DSS colitis

DSS 结肠炎
  • 文章类型: Journal Article
    目的:结肠炎患者的腹部不适可能部分归因于小肠动力障碍的存在,然而,将结肠炎症与小肠运动联系起来的机制仍未被研究。我们假设结肠炎由于小肠内的肠内分泌细胞(EEC)的损失而导致小肠运动不足,这可以使用5-羟色胺能调节剂来挽救。
    方法:雄性C57BL/6J小鼠,以及过度表达(EECOVER)或缺乏(EECDEL)NeuroD1+肠内分泌细胞的小鼠,暴露于DSS结肠炎(2.5%或5%,持续7天),并通过70kdFITC-葡聚糖荧光转运评估小肠运动性。通过免疫组织化学评估EEC的数量和分化,TUNEL染色,和qRT-PCR。小鼠用5-HT4激动剂普鲁卡洛必利(5mg/kg口服,每天)恢复血清素信号。
    结果:DSS诱导的结肠炎与在小肠中没有明显炎症的情况下发生的显著小肠动力不足相关,并且与EEC密度的显著降低相关。EEC丢失与关键5-羟色胺合成和转运蛋白基因表达的改变有关,包括Tph1、Ddc、还有Slc6a4.重要的是,小鼠过度表达肠内分泌细胞显示出改善的小肠运动,而缺乏EECs的小鼠在暴露于DSS时肠道动力较差。最后,用5-HT4激动剂普鲁卡洛必利治疗暴露于DSS的小鼠可恢复小肠运动并减轻结肠炎。
    结论:实验性DSS结肠炎由于肠内分泌损失而在小鼠中诱导显著的小肠动力障碍,这可以通过EEC的遗传调节或施用5-羟色胺类似物来逆转,为有症状的结肠炎患者提供新的治疗方法。
    OBJECTIVE: The abdominal discomfort experienced by patients with colitis may be attributable in part to the presence of small intestinal dysmotility, yet mechanisms linking colonic inflammation with small-bowel motility remain largely unexplored. We hypothesize that colitis results in small intestinal hypomotility owing to a loss of enteroendocrine cells (EECs) within the small intestine that can be rescued using serotonergic-modulating agents.
    METHODS: Male C57BL/6J mice, as well as mice that overexpress (EECOVER) or lack (EECDEL) NeuroD1+ enteroendocrine cells, were exposed to dextran sulfate sodium (DSS) colitis (2.5% or 5% for 7 days) and small intestinal motility was assessed by 70-kilodalton fluorescein isothiocyanate-dextran fluorescence transit. EEC number and differentiation were evaluated by immunohistochemistry, terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling staining, and quantitative reverse-transcriptase polymerase chain reaction. Mice were treated with the 5-hydroxytryptamine receptor 4 agonist prucalopride (5 mg/kg orally, daily) to restore serotonin signaling.
    RESULTS: DSS-induced colitis was associated with a significant small-bowel hypomotility that developed in the absence of significant inflammation in the small intestine and was associated with a significant reduction in EEC density. EEC loss occurred in conjunction with alterations in the expression of key serotonin synthesis and transporter genes, including Tph1, Ddc, and Slc6a4. Importantly, mice overexpressing EECs revealed improved small intestinal motility, whereas mice lacking EECs had worse intestinal motility when exposed to DSS. Finally, treatment of DSS-exposed mice with the 5-hydroxytryptamine receptor 4 agonist prucalopride restored small intestinal motility and attenuated colitis.
    CONCLUSIONS: Experimental DSS colitis induces significant small-bowel dysmotility in mice owing to enteroendocrine loss that can be reversed by genetic modulation of EEC or administering serotonin analogs, suggesting novel therapeutic approaches for patients with symptomatic colitis.
