Intestinal homeostasis

肠道稳态
  • 文章类型: Journal Article
    背景:溃疡性结肠炎(UC)是由病因不确定的胃肠道内持续的炎症过程定义的。目前的治疗方法在解决氧化应激的能力方面受到限制,炎症,屏障功能恢复,以协调的方式调节肠道菌群,以维持肠道稳态。
    结果:该研究涉及通过铁离子介导的姜黄素氧化偶联来构建金属-酚类纳米酶(Cur-Fe)。Cur-Fe纳米酶表现出超氧化物歧化酶(SOD)样和•OH清除活性,证明了在体外维持细胞内氧化还原平衡的显着抗炎和抗氧化特性。从大肠杆菌Nissle1917(EcN)中汲取灵感,随后通过将Cur-Fe整合到EcN膜(EM)中来开发仿生Cur-Fe纳米酶(CF@EM),以提高Cur-Fe纳米酶的体内靶向能力和治疗效果。口服时,CF@EM在DSS诱导的结肠炎模型中显示出强的定植发炎的结肠和恢复肠氧化还原平衡和屏障功能的能力。重要的是,CF@EM通过增强细菌多样性和将组成结构转变为抗炎表型来影响肠道微生物组朝向有益状态。此外,肠道微生物代谢产物的分析支持CF@EM的疗效与胆汁酸代谢密切相关的观点。
    结论:受肠道微生物的启发,我们已经成功地合成了一种具有抑制炎症和恢复肠道稳态能力的仿生Cur-Fe纳米酶。总的来说,没有明显的全身毒性,这项工作为靶向口服纳米药物治疗溃疡性结肠炎提供了前所未有的机会.
    BACKGROUND: Ulcerative colitis (UC) is defined by persistent inflammatory processes within the gastrointestinal tract of uncertain etiology. Current therapeutic approaches are limited in their ability to address oxidative stress, inflammation, barrier function restoration, and modulation of gut microbiota in a coordinated manner to maintain intestinal homeostasis.
    RESULTS: This study involves the construction of a metal-phenolic nanozyme (Cur-Fe) through a ferric ion-mediated oxidative coupling of curcumin. Cur-Fe nanozyme exhibits superoxide dismutase (SOD)-like and •OH scavenging activities, demonstrating significant anti-inflammatory and anti-oxidant properties for maintaining intracellular redox balance in vitro. Drawing inspiration from Escherichia coli Nissle 1917 (EcN), a biomimetic Cur-Fe nanozyme (CF@EM) is subsequently developed by integrating Cur-Fe into the EcN membrane (EM) to improve the in vivo targeting ability and therapeutic effectiveness of the Cur-Fe nanozyme. When orally administered, CF@EM demonstrates a strong ability to colonize the inflamed colon and restore intestinal redox balance and barrier function in DSS-induced colitis models. Importantly, CF@EM influences the gut microbiome towards a beneficial state by enhancing bacterial diversity and shifting the compositional structure toward an anti-inflammatory phenotype. Furthermore, analysis of intestinal microbial metabolites supports the notion that the therapeutic efficacy of CF@EM is closely associated with bile acid metabolism.
    CONCLUSIONS: Inspired by gut microbes, we have successfully synthesized a biomimetic Cur-Fe nanozyme with the ability to inhibit inflammation and restore intestinal homeostasis. Collectively, without appreciable systemic toxicity, this work provides an unprecedented opportunity for targeted oral nanomedicine in the treatment of ulcerative 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
    背景:人类饮食中塑料含量的增加正在引起公众对纳米塑料(NP)颗粒潜在风险的关注,可以从塑料碎片的降解中出现。NP摄入对炎症性肠病(IBD)患者构成特殊风险,因为受损的上皮屏障可能促进NP易位。
    方法:体外,将骨髓源性巨噬细胞(BMDMs)暴露于25nm聚甲基丙烯酸酯(PMMA)或50nm聚苯乙烯(PS)颗粒,以评估形态变化以及促炎和抗炎基因表达的改变.在体内,小鼠在诱导急性葡聚糖硫酸钠(DSS)结肠炎前接受PMMANP颗粒6个月,以研究NP对肠道健康和炎症的影响.
