intestinal epithelial cells

肠上皮细胞
  • 文章类型: Journal Article
    E3泛素连接酶(E3)通过介导泛素化在调节炎症反应中起着至关重要的作用。先前的研究表明,锚蛋白重复序列和含SOCS盒的蛋白3(ASB3)参与与癌症相关的免疫调节功能。然而,ASB3对炎症性肠病(IBD)中菌群和炎症反应的动态相互作用的影响尚不清楚.这里,我们系统地鉴定了E3连接酶ASB3作为IBD发生和进展的促进调节因子.我们观察到ASB3在IBD患者的病变中表现出明显的上调。ASB3-/-小鼠对葡聚糖硫酸钠诱导的结肠炎具有抗性。IκBα磷酸化水平与促炎因子IL-1β的产生,与WT小鼠相比,ASB3-/-小鼠的结肠组织中IL-6和TNF-α降低。这种结肠炎抗性表型在共消化微生物转移后被抑制,在联合抗生素去除肠道共生微生物组后被逆转。机械上,ASB3特异性催化肠上皮细胞中TRAF6的K48连接的多泛素化。相比之下,在缺乏ASB3的类器官中,TRAF6蛋白的完整性被屏蔽,因此减缓肠道炎症的发作。ASB3与结肠炎菌群失调有关,并通过破坏TRAF6稳定性促进促炎因子的产生。限制肠上皮细胞ASB3蛋白水平的策略可能有助于结肠炎的治疗。
    目的:泛素化是控制蛋白质稳定性的关键过程。我们确定了结肠炎症过程中肠上皮细胞中ASB3对TRAF6的泛素化。炎症性肠病患者在病灶部位表现出上调的ASB3表达,支持TRAF6降解的参与,促进TLR-Myd88/TRIF非依赖性NF-κB异常激活和肠道微生物群失衡。肠上皮细胞中持续的炎症信号传导和由ASB3介导的保护性益生菌免疫应答失调共同导致炎症性肠病的恶化。这些发现为炎症性肠病的发病机制提供了见解,并提出了ASB3增加结肠炎风险的新机制。我们的结果表明,将来在肠上皮细胞中抑制ASB3可能是一种新的临床策略。
    E3 ubiquitin ligase (E3) plays a vital role in regulating inflammatory responses by mediating ubiquitination. Previous studies have shown that ankyrin repeat and SOCS box-containing protein 3 (ASB3) is involved in immunomodulatory functions associated with cancer. However, the impact of ASB3 on the dynamic interplay of microbiota and inflammatory responses in inflammatory bowel disease (IBD) is unclear. Here, we systematically identify the E3 ligase ASB3 as a facilitative regulator in the development and progression of IBD. We observed that ASB3 exhibited significant upregulation in the lesions of patients with IBD. ASB3-/- mice are resistant to dextran sodium sulfate-induced colitis. IκBα phosphorylation levels and production of proinflammatory factors IL-1β, IL-6, and TNF-α were reduced in the colonic tissues of ASB3-/- mice compared to WT mice. This colitis-resistant phenotype was suppressed after coprophagic microbial transfer and reversed after combined antibiotics removed the gut commensal microbiome. Mechanistically, ASB3 specifically catalyzes K48-linked polyubiquitination of TRAF6 in intestinal epithelial cells. In contrast, in ASB3-deficient organoids, the integrity of the TRAF6 protein is shielded, consequently decelerating the onset of intestinal inflammation. ASB3 is associated with dysregulation of the colitis microbiota and promotes proinflammatory factors\' production by disrupting TRAF6 stability. Strategies to limit the protein level of ASB3 in intestinal epithelial cells may help in the treatment of colitis.
    OBJECTIVE: Ubiquitination is a key process that controls protein stability. We determined the ubiquitination of TRAF6 by ASB3 in intestinal epithelial cells during colonic inflammation. Inflammatory bowel disease patients exhibit upregulated ASB3 expression at focal sites, supporting the involvement of degradation of TRAF6, which promotes TLR-Myd88/TRIF-independent NF-κB aberrant activation and intestinal microbiota imbalance. Sustained inflammatory