    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
    炎症性肠病(IBD)的患病率在全球范围内呈上升趋势;然而,其病因尚不完全清楚。患者遗传学,免疫系统,和肠道微生物群被认为是导致IBD的关键因素。临床前动物模型对于更好地理解个体影响因素的重要性至关重要。其中,葡聚糖硫酸钠(DSS)结肠炎模型应用最广泛。DSS治疗诱导肠道炎症和生态失调。然而,其确切的作用方式尚不清楚。为了确定DSS治疗是否诱导微生物群的致病性变化,我们在体外研究了DSS对小鼠微生物群的调节作用。为此,我们在六个重复的连续生物反应器中培养了来自结肠的鼠微生物群。用1%DSS补充三个生物反应器,并通过微生物群分类学和功能性与剩余的PBS处理的对照生物反应器进行比较。使用元蛋白质组学,我们没有发现微生物分类学的显著变化,在门或属水平。没有观察到代谢途径的差异。此外,全球代谢组和靶向短链脂肪酸(SCFA)定量未发现任何DSS相关变化.在没有宿主环境的情况下,DSS对体外微生物功能和分类学的影响可以忽略不计。我们的结果强调DSS结肠炎小鼠模型是研究宿主-微生物群相互作用的合适模型。这可能有助于了解肠道炎症如何在分类和功能水平上调节微生物群。
    The prevalence of inflammatory bowel disease (IBD) is rising globally; however, its etiology is still not fully understood. Patient genetics, immune system, and intestinal microbiota are considered critical factors contributing to IBD. Preclinical animal models are crucial to better understand the importance of individual contributing factors. Among these, the dextran sodium sulfate (DSS) colitis model is the most widely used. DSS treatment induces gut inflammation and dysbiosis. However, its exact mode of action remains unclear. To determine whether DSS treatment induces pathogenic changes in the microbiota, we investigated the microbiota-modulating effects of DSS on murine microbiota in vitro. For this purpose, we cultured murine microbiota from the colon in six replicate continuous bioreactors. Three bioreactors were supplemented with 1% DSS and compared with the remaining PBS-treated control bioreactors by means of microbiota taxonomy and functionality. Using metaproteomics, we did not identify significant changes in microbial taxonomy, either at the phylum or genus levels. No differences in the metabolic pathways were observed. Furthermore, the global metabolome and targeted short-chain fatty acid (SCFA) quantification did not reveal any DSS-related changes. DSS had negligible effects on microbial functionality and taxonomy in vitro in the absence of the host environment. Our results underline that the DSS colitis mouse model is a suitable model to study host-microbiota interactions, which may help to understand how intestinal inflammation modulates the microbiota at the taxonomic and functional levels.
    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
    本研究旨在探讨低剂量壳寡糖(COS)通过调节微生物菌群失调和促炎反应改善葡聚糖硫酸钠(DSS)诱导的慢性结肠炎的潜力。DSS诱导BALB/c小鼠慢性结肠炎(4%w/v,3个周期的5天)给药。将小鼠分为四组:赋形剂,DSS,DSS+美沙拉嗪和DSS+COS。口服COS和美沙拉嗪,每天一次,从第1天到第30天,分别以20mg/kg和50mg/kg的剂量。疾病活动指数(DAI),结肠长度,组织病理学评分,微生物组成,评估促炎细胞因子的表达。COS(20mg/kg,COSLow)给药降低了疾病活动指数,结肠缩短,由DSS引起的显著。此外,COSLow恢复了肠道中改变的微生物组,并抑制了结肠中升高的促炎细胞因子(IL-1和IL-6),以对抗DSS诱导的小鼠慢性结肠炎。此外,COSLow治疗改善了益生菌菌群,从而恢复了肠道稳态。总之,这是第一项通过慢性COSLow治疗Balb/c小鼠DSS诱导的慢性结肠炎来调节微生物菌群失调和促炎反应的研究.因此,以相对低的剂量补充COS对于慢性炎症性肠病可能是有效的。
    The study aimed to investigate the potential of low dose chitooligosaccharide (COS) in ameliorating dextran sodium sulfate (DSS) induced chronic colitis by regulating microbial dysbiosis and pro-inflammatory responses. Chronic colitis was induced in BALB/c mice by DSS (4% w/v, 3 cycles of 5 days) administration. The mice were divided into four groups: vehicle, DSS, DSS + mesalamine and DSS+COS. COS and mesalamine were administered orally, daily once, from day 1 to day 30 at a dose of 20 mg/kg and 50 mg/kg respectively. The disease activity index (DAI), colon length, histopathological score, microbial composition, and pro-inflammatory cytokine expression were evaluated. COS (20 mg/kg, COSLow) administration reduced the disease activity index, and colon shortening, caused by DSS significantly. Furthermore, COSLow restored the altered microbiome in the gut and inhibited the elevated pro-inflammatory cytokines (IL-1 and IL-6) in the colon against DSS-induced chronic colitis in mice. Moreover, COSLow treatment improved the probiotic microflora thereby restoring the gut homeostasis. In conclusion, this is the first study where microbial dysbiosis and pro-inflammatory responses were modulated by chronic COSLow treatment against DSS-induced chronic colitis in Balb/c mice. Therefore, COS supplementation at a relatively low dose could be efficacious for chronic inflammatory bowel 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
    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
    BACKGROUND: The laminin gamma 1 chain (LMγ1) is abundant along the crypt-villus axis in the intestinal basement membrane.
    OBJECTIVE: We investigated whether a serological biomarker of laminin degradation was associated with disease activity in patients with Crohn\'s disease (CD) and in rats with dextran sulfate sodium (DSS)-induced colitis.
    METHODS: Serum samples from CD patients (n = 43), healthy subjects (n = 19), and Sprague Dawley rats receiving 5-6% DSS water for five days and regular drinking water for 11 days were included in this study. The LG1M biomarker, a neo-epitope degradation fragment of the LMγ1 chain generated by matrix metalloproteinases-9 (MMP-9), was measured in serum to estimate the level of laminin degradation.