    结果:在BMDMs中,PMMA和PSNP暴露诱导了形态学变化,表明以变形虫细胞形状增大为特征的促炎表型。它还引发了炎症反应,由促炎细胞因子如Tnfa和Il6的表达增加指示。出乎意料的是,长期施用PMMANP不会影响小鼠的肠上皮屏障或加重急性DSS诱导的结肠炎.结肠镜检查和组织学分析显示没有NP相关的变化,提示对肠道健康或炎症的不利影响。
    结论:我们的动物模型研究结果为IBD患者关于NP摄入的影响提供了一些保证。然而,生活方式和饮食习惯的变化可能导致某些人的塑料摄入量显着增加,引起人们对终身塑料消费潜在的长期胃肠道影响的担忧。
    BACKGROUND: The increasing presence of plastics in the human diet is raising public concern about the potential risks posed by nanoplastic (NP) particles, which can emerge from the degradation of plastic debris. NP ingestion poses particular risks to individuals with inflammatory bowel disease (IBD), as compromised epithelial barriers may facilitate NP translocation.
    METHODS: In vitro, bone-marrow-derived macrophages (BMDMs) were exposed to 25 nm polymethacrylate (PMMA) or 50 nm polystyrene (PS) particles to assess morphological changes and alterations in pro- and anti-inflammatory gene expression. In vivo, mice received PMMA NP particles for 6 months before acute dextran sodium sulfate (DSS) colitis was induced to investigate NP impacts on intestinal health and inflammation.
    RESULTS: PMMA and PS NP exposure in BMDMs induced morphological changes indicative of a proinflammatory phenotype characterized by enlarged amoeboid cell shapes. It also triggered an inflammatory response, indicated by increased expression of proinflammatory cytokines such as Tnfa and Il6. Unexpectedly, long-term PMMA NP administration did not affect the intestinal epithelial barrier or exacerbate acute DSS-induced colitis in mice. Colonoscopy and histological analysis revealed no NP-related changes, suggesting adverse effects on intestinal health or inflammation.
    CONCLUSIONS: Our findings from animal models offer some reassurance to IBD patients regarding the effects of NP ingestion. However, variations in lifestyle and dietary habits may lead to significantly higher plastic intake in certain individuals, raising concerns about potential long-term gastrointestinal effects of lifelong plastic consumption.
    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
    昆虫内脏为微生物定植提供了独特的栖息地,肠道细菌可能为它们的宿主提供许多好处。尽管肠球菌已成为昆虫中主要的肠道共生细菌之一,它在肠道内各种壁龛中的建立并没有得到很好的表征。在这项研究中,将肠球菌接种到家蚕(BombyxmoriL.)中以研究其生物学功能。基于基因组的分析显示,其成功定植与粘附基因(ebpA,ebpC,efaA,srtC,和SCM)。该细菌没有改变相关代谢酶的活性或肠屏障功能。然而,Att2、CecA、Lys和Lys提出了宿主对共生E.mundtii免疫的潜在适应性机制。此外,16S宏基因组学分析显示,接种后,蚕肠道中E.mundtii的相对丰度显着增加。肠道微生物组表现出明显的异质性,肠道微生物组健康指数升高,微生物菌群失调指数降低,治疗组潜在致病性低。此外,E.mundtii增强了宿主肠道中碳水化合物的分解。总的来说,E.mundtii是昆虫的有益微生物,通过提供竞争优势来促进肠道稳态。这种特征有助于E.mundtii控制复杂的微生物环境,并在鳞翅目中保持流行,可能促进双方之间的长期共生。本研究有助于阐明鳞翅目昆虫肠道中E.mundtii的生态位,并进一步揭示其在昆虫宿主中的潜在作用。
    Insect guts offer unique habitats for microbial colonization, with gut bacteria potentially offering numerous benefits to their hosts. Although Enterococcus has emerged as one of the predominant gut commensal bacteria in insects, its establishment in various niches within the gut has not been characterized well. In this study, Enterococcus mundtii was inoculated into the silkworm (Bombyx mori L.) to investigate its biological functions. Genome-based analysis revealed that its successful colonization is related to adherence genes (ebpA, ebpC, efaA, srtC, and scm). This bacterium did not alter the activities of related metabolic enzymes or the intestinal barrier function. However, significant changes in the gene expressions levels of Att2, CecA, and Lys suggest potential adaptive mechanisms of host immunity to symbiotic