DSS colitis

DSS 结肠炎
  • 文章类型: Journal Article
    炎症性肠病(IBD)是一种突出的慢性免疫介导的炎症性疾病,然而其病因仍然知之甚少,涵盖了遗传学之间复杂的相互作用,豁免权,和肠道微生物组。这项研究发现了一种新的结肠炎相关风险基因,即Ring1a,调节粘膜免疫反应和肠道微生物群。Ring1a缺乏通过损害免疫系统而加剧结肠炎。同时,Ring1a缺乏导致Prevotella属为主的致病微环境,可以水平传播给共同饲养的野生型(WT)小鼠,因此加剧葡聚糖硫酸钠(DSS)诱导的结肠炎。此外,我们发现了一种潜在的机制,将Ring1aKO小鼠中改变的微生物群与降低的IgA水平联系起来,我们证明了甲硝唑可以改善Ring1aKO小鼠的结肠炎进展,可能是通过减少普雷沃氏菌属的丰度。我们还阐明了DSS结肠炎的免疫景观,并揭示了与Ring1a缺乏相关的肠道免疫稳态的破坏。总的来说,这些发现强调了Ring1a是结肠炎的前瞻性候选风险基因,并提示甲硝唑是临床治疗普雷沃氏菌属为主的结肠炎的潜在治疗选择.
    我们发现PcG蛋白Ring1a可能是结肠炎的新风险基因。Ring1a缺乏通过调节粘膜免疫系统和结肠微生物生态导致结肠炎加重。
    Inflammatory bowel disease (IBD) represents a prominent chronic immune-mediated inflammatory disorder, yet its etiology remains poorly comprehended, encompassing intricate interactions between genetics, immunity, and the gut microbiome. This study uncovers a novel colitis-associated risk gene, namely Ring1a, which regulates the mucosal immune response and intestinal microbiota. Ring1a deficiency exacerbates colitis by impairing the immune system. Concomitantly, Ring1a deficiency led to a Prevotella genus-dominated pathogenic microenvironment, which can be horizontally transmitted to co-housed wild type (WT) mice, consequently intensifying dextran sodium sulfate (DSS)-induced colitis. Furthermore, we identified a potential mechanism linking the altered microbiota in Ring1aKO mice to decreased levels of IgA, and we demonstrated that metronidazole administration could ameliorate colitis progression in Ring1aKO mice, likely by reducing the abundance of the Prevotella genus. We also elucidated the immune landscape of DSS colitis and revealed the disruption of intestinal immune homeostasis associated with Ring1a deficiency. Collectively, these findings highlight Ring1a as a prospective candidate risk gene for colitis and suggest metronidazole as a potential therapeutic option for clinically managing Prevotella genus-dominated colitis.
    We found that PcG protein Ring1a could be a new risk gene for colitis. Ring1a deficiency causes aggravated colitis by regulating the mucosal immune system and colonic microbial ecology.
    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
    食物过敏(FA)已成为全球性的食品安全问题。有证据表明,炎症性肠病(IBD)可以增加FA的发病率,但它主要是基于流行病学研究。动物模型对于揭示所涉及的机制至关重要。然而,葡聚糖硫酸钠(DSS)诱导的IBD模型可能会导致大量动物损失。为了更好地研究IBD对FA的影响,本研究旨在建立一个符合IBD和FA症状的小鼠模型。首先,我们通过监测生存率比较了三种DSS诱导的结肠炎模型,疾病活动指数,结肠长度,和脾脏指数,然后用4%的7天给药消除结肠炎模型,因为死亡率高。此外,我们评估了所选择的两种模型对FA和肠组织病理学的建模效果,发现3%DSS给药7天的结肠炎模型和DSS长期给药的结肠炎模型的建模效果相似.然而,为了动物生存的原因,我们推荐长期使用DSS的结肠炎模型.