    RESULTS: Serum LG1M was elevated in CD patients with active and inactive disease compared to healthy subjects (p < 0.0001). LG1M distinguished CD patients from healthy subjects, with an area under the curve (AUC) of 0.81 (p < 0.0001). Serum LG1M was decreased in DSS rats compared to controls 2 days after DSS withdrawal, and increased upon reversal of the disease.
    CONCLUSIONS: Increased serum LG1M in active and inactive CD patients supports the evidence of altered LM expression in both inflamed and non-inflamed tissue. Moreover, lower LG1M levels in the early healing phase of DSS-induced colitis may reflect ongoing mucosal repair.
    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
    目的:炎性损伤后受损的粘膜表面的恢复需要多种再生机制,但仍未明确。以前,我们证明了三叶因子3(TFF3)的修复作用取决于神秘受体,富含亮氨酸的重复序列和免疫球蛋白样结构域含有nogo受体2(LINGO2)。这项研究在肠组织损伤的情况下检查了相关的孤儿受体LINGO3,以确定LINGO家族成员对于粘膜伤口愈合和维持粘膜上皮更新所需的肠干细胞(ISC)隔室是否通常很重要。方法和结果:我们发现LINGO3在人肠上皮细胞上广泛表达,在隐窝小生境内的离散细胞上稀疏表达,包含ISC。功能缺失研究表明,LINGO3参与右旋糖酐硫酸钠(DSS)诱导的结肠炎后正常肠道结构的恢复,并且LINGO3是长效TFF2融合蛋白(TFF2-Fc)的治疗作用所必需的,包括许多对细胞增殖和伤口修复至关重要的信号通路。LINGO3-TFF2蛋白-蛋白相互作用相对较弱,LINGO3仅部分负责TFF2诱导的MAPK信号传导,表明受体复合物的其他未鉴定成分。然而,TFF2或LINGO3的缺乏消除了肠道类器官的出芽/生长,并降低了肠道ISC基因富含亮氨酸重复序列的G蛋白偶联受体5(LGR5)的表达,表明这些蛋白质在组织再生中的同源作用,可能是通过调节隐窝生态位的ISC。结论:我们认为LINGO3在促进粘膜伤口愈合方面具有以前未被重视的作用。
    Aim: Recovery of damaged mucosal surfaces following inflammatory insult requires diverse regenerative mechanisms that remain poorly defined. Previously, we demonstrated that the reparative actions of Trefoil Factor 3 (TFF3) depend upon the enigmatic receptor, leucine rich repeat and immunoglobulin-like domain containing nogo receptor 2 (LINGO2). This study examined the related orphan receptor LINGO3 in the context of intestinal tissue damage to determine whether LINGO family members are generally important for mucosal wound healing and maintenance of the intestinal stem cell (ISC) compartment needed for turnover of mucosal epithelium.Methods and Results: We find that LINGO3 is broadly expressed on human enterocytes and sparsely on discrete cells within the crypt niche, that contains ISCs. Loss of function studies indicate that LINGO3 is involved in recovery of normal intestinal architecture following dextran sodium sulfate (DSS)-induced colitis, and that LINGO3 is needed for therapeutic action of the long acting TFF2 fusion protein (TFF2-Fc), including a number of signaling pathways critical for cell proliferation and wound repair. LINGO3-TFF2 protein-protein interactions were relatively weak however and LINGO3 was only partially responsible for TFF2 induced MAPK signaling suggesting additional un-identified components of a receptor complex. However, deficiency in either TFF2 or LINGO3 abrogated budding/growth of intestinal organoids and reduced expression of the intestinal ISC gene leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5), indicating homologous roles for these proteins in tissue regeneration, possibly via regulation of ISCs in the crypt niche.Conclusion: We propose that LINGO3 serves a previously unappreciated role in promoting mucosal wound healing.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Inflammatory bowel diseases (IBD) are chronic inflammatory disorders with increasing incidence and prevalence worldwide. Here, we investigated thymoquinone (TQ), a naturally occurring phytochemical present in Nigella sativa, for anti-inflammatory effects in colonic inflammation. To address this, we used in vivo (mice) and in vitro (HT-29 cells) models in this investigation. Our results showed that TQ treatment significantly reduced the disease activity index (DAI), myeloperoxidase (MPO) activity, and protected colon microscopic architecture. In addition, TQ also reduced the expression of proinflammatory cytokines and mediators at both the mRNA and protein levels. Further, TQ decreased phosphorylation of the activated mitogen-activated protein kinase (MAPK) signaling pathway and nuclear factor kappa B (NF-κB) proteins and enhanced colon epithelial PPAR-γ transcription factor expression. TQ significantly decreased proinflammatory chemokines (CXCL-1 and IL-8), and mediator (COX-2) mRNA expression in HT-29 cells treated with TNF-α. TQ also increased HT-29 PPAR-γ mRNA, PPAR-γ protein expression, and PPAR-γ promoter activity. These results indicate that TQ inhibits MAPK and NF-κB signaling pathways and transcriptionally regulates PPAR-γ expression to induce potent anti-inflammatory activity in vivo and in vitro models of colon inflammation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Vedolizumab is a widely used and safe therapy in inflammatory bowel disease, particularly in ulcerative colitis (UC), making it a promising candidate for enhanced efficacy by combining it with additional immunomodulatory medications. In this study, we studied the impact of vedolizumab monotreatment vs vedolizumab coadministration with other immunomodulatory drugs on intestinal inflammation and intestinal immune cells in vivo.