E. mundtii. Moreover, 16S metagenomics analysis revealed a significant increase in the relative abundance of E. mundtii in the intestines of silkworms following inoculation. The intestinal microbiome displayed marked heterogeneity, an elevated gut microbiome health index, a reduced microbial dysbiosis index, and low potential pathogenicity in the treatment group. Additionally, E. mundtii enhanced the breakdown of carbohydrates in host intestines. Overall, E. mundtii serves as a beneficial microbe for insects, promoting intestinal homeostasis by providing competitive advantage. This characteristic helps E. mundtii dominate complex microbial environments and remain prevalent across Lepidoptera, likely fostering long-term symbiosis between the both parties. The present study contributes to clarifying the niche of E. mundtii in the intestine of lepidopteran insects and further reveals its potential roles in their insect hosts.
    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
    我们先前的研究表明,重金属铅(Pb)暴露会加剧高脂饮食(HFD)诱导的代谢损伤,并显着消耗肠道微生物群衍生的代谢产物短链脂肪酸(SCFA)水平。然而,目前尚不清楚SCFA是否是促进铅暴露后不良后果的关键代谢产物。在这项研究中,我们探索了外源补充乙酸盐的效果,丙酸盐,和丁酸盐对铅暴露的HFD小鼠代谢紊乱模型的影响。我们发现三种SCFA干预措施可以减轻糖脂代谢紊乱和肝损伤,丁酸盐在改善肥胖相关症状方面效果最佳。所有三个SCFA都促进了Muribaculaceae和Muibaculum的丰度,醋酸盐特别丰富的Christensenellaceae,Blautia,和Ruminococus,和丁酸盐特别富含副曲霉,Rikenella,Prevotellaceae_UCG-001和拟杆菌,促进了SCFA生产形成良性循环的积极推进。此外,丁酸抑制革兰氏阴性菌大肠杆菌-志贺氏菌。所有这些事件通过G蛋白偶联受体(GPR)/组蛋白脱乙酰酶3(HDAC3)和脂多糖(LPS)/toll样受体4(TLR4)/核因子κ-B(NF-κB)途径之间的串扰减轻了肠道Th17/Treg失衡和炎症反应,并最终改善了肠道屏障功能。SCFA进一步上调单羧酸转运蛋白1(MCT1)和GPR43/腺苷5'-单磷酸活化蛋白激酶(AMPK)途径以抑制肝脂质积累。总的来说,SCFA,尤其是丁酸,是一种有效的调节剂,通过靶向肠道微生态来改善暴露于重金属的肥胖个体的代谢紊乱。
    Our previous study showed that heavy metal lead (Pb) exposure exacerbates high-fat-diet (HFD)-induced metabolic damage and significantly depletes the gut microbiota-derived metabolite short-chain fatty acid (SCFA) levels. However, it remains unclear whether SCFA is a key metabolite involved in accelerating adverse consequences after Pb exposure. In this study, we explored the effects of exogenous supplementation of acetate, propionate, and butyrate on a metabolic disorder model in Pb-exposed HFD mice. We found that three SCFA interventions attenuated glycolipid metabolism disorders and liver damage, with butyrate performing the best effects in improving obesity-related symptoms. All three SCFA promoted the abundance of Muribaculaceae and Muribaculum, acetate specifically enriched Christensenellaceae, Blautia, and Ruminococcus, and butyrate specifically enriched Parasutterella, Rikenella, Prevotellaceae_UCG-001, and Bacteroides, which contributed to the positive promotion of SCFA production forming a virtuous cycle. Besides, butyrate inhibited Gram-negative bacteria Escherichia-Shigella. All of these events alleviated the intestinal Th17/Treg imbalance and inflammatory response through crosstalk between the G protein-coupled receptor (GPR)/histone deacetylase 3 (HDAC3) and lipopolysaccharide (LPS)/toll-like receptors 4 (TLR4)/nuclear factor κ-B (NF-κB) pathways and ultimately improved the intestinal barrier function. SCFA further upregulated the monocarboxylate transporter 1 (MCT1) and GPR43/adenosine 5\'-monophosphate-activated protein kinase (AMPK) pathways to inhibit hepatic lipid accumulation. Overall, SCFA, especially butyrate, is an effective modulator to improve metabolic disorders in obese individuals exposed to heavy metals by targeting gut microecology.
