胞在包括肿瘤进展在内的其他病理状况中的作用。然而,MC生物学的几个方面仍有待澄清。单细胞RNA测序平台的出现为了解MCs的起源和分化以及它们在不同组织中的表型和功能提供了机会。包括肠子.这篇综述概述了单细胞转录组学研究如何深入了解MC的发育以及肠道MC亚群在健康和疾病中的功能作用。
    Mast cells (MCs) are tissue-resident immune cells distributed in all tissues and strategically located close to blood and lymphatic vessels and nerves. Thanks to the expression of a wide array of receptors, MCs act as tissue sentinels, able to detect the presence of bacteria and parasites and to respond to different environmental stimuli. MCs originate from bone marrow (BM) progenitors that enter the circulation and mature in peripheral organs under the influence of microenvironment factors, thus differentiating into heterogeneous tissue-specific subsets. Even though MC activation has been traditionally linked to IgE-mediated allergic reactions, a role for these cells in other pathological conditions including tumor progression has recently emerged. However, several aspects of MC biology remain to be clarified. The advent of single-cell RNA sequencing platforms has provided the opportunity to understand MCs\' origin and differentiation as well as their phenotype and functions within different tissues, including the gut. This review recapitulates how single-cell transcriptomic studies provided insight into MC development as well as into the functional role of intestinal MC subsets in health and disease.
    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:据报道,肠道菌群失调和肠道通透性增加先于1型糖尿病相关的自身免疫。肠道炎症在自身免疫中的作用尚不清楚。
    目的:评估肠道炎症标志物是否与胰岛自身免疫风险相关,以及饮食是否与肠道炎症标志物有关。
    方法:从芬兰1型糖尿病预测和预防队列中获得了75例胰岛自身免疫性儿童和88例对照儿童的巢式病例对照样本。用3天的食物记录来评估饮食,从6个月和12个月大的粪便样本中分析钙卫蛋白和人β-防御素-2(HBD-2)。在匹配的病例对照设置中使用条件逻辑回归分析来评估自身免疫的风险。方差分析,独立样本t检验,和一般线性模型用于二次分析,以检验背景特征和饮食因素与炎症标志物的关联.
    结果:在未调整的分析中,钙卫蛋白与胰岛自身免疫风险无关,而HBD-2在中间(比值比[OR]3.23;95%置信区间[CI]:1.03,10.08)或最高三分位数(OR3.02;95%CI:1.05,8.69)与12个月大时最低的HBD-2显示出边缘关联(P趋势=0.063),胰岛自身免疫风险较高。在敏感性分析中排除牛奶过敏的儿童加强了HBD-2与胰岛自身免疫的关联,而调整饮食因素和母亲教育削弱了它。在12个月大的时候,较高的脂肪摄入量与较高的HBD-2(β=0.219;95%CI:0.110,0.328),和更高的膳食纤维摄入量(β=-0.294;95%CI:-0.510,-0.078),镁(β=-0.036;95%CI:-0.059,-0.014),和钾(β=-0.003;95%CI:-0.005,-0.001)与较低的HBD-2。
    结论:婴儿期HBD-2水平升高可能与胰岛自身免疫风险升高有关。饮食因素在肠道炎症状态中起作用。
    BACKGROUND: Gut dysbiosis and increased intestinal permeability have been reported to precede type 1 diabetes-related autoimmunity. The role of gut inflammation in autoimmunity is not understood.
    OBJECTIVE: This study aimed to assess whether gut inflammation markers are associated with risk of islet autoimmunity and whether diet is associated with gut inflammation markers.
    METHODS: A nested case-control sample of 75 case children with islet autoimmunity and 88 control children was acquired from the Finnish Type 1 Diabetes Prediction and Prevention cohort. Diet was assessed with 3-d food records, and calprotectin and human β-defensin-2 (HBD-2) were analyzed from stool samples at 6 and 12 mo of age. Conditional logistic regression analysis was used in a matched case-control setting to assess risk of autoimmunity. Analysis of variance, independent samples t test, and a general linear model were used in secondary analyses to test associations of background characteristics and dietary factors with inflammation markers.
    RESULTS: In unadjusted analyses, calprotectin was not associated with risk of islet autoimmunity, whereas HBD-2 in the middle (odds ratio [OR]: 3.23; 95% confidence interval [CI]: 1.03, 10.08) or highest tertile (OR: 3.02; 95% CI: 1.05, 8.69) in comparison to the lowest at 12 mo of age showed borderline association (P-trend = 0.063) with higher risk of islet autoimmunity. Excluding children with cow milk allergy in sensitivity analyses strengthened the association of HBD-2 with islet autoimmunity, whereas adjusting for dietary factors and maternal education weakened it. At age 12 mo, higher fat intake was associated with higher HBD-2 (β: 0.219; 95% CI: 0.110, 0.328) and higher intake of dietary fiber (β: -0.294; 95% CI: -0.510, -0.078), magnesium (β: -0.036; 95% CI: -0.059, -0.014), and potassium (β: -0.003; 95% CI: -0.005, -0.001) with lower HBD-2.