    Colon tissue from human patients with UC with active disease or in remission with or without vedolizumab treatment was stained by immunohistochemistry. We reconstituted NOD-SCID-SGM3 mice with human CD34+ cells and treated them with dextran sodium sulfate to induce acute colitis. Mice were treated with vedolizumab alone, or in combination with tacrolimus, ozanimid, or tofacitinib.
    Vedolizumab reduced the number of CD3+ T cells and CD68+ monocytes/macrophages in the colon of patients with UC with active disease. Vedolizumab moderately decreased immune cell numbers in acute dextran sodium sulfate-induced colitis. The combination of vedolizumab with tacrolimus further reduced the number of infiltrating CD3+ T cells and CD68+ monocytes/macrophages and was superior in ameliorating intestinal inflammation when compared to vedolizumab monotreatment. In contrast, cotreatment using vedolizumab with ozanimod or tofacitinib had no additive effect.
    Our data show that vedolizumab reduces the number of innate and adaptive immune cells in the mucosa of patients with UC. Further, the combination of vedolizumab with tacrolimus was more efficient to reduce immune cell numbers and to increase therapeutic efficacy than vedolizumab monotreatment. This finding indicates that combination treatment using these two drugs may be beneficial for patients who do not respond to vedolizumab monotherapy.
    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
    Glucocorticoids are the first line treatment for the flare-ups of inflammatory bowel disease, but they have significant limitations. The objective of this study is to investigate whether glucocorticoid epithelial actions contribute to such limitations.
    Intestinal epithelium glucocorticoid receptor knockout mice (Nr3c1ΔIEC ) received dextran sulfate sodium (DSS) to induce colitis. Inflammatory status was assessed by morphological and biochemical methods, and corticoid production was measured in colonic explants. Some mice were administered budesonide.
    After 7 days of DSS Nr3c1ΔIEC , mice exhibited 23.1% lower disease activity index (DAI) and 37% lower diarrheal score than WT mice, with decreased weight loss in days 5-7 of colitis, attenuated tissue damage, reduced colonic expression of S100A9 and STAT3 phosphorylation, and a better overall state. Ki67 immunoreactivity was increased at the crypt base, indicating enhanced epithelial proliferation. Mice administered budesonide (6 μg·day-1 PO) showed modest antiinflammatory effects but increased weight loss, which was prevented in knockout mice. Epithelial deletion of the glucocorticoid receptor also protected mice in a protracted colitis protocol. Conversely, knockout mice presented a worse status compared to the control group at 1 day post DSS. In a separate experiment, colonic corticosterone production was shown to be significantly increased in knockout mice at 7 days of colitis but not at earlier stages.
    The intestinal epithelial glucocorticoid receptor has deleterious effects in experimental colitis induced by DSS, probably related to inhibition of epithelial proliferative responses leading to impaired wound healing and reduced endogenous corticosterone production.
    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
    Inflammatory bowel disease (IBD) is a complex disease which leads to life-threatening complications and decreased quality of life. The dextran sulfate sodium (DSS) colitis model in mice is known for rapid screening of candidate compounds. Efficacy assessment in this model relies partly on microscopic semiquantitative scoring, which is time-consuming and subjective. We hypothesized that deep learning artificial intelligence (AI) could be used to identify acute inflammation in H&E-stained sections in a consistent and quantitative manner. Training sets were established using ×20 whole slide images of the entire colon. Supervised training of a Convolutional Neural Network (CNN) was performed using a commercial AI platform to detect the entire colon tissue, the muscle and mucosa layers, and 2 categories within the mucosa (normal and acute inflammation E1). The training sets included slides of naive, vehicle-DSS and cyclosporine A-DSS mice. The trained CNN was able to segment, with a high level of concordance, the different tissue compartments in the 3 groups of mice. The segmented areas were used to determine the ratio of E1-affected mucosa to total mucosa. This proof-of-concept work shows promise to increase efficiency and decrease variability of microscopic scoring of DSS colitis when screening candidate compounds for IBD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号