    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
    背景:抑郁症不仅损害中枢神经系统,还包括主机的外围系统。肠道菌群已被证明与抑郁症的发病机制有关。逍遥散(XYS)在中国已有一千多年的历史,用于治疗抑郁症,大大缓解焦虑,认知障碍,尤其是胃肠功能紊乱.然而,它仍然只是划伤XYS的抗抑郁机制的表面。
    目的:本研究旨在从“微生物群-肠-脑”轴的角度阐明XYS的作用机制。
    方法:我们首先评估了XYS对慢性不可预知轻度应激(CUMS)引起的抑郁大鼠宏观行为的影响。其次,利用菌群失调模型揭示了XYS对抑郁大鼠肠道稳态的影响。随后,通过16SrRNA基因测序技术和分子生物学方法证明了其潜在机制。最后,从“微生物群-肠-脑”的角度对XYS的抗抑郁作用进行了相关分析和可视化。
    结果:我们的数据表明XYS改善了CUMS大鼠的抑郁样症状,部分取决于肠道微生物群的存在。此外,我们说明了XYS逆转了CUMS诱导的抑郁大鼠的肠道菌群失调,降低了拟杆菌/Firmicutes比率和拟杆菌的丰度,和棒状杆菌,同时增加乳杆菌和阿德氏杆菌的丰度。拟杆菌的显着富集和脂多糖(LPS)的水平表明抑郁症损害了免疫反应和肠道屏障。机械上,XYS显著下调抑郁大鼠结肠和脑组织TLR4/NLRP3信号通路相关因子的表达水平。此外,XYS显着增加了claudin1和ZO-1的水平,表明XYS积极维持了肠道和血脑屏障(BBB)的完整性。
    结论:我们的研究通过“微生物群-TLR4/NLRP3信号通路-屏障”的镜头,为XYS的抗抑郁作用提供了见解,为提高临床效率和丰富药物选择提供基础,并有助于我们了解中药治疗抑郁症的机制。
    BACKGROUND: Depression impairs not only central nervous system, but also peripheral systems of the host. Gut microbiota have been proved to be involved in the pathogenesis of depression. Xiaoyaosan (XYS) has a history of over a thousand years in China for treating depression, dramatically alleviating anxiety, cognitive disorders, and especially gastrointestinal dysfunctions. Yet, it still just scratches the surface of the anti-depression mechanisms of XYS.
    OBJECTIVE: This study aims to elucidate the mechanism of actions of XYS from the perspective of \"microbiota-gut-brain\" axis.
    METHODS: We firstly evaluated the effects of XYS on the macroscopic behaviors of depressed rats that induced by chronic unpredictable mild stress (CUMS). Secondly, the effects of XYS on intestinal homeostasis of depressed rats were revealed by using dysbacteriosis model. Subsequently, the underlying mechanisms were demonstrated by 16S rRNA gene sequencing technology and molecular biology methods. Finally, correlation analysis and visualization of the anti-depression effects of XYS were performed from the \"microbiota - gut - brain\" perspective.