    Food allergy (FA) has become a global food safety issue. Evidence suggests that inflammatory bowel disease (IBD) can increase the incidence of FA, but it is mostly based on epidemiological studies. An animal model is pivotal for unraveling the mechanisms involved. However, dextran sulfate sodium (DSS)-induced IBD models may cause substantial animal losses. To better investigate the effect of IBD on FA, this study aimed to establish a murine model to fit both IBD and FA symptoms. Firstly, we compared three DSS-induced colitis models by monitoring survival rate, disease activity index, colon length, and spleen index, and then eliminated the colitis model with a 7-day administration of 4% due to high mortality. Moreover, we evaluated the modeling effects on FA and intestinal histopathology of the two models selected and found the modeling effects were similar in both the colitis model with a 7-day administration of 3% DSS and the colitis model with long-term administration of DSS. However, for animal survival reasons, we recommend the colitis model with long-term administration of DSS.
    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(IBDs)包括克罗恩病(CD)和溃疡性结肠炎(UC),其特征是慢性和衰弱性肠道炎症。肠道细菌群落的改变与IBD的发生和进展密切相关。肠道病毒,主要由细菌病毒(噬菌体,噬菌体),被认为是调节和塑造肠道微生物群落的重要因素。虽然已经在IBD患者中观察到肠道病毒改变,这些病毒对IBD患者相关的细菌群落改变和炎症反应增强的作用仍在很大程度上未知.
    这里,我们进行了体内微生物交叉感染实验,以观察从UC患者和健康对照中分离的粪便病毒样颗粒(VLP)对人类微生物群相关(HMA)小鼠实验性结肠炎的细菌多样性和严重程度的影响.鸟枪宏基因组学证实,几种噬菌体被转移到HMA小鼠中,导致肠道病毒的治疗特异性改变。来自健康和UC患者的VLP也改变了这些小鼠的肠道细菌多样性,在实验性结肠炎期间放大的效应。从UC患者分离的VLP特异性改变了先前与IBD进展有关的几种细菌分类群的相对丰度。此外,UCVLP给药增加了HMA小鼠结肠炎的严重程度,如结肠长度缩短和促炎细胞因子产生增加所示。重要的是,这种效应依赖于完整的VLP。
    我们的发现建立在最近的文献上,表明噬菌体是肠道细菌群落的动态调节因子,并暗示肠道病毒在调节肠道炎症和疾病中。视频摘要。
    BACKGROUND: Inflammatory bowel diseases (IBDs) including Crohn\'s disease (CD) and ulcerative colitis (UC) are characterized by chronic and debilitating gut inflammation. Altered bacterial communities of the intestine are strongly associated with IBD initiation and progression. The gut virome, which is primarily composed of bacterial viruses (bacteriophages, phages), is thought to be an important factor regulating and shaping microbial communities in the gut. While alterations in the gut virome have been observed in IBD patients, the contribution of these viruses to alterations in the bacterial community and heightened inflammatory responses associated with IBD patients remains largely unknown.
    RESULTS: Here, we performed in vivo microbial cross-infection experiments to follow the effects of fecal virus-like particles (VLPs) isolated from UC patients and healthy controls on bacterial diversity and severity of experimental colitis in human microbiota-associated (HMA) mice. Shotgun metagenomics confirmed that several phages were transferred to HMA mice, resulting in treatment-specific alterations in the gut virome. VLPs from healthy and UC patients also shifted gut bacterial diversity of these mice, an effect that was amplified during experimental colitis. VLPs isolated from UC patients specifically altered the relative abundance of several bacterial taxa previously implicated in IBD progression. Additionally, UC VLP administration heightened colitis severity in HMA mice, as indicated by shortened colon length and increased pro-inflammatory cytokine production. Importantly, this effect was dependent on intact VLPs.
    CONCLUSIONS: Our findings build on recent literature indicating that phages are dynamic regulators of bacterial communities in the gut and implicate the intestinal virome in modulating intestinal inflammation and disease. Video Abstract.