    CONCLUSIONS: Higher HBD-2 in infancy may be associated with higher risk of islet autoimmunity. Dietary factors play a role in gut inflammatory status.
    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
    跳过病变是克罗恩病(CD)特征的神秘空间特征。它们包括具有清晰分界的发炎和相邻的非发炎组织切片。目前,人类胃肠(GI)系统的空间特征缺乏关于微生物组织的清晰度,粘液,组织,和炎症期间的宿主细胞。具有多路复用能力和创新方法的新技术提供了检查CD中发炎和非发炎组织的空间组织的方法,这可能会为诊断开辟新的途径,预后,和治疗。在这次审查中,我们提供了CD患者中空间情境相关性的证据,以及最近发表在炎症过程中的空间性研究中的方法和观点.通过这次审查,我们的目标是为进一步研究解决现有差距提供灵感。
    Skip lesions are an enigmatic spatial feature characterizing Crohn\'s disease (CD). They comprise inflamed and adjacent non-inflamed tissue sections with a clear demarcation. Currently, spatial features of the human gastrointestinal (GI) system lack clarity regarding the organization of microbes, mucus, tissue, and host cells during inflammation. New technologies with multiplexing abilities and innovative approaches provide ways of examining the spatial organization of inflamed and non-inflamed tissues in CD, which may open new avenues for diagnosis, prognosis, and treatment. In this review, we present evidence of the relevance of spatial context in patients with CD and the methods and ideas recently published in studies of spatiality during inflammation. With this review, we aim to provide inspiration for further research to address existing gaps.
    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
    目标:迄今为止,目前尚不清楚环境因素如何影响克罗恩病(CD)风险以及它们如何与生物过程相互作用。这项研究调查了环境暴露与CD风险之间的关联,并评估了它们与疾病前生物标志物的关联。
    方法:我们研究了来自加拿大克罗恩病和结肠炎的4,289名健康的一级亲属(FDRs)的CD患者-遗传,Environmental,微生物(CCC-GEM)项目。回归模型确定了与未来CD发病相关的环境因素及其与疾病前生物学因素的关联。包括通过乳果糖与甘露醇比率(LMR)的尿排泄分数测量的肠道通透性改变;通过粪便钙卫蛋白(FCP)水平引起的肠道炎症;以及通过16SrRNA测序的粪便微生物组组成。
    结果:经过5.62年的中位随访,86FDRs开发了CD。与5-15岁的狗一起生活(HR=0.62;95%CI=0.40-0.96;P=0.034),并且在出生后第一年生活在一个大家庭中(HR=0.43;95%CI=0.21-0.85;P=.016)与降低CD风险相关;而在招募时养鸟(HR=2.78;CI=1.36-5.68;P=.005)与增加CD风险相关。此外,和狗一起生活与LMR降低有关,改变了多个细菌属的相对丰度,并增加了Chao1多样性;而鸟类所有者的FCP水平较高。参与者生命第一年的大家庭与微生物群组成改变有关,而不影响FCP或LMR。
    结论:本研究确定了与CD风险相关的环境变量。这些变量也与屏障功能的改变有关,亚临床炎症,和肠道微生物组组成变化,提示在CD发病机制中的潜在作用。
    OBJECTIVE: To date, it is unclear how environmental factors influence Crohn\'s disease (CD) risk and how they interact with biological processes. This study investigates the association between environmental exposures and CD risk and evaluates their association with pre-disease biomarkers.
    METHODS: We studied 4289 healthy first-degree relatives (FDRs) of patients with CD from the Crohn\'s and Colitis Canada - Genetic, Environmental, Microbial (CCC-GEM) project. Regression models identified environmental factors associated with future CD onset and their association with pre-disease biological factors, including altered intestinal permeability measured by urinary fractional excretion of lactulose to mannitol ratio (LMR); gut inflammation via fecal calprotectin (FCP) levels; and fecal microbiome composition through 16S rRNA sequencing.