    RESULTS: Our data indicated that XYS ameliorated the depression-like symptoms of CUMS rats, partly depending on the presence of gut microbiota. Furthermore, we illustrated that XYS reversed CUMS-induced gut dysbiosis of depressed rats in terms of decreasing the Bacteroidetes/Firmicutes ratio and the abundances of Bacteroides, and Corynebacterium, while increasing the abundances of Lactobacillus and Adlercreutzia. The significant enrichment of Bacteroides and the level of lipopolysaccharides (LPS) suggested that depression damaged the immune responses and gut barrier. Mechanistically, XYS significantly down-regulated the expression levels of factors that involved in TLR4/NLRP3 signaling pathway in the colon and brain tissues of depressed rats. In addition, XYS significantly increased the levels of claudin 1 and ZO-1, showing that XYS positively maintained the integrity of gut and blood-brain barriers (BBB).
    CONCLUSIONS: Our study offers insights into the anti-depression effects of XYS through a lens of \"microbiota-TLR4/NLRP3 signaling pathway-barriers\", providing a foundation for enhancing clinical efficiency and enriching drug selection, and contributing to our understanding of the mechanisms of traditional Chinese medicines (TCMs) in treating depression.
    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
    肠内分泌细胞(EEC)以其全身激素作用而闻名,尤其是在调节食欲和血糖方面。人们对EEC生产的产品如何调节肠道内的局部环境知之甚少。这里,我们专注于EECs与其他肠细胞之间的旁分泌相互作用,因为它们调节肠稳态和生理的三个基本方面:1)肠干细胞功能和增殖;2)营养吸收;和3)粘膜屏障功能。我们还讨论了EEC表达多种激素的能力,描述体外和体内模型以研究EEC,并考虑胃肠道疾病中EECs是如何改变的。
    Enteroendocrine cells (EECs) are well-known for their systemic hormonal effects, especially in the regulation of appetite and glycemia. Much less is known about how the products made by EECs regulate their local environment within the intestine. Here, we focus on paracrine interactions between EECs and other intestinal cells as they regulate three essential aspects of intestinal homeostasis and physiology: 1) intestinal stem cell function and proliferation; 2) nutrient absorption; and 3) mucosal barrier function. We also discuss the ability of EECs to express multiple hormones, describe in vitro and in vivo models to study EECs, and consider how EECs are altered in GI disease.
    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
    肠上皮细胞(IECs)是通过自我更新维持肠道稳态的关键,扩散,分化,和调节细胞死亡。虽然细胞凋亡和坏死被认为是不同的途径,它们错综复杂的相互作用仍然难以捉摸。在这项研究中,我们报道Mettl3介导的m6A修饰通过阻止上皮细胞死亡维持肠道稳态.Mettl3敲除诱导IECs中的细胞凋亡和坏死。用特异性抑制剂靶向不同的细胞死亡模式揭示了RIPK1激酶活性对于Mettl3敲除引发的细胞死亡至关重要。机械上,这是通过m6A介导的Atf3转录调节而发生的,Atf3是一种直接与Cflar结合的转录因子,编码抗细胞死亡蛋白cFLIP的基因。cFLIP抑制RIPK1活性,从而抑制下游凋亡和坏死信号传导。一起,这些发现描述了METTL3-ATF3-cFLIP轴通过阻断RIPK1活性在肠上皮稳态调节中的重要作用.
    Intestinal epithelial cells (IECs) are pivotal for maintaining intestinal homeostasis through self-renewal, proliferation, differentiation, and regulated cell death. While apoptosis and necroptosis are recognized as distinct pathways, their intricate interplay remains elusive. In this study, we report that Mettl3-mediated m6A modification maintains intestinal homeostasis by impeding epithelial cell death. Mettl3 knockout induces both apoptosis and necroptosis in IECs. Targeting different modes of cell death with specific inhibitors unveils that RIPK1 kinase activity is critical for the cell death triggered by Mettl3 knockout. Mechanistically, this occurs via the m6A-mediated transcriptional regulation of Atf3, a transcription factor that directly binds to Cflar, the gene encoding the anti-cell death protein cFLIP. cFLIP inhibits RIPK1 activity, thereby suppressing downstream apoptotic and necroptotic signaling. Together, these findings delineate the essential role of the METTL3-ATF3-cFLIP axis in homeostatic regulation of the intestinal epithelium by blocking RIPK1 activity.