    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
    肠道菌群已成为炎症性肠病(IBD)治疗的新靶点。益生菌因其有益作用而闻名,并在IBD的临床治疗和结肠炎的动物模型中显示出良好的疗效。然而,这些益生菌如何促进IBD的改善在很大程度上是未知的。在目前的研究中,通过口服植物乳杆菌菌株治疗DSS诱导的小鼠结肠炎模型,以研究其对结肠炎的影响。结果表明,植物乳杆菌菌株改善了菌群失调,并增强了与短链脂肪酸(SCFA)生产相关的有益菌的丰度。此外,植物乳杆菌菌株降低了促炎细胞因子的水平,即,IL-17A,IL-17F,IL-6,IL-22和TNF-α,增加抗炎细胞因子的水平,即,TGF-β,IL-10.我们的结果表明,植物乳杆菌菌株具有益生菌作用,可以通过调节常驻肠道微生物群和免疫反应来改善小鼠的DSS结肠炎。
    Gut microbiota has become a new therapeutic target in the treatment of inflammatory Bowel Disease (IBD). Probiotics are known for their beneficial effects and have shown good efficacy in the clinical treatment of IBD and animal models of colitis. However, how these probiotics contribute to the amelioration of IBD is largely unknown. In the current study, the DSS-induced mouse colitis model was treated with oral administration of Lactobacillus plantarum strains to investigate their effects on colitis. The results indicated that the L. plantarum strains improved dysbiosis and enhanced the abundance of beneficial bacteria related to short-chain fatty acids (SCFAs) production. Moreover, L. plantarum strains decreased the level of pro-inflammatory cytokines, i.e., IL-17A, IL-17F, IL-6, IL-22, and TNF-α and increased the level of anti-inflammatory cytokines, i.e., TGF-β, IL-10. Our result suggests that L. plantarum strains possess probiotic effects and can ameliorate DSS colitis in mice by modulating the resident gut microbiota and immune response.
    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
    l-Arabinose is a monosaccharide extracted from plants or fibers, which is known to have a variety of functional properties. In this study, we aim to investigate whether l-arabinose could inhibit colitis by modulating gut microbiota. l-Arabinose was administered in mice daily in a dextran sodium sulfate (DSS)-induced colitis model. The histological analysis, disease index, and the expression of inflammatory genes were measured. 16S-rRNA sequence analysis was performed to investigate gut microbiota. Intriguingly, we found that l-arabinose could repress DSS-induced colitis and inhibit p38-/p65-dependent inflammation activation. Besides that, our data revealed that l-arabinose-modulated DSS-induced gut microbiota were disturbed. Additionally, the perturbed gut microbiota was responsible for the suppressive effects of l-arabinose on DSS-induced colitis treated with antibiotics. Lastly, Caco-2 cells were used to confirm the protective effects of l-arabinose in colitis or inflammatory bowel disease. As expected, the protein expression levels in Caco-2 cells of pro-inflammatory genes, which were treated with l-arabinose and incubated with or without tumor necrosis factor alpha. Our work suggested that l-arabinose exerts anti-inflammation effects in DSS-induced colitis. These beneficial effects have correlations with the composition, diversity, and abundance of the gut microbiota regulated by l-arabinose. l-Arabinose could be a remarkable candidate as a functional food or novel therapeutic strategy for intestinal health.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Tim-3 is involved in the physiopathology of inflammatory bowel disease (IBD), but the underlying mechanism is unknown. Here, we demonstrated that, in mouse with DSS colitis, Tim-3 inhibited the polarization of pathogenic pro-inflammatory M1 macrophages, while Tim-3 downregulation or blockade resulted in an increased M1 response. Adoptive transfer of Tim-3-silenced macrophages worsened DSS colitis and enhanced inflammation, while Tim-3 overexpression attenuated DSS colitis by decreasing the M1 macrophage response. Co-culture of Tim-3-overexpressing macrophages with intestinal lymphocytes decreased the pro-inflammatory response. Tim-3 shaped intestinal macrophage polarization may be TLR-4 dependent since Tim-3 blockade failed to exacerbate colitis or increase M1 macrophage response in the TLR-4 KO model. Finally, Tim-3 signaling inhibited phosphorylation of IRF3, a TLR-4 downstream transcriptional factor regulating macrophage polarization. A better understanding of this pathway may shed new light on colitis pathogenesis and result in a new therapeutic strategy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号