    RESULTS: Over a 5.62-year median follow-up, 86 FDRs developed CD. Living with a dog between ages 5 and 15 (hazard ratio [HR], 0.62; 95% confidence interval [CI], 0.40-0.96; P = .034), and living with a large family size in the first year of life (HR, 0.43; 95% CI, 0.21-0.85; P = .016) were associated with decreased CD risk, whereas having a bird at the time of recruitment (HR, 2.78; 95% CI, 1.36-5.68; P = .005) was associated with an increased CD risk. Furthermore, living with a dog was associated with reduced LMR, altered relative abundance of multiple bacterial genera, and increased Chao1 diversity, whereas bird owners had higher FCP levels. Large family during participants\' first year of life was associated with altered microbiota composition without affecting FCP or LMR.
    CONCLUSIONS: This study identifies environmental variables associated with CD risk. These variables were also associated with altered barrier function, subclinical inflammation, and gut microbiome composition shifts, suggesting potential roles in CD pathogenesis.
    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
    炎症性肠病(IBD)是一组与多种致病因素相关的胃肠道慢性炎症,包括免疫反应的失调。效应CD4+T细胞和调节性CD4+T细胞(Treg)是维持耐受性和炎症平衡的核心角色。有趣的是,这些细胞中的遗传修饰与调节特定表型和免疫功能的承诺有关。然而,在IBD进展中控制T辅助细胞致病行为的转录程序仍然未知。在这项研究中,我们的目的是找到控制肠道炎症时效应CD4+T细胞致病行为的主要转录调节因子。为了实现这一目标,我们使用了一个IBD的动物模型,通过将初始CD4+T细胞转移到重组激活基因1(Rag1)缺陷小鼠中,缺乏淋巴细胞。作为一种控制,一组Rag1-/-小鼠接受了整个CD4+T细胞群的转移,其中包括效应T细胞和Treg。当肠道炎症进展时,我们从结肠固有层和脾组织中分离出CD4+T细胞,并进行批量RNA-seq。我们通过比较来自两个实验组的样品鉴定了差异上调和下调的基因。我们在结肠中发现了532个差异表达基因(DEGs),在脾脏中发现了30个DEGs,主要与Th1反应有关,白细胞迁移,以及对固有层T细胞中细胞因子的反应。我们将这些数据整合到基因调控网络中,以识别主调控者,鉴定四个上调的主基因调节因子(Lef1、Dnmt1、Mybl2和Jup)和仅一个下调的主基因调节因子(Foxo3)。通过qRT-PCR分析证实了转录组学分析中观察到的主调节因子的表达改变,发现Lef1和Mybl2上调,但在Dnmt1,Jup,Foxo3这两个主调节因子参与了T细胞功能和细胞周期进程,分别。我们在IBD动物模型中鉴定了与效应CD4+T细胞致病行为相关的两个主要调节基因。这些发现为治疗IBD提供了两个新的潜在分子靶标。
    Inflammatory bowel diseases (IBD) are a group of chronic inflammatory conditions of the gastrointestinal tract associated with multiple pathogenic factors, including dysregulation of the immune response. Effector CD4+ T cells and regulatory CD4+ T cells (Treg) are central players in maintaining the balance between tolerance and inflammation. Interestingly, genetic modifications in these cells have been implicated in regulating the commitment of specific phenotypes and immune functions. However, the transcriptional program controlling the pathogenic behavior of T helper cells in IBD progression is still unknown. In this study, we aimed to find master transcription regulators controlling the pathogenic behavior of effector CD4+ T cells upon gut inflammation. To achieve this goal, we used an animal model of IBD induced by the transfer of naïve CD4+ T cells into recombination-activating gene 1 (Rag1) deficient mice, which are devoid of lymphocytes. As a control, a group of Rag1-/- mice received the transfer of the whole CD4+ T cells population, which includes both effector T cells and Treg. When gut inflammation progressed, we isolated CD4+ T cells from the colonic lamina propria and spleen tissue, and performed bulk RNA-seq. We identified differentially up- and down-regulated genes by comparing samples from both experimental groups. We found 532 differentially expressed genes (DEGs) in the colon and 30 DEGs in the spleen, mostly related to Th1 response, leukocyte migration, and response to cytokines in lamina propria T-cells. We integrated these data into Gene Regulatory Networks to identify Master Regulators, identifying four up-regulated master gene regulators (Lef1, Dnmt1, Mybl2, and Jup) and only one down-regulated master regulator (Foxo3). The altered expression of master regulators observed in the transcriptomic analysis was confirmed by qRT-PCR analysis and found an up-regulation of Lef1 and Mybl2, but without differences on Dnmt1, Jup, and Foxo3. These two master regulators have been involved in T cells function and cell cycle progression, respectively. We identified two master regulator genes associated with the pathogenic behavior of effector CD4+ T cells in an animal model of IBD. These findings provide two new potential molecular targets for treating IBD.