    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
    去泛素酶(DUB)对于维持蛋白质稳态和将蛋白质组装成功能复合物至关重要。尽管人们对DUBs的生物学功能越来越感兴趣,DUB在调节肠干细胞(ISC)和肠道稳态中的作用仍然未知。这里,我们通过诱导敲低成人中肠ISC和肠母细胞(EB)中DUBs的表达进行体内RNAi筛选,以鉴定果蝇中肠道稳态的DUB调节因子。我们筛选了43个DUB,并确定了ISC稳态所需的8个DUB。usp1,CG7857,usp5,rpn8,usp10和csn5的敲除减少了ISC/EB的数量,而CG4968和usp8的敲低增加了ISC/EB的数量。此外,ISC/EBs中usp1,CG4968,CG7857或rpn8的敲低破坏了肠屏障的完整性并缩短了寿命,表明这些DUB对维持肠道稳态的要求。此外,我们提供证据表明Usp1通过调节Notch信号活性介导ISC谱系分化。我们的研究表明,第一次,维持果蝇肠道稳态所需的去泛素酶,并为DUB和肠道稳态之间的功能联系提供新的见解。
    Deubiquitinases (DUBs) are essential for the maintenance of protein homeostasis and assembly of proteins into functional complexes. Despite growing interest in DUBs biological functions, the roles of DUBs in regulating intestinal stem cells (ISCs) and gut homeostasis remain largely unknown. Here, we perform an in vivo RNAi screen through induced knock-down of DUBs expression in adult midgut ISCs and enteroblasts (EBs) to identify DUB regulators of intestinal homeostasis in Drosophila. We screen 43 DUBs and identify 8 DUBs that are required for ISCs homeostasis. Knocking-down of usp1, CG7857, usp5, rpn8, usp10 and csn5 decreases the number of ISCs/EBs, while knocking-down of CG4968 and usp8 increases the number of ISCs/EBs. Moreover, knock-down of usp1, CG4968, CG7857, or rpn8 in ISCs/EBs disrupts the intestinal barrier integrity and shortens the lifespan, indicating the requirement of these DUBs for the maintenance of gut homeostasis. Furthermore, we provide evidences that USP1 mediates ISC lineage differentiation via modulating the Notch signaling activity. Our study identifies, for the first time, the deubiquitinases required for the maintenance of intestinal homeostasis in Drosophila, and provide new insights into the functional links between the DUBs and intestinal homeostasis.
    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
    粘膜屏障对肠道稳态至关重要,杯状细胞是维持粘膜屏障完整性所必需的。莫洛尼鼠白血病病毒1(PIM1)激酶的前病毒整合位点调节多种细胞功能,但其在结肠炎期间的肠道稳态中的作用尚不清楚。这里,我们证明,PIM1在溃疡性结肠炎患者和小鼠模型的结肠上皮中显著升高,在肠道微生物群的存在。上皮PIM1导致杯状细胞减少,从而损害小鼠对结肠炎和结肠炎相关性结直肠癌(CAC)的抵抗力。机械上,PIM1通过Wnt和Notch信号通路调节杯状细胞分化。有趣的是,PIM1与组蛋白脱乙酰酶2(HDAC2)相互作用,并通过磷酸化下调其水平,从而改变Wnt信号通路基因的表观遗传谱。总的来说,这些发现探讨了PIM1-HDAC2轴在杯状细胞分化和溃疡性结肠炎/CAC发病机制中的未知功能,这表明PIM1靶向治疗溃疡性结肠炎和CAC的潜力。
    The mucosal barrier is crucial for intestinal homeostasis, and goblet cells are essential for maintaining the mucosal barrier integrity. The proviral integration site for Moloney murine leukemia virus-1 (PIM1) kinase regulates multiple cellular functions, but its role in intestinal homeostasis during colitis is unknown. Here, we demonstrate that PIM1 is prominently elevated in the colonic epithelia of both ulcerative colitis patients and murine models, in the presence of intestinal microbiota. Epithelial PIM1 leads to decreased goblet cells, thus impairing resistance to colitis and colitis-associated colorectal cancer (CAC) in mice. Mechanistically, PIM1 modulates goblet cell differentiation through the Wnt and Notch signaling pathways. Interestingly, PIM1 interacts with histone deacetylase 2 (HDAC2) and downregulates its level via phosphorylation, thereby altering the epigenetic profiles of Wnt signaling pathway genes. Collectively, these findings investigate the unknown function of the PIM1-HDAC2 axis in goblet cell differentiation and ulcerative colitis/CAC pathogenesis, which points to the potential for PIM1-targeted therapies of ulcerative colitis and CAC.