    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:早老素基因突变是阿尔茨海默病的主要病因。然而,人们对它们在肠道中的表达和功能知之甚少。在这项研究中,我们确定PresenilinsPsen1和Psen2是维持肠道稳态的关键分子。
    方法:分析人炎性肠病(IBD)和对照样品的Psen1表达。新产生的肠上皮特异性Psen1缺陷,Psen2缺陷型和诱导型Psen1/Psen2双缺陷型小鼠用于剖析早老素在肠道稳态中的功能作用。
    结果:Psen1表达在实验性肠道炎症和IBD患者中受到调节。在小鼠中诱导的Psen1和Psen2缺失导致快速的体重减轻和肠道炎症的自发发展。小鼠表现出上皮屏障破坏,细菌易位和营养摄取的关键途径失调。浪费疾病与肠道炎症和生态失调无关,由于微生物群的消耗使Psen缺乏的动物免于自发性结肠炎的发展,而不是体重减轻。在分子水平上,缺乏Psen的肠上皮细胞显示Notch信号传导受损和上皮分化失调。
    结论:总体而言,我们的研究提供了证据,证明Psen1和Psen2是肠道稳态的重要守护者,也是IBD促进屏障治疗策略的未来靶点.
    OBJECTIVE: Mutations in presenilin genes are the major cause of Alzheimer\'s disease. However, little is known about their expression and function in the gut. In this study, we identify the presenilins Psen1 and Psen2 as key molecules that maintain intestinal homoeostasis.
    METHODS: Human inflammatory bowel disease (IBD) and control samples were analysed for Psen1 expression. Newly generated intestinal epithelium-specific Psen1-deficient, Psen2-deficient and inducible Psen1/Psen2 double-deficient mice were used to dissect the functional role of presenilins in intestinal homoeostasis.
    RESULTS: Psen1 expression was regulated in experimental gut inflammation and in patients with IBD. Induced deletion of Psen1 and Psen2 in mice caused rapid weight loss and spontaneous development of intestinal inflammation. Mice exhibited epithelial barrier disruption with bacterial translocation and deregulation of key pathways for nutrient uptake. Wasting disease was independent of gut inflammation and dysbiosis, as depletion of microbiota rescued Psen-deficient animals from spontaneous colitis development but not from weight loss. On a molecular level, intestinal epithelial cells lacking Psen showed impaired Notch signalling and dysregulated epithelial differentiation.
    CONCLUSIONS: Overall, our study provides evidence that Psen1 and Psen2 are important guardians of intestinal homoeostasis and future targets for barrier-promoting therapeutic strategies in IBD.
    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
    肠道菌群在炎症性肠病的疾病进展中起着重要作用。虽然益生菌对IBD有效,在小鼠结肠炎模型中,没有多少研究研究它们对肠道菌群组成和免疫调节的影响。我们的研究首次针对副干酪乳杆菌BNCC345679的治疗效果,并探讨了其对肠道微生物群菌群失调的影响,炎性细胞因子,相关的miRNA,VCAM-1,氧化应激,肠道完整性,和粘液屏障。我们发现,对副干酪乳杆菌BNCC345679的口服干预会影响有益微生物分类群的恢复,包括乳酸菌。和阿克曼西亚。,随之而来的是体重的改善,DAI得分,和炎性细胞因子。副干酪乳杆菌BNCC345679减轻氧化应激并增加肠完整性蛋白MUC2和ZO-1的表达。这些结果表明,副干酪乳杆菌BNCC345679具有减轻DSS诱导的结肠炎的能力,并有可能作为缓解IBD的补充。
    The gut microbiota plays an important role in the disease progression of inflammatory bowel disease. Although probiotics are effective against IBD, not many studies have investigated their effects on gut microbiota composition and immunomodulation in mouse colitis models. Our study aimed at the therapeutic effects of Lacticaseibacillus paracasei BNCC345679 for the first time and explored its impact on gut microbiome dysbiosis, inflammatory cytokines, related miRNAs, VCAM-1, oxidative stress, intestinal integrity, and mucus barrier. We found that oral intervention of L. paracasei BNCC345679 affects recovering beneficial microbial taxa, including lactobacillus spp. and akkermansia spp., followed by improved body weight, DAI score, and inflammatory cytokines. L. paracasei BNCC345679 mitigated oxidative stress and increased the expression of intestinal integrity proteins MUC2 and ZO-1. These results suggested that L. paracasei BNCC345679 has the capacity to reduce DSS-induced colitis and has the potential as a supplement for the mitigation of IBD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号