    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
    适当的宿主-微生物群相互作用对于维持肠道稳态至关重要;因此,这些相互作用的不平衡导致炎症相关的肠道疾病。Toll样受体(TLR)识别微生物配体,并在健康和疾病中的宿主-微生物相互作用中起关键作用。TLR13在增强宿主对病原菌的防御方面具有良好的功能。然而,其在维持肠道稳态和控制结肠炎相关性结肠癌(CAC)中的作用尚不清楚.本研究旨在使用离体细胞和体内CAC动物模型研究TLR13介导的信号传导在肠道稳态和结肠肿瘤发生中的参与。Tlr13缺陷型小鼠易于发生葡聚糖硫酸钠(DSS)诱导的结肠炎。在CAC方案的早期阶段(AOM/DSS治疗),Tlr13缺乏导致严重溃疡性结肠炎。此外,Tlr13缺陷小鼠表现出增加的肠道通透性,正如异硫氰酸荧光素(FITC)-葡聚糖水平升高所证明的那样,内毒素,和细菌移位。在Tlr13缺陷小鼠中观察到结肠肠细胞的增强的细胞存活和增殖。转录组分析显示,Tlr13缺乏与结肠肿瘤组织基因表达谱的实质性变化有关。Tlr13缺陷小鼠更容易受到CAC的影响,随着白细胞介素(IL)-6,IL-12和TNF-α细胞因子的产生增加,STAT3,NF-κB增强,和结肠组织中的MAPK信号传导。这些发现表明TLR13在维持肠道稳态和控制CAC中起保护作用。我们的研究通过TLR13介导的信号传导为肠道健康提供了新的视角,这对于破译宿主-微生物群相互作用在健康和疾病中的作用至关重要。
    Appropriate host-microbiota interactions are essential for maintaining intestinal homeostasis; hence, an imbalance in these interactions leads to inflammation-associated intestinal diseases. Toll-like receptors (TLRs) recognize microbial ligands and play a key role in host-microbe interactions in health and disease. TLR13 has a well-established function in enhancing host defenses against pathogenic bacteria. However, its role in maintaining intestinal homeostasis and controlling colitis-associated colon cancer (CAC) is largely unknown. This study aimed to investigate the involvement of TLR13-mediated signaling in intestinal homeostasis and colonic tumorigenesis using ex vivo cell and in vivo CAC animal model. Tlr13-deficient mice were prone to dextran sodium sulfate (DSS)-induced colitis. During the early stages of the CAC regimen (AOM/DSS-treated), Tlr13 deficiency led to severe ulcerative colitis. Moreover, Tlr13-deficient mice exhibited increased intestinal permeability, as evidenced by elevated levels of fluorescein isothiocyanate (FITC)-dextran, endotoxins, and bacterial translocation. Enhanced cell survival and proliferation of colonic intestinal cells were observed in Tlr13-deficient mice. A transcriptome analysis revealed that Tlr13 deficiency is associated with substantial changes in gene expression profile of colonic tumor tissue. Tlr13-deficient mice were more susceptible to CAC, with increased production of interleukin (IL)-6, IL-12, and TNF-α cytokines and enhanced STAT3, NF-κB, and MAPK signaling in colon tissues. These findings suggest that TLR13 plays a protective role in maintaining intestinal homeostasis and controlling CAC. Our study provides a novel perspective on intestinal health via TLR13-mediated signaling, which is crucial for deciphering the role of host-microbiota interactions in health and disease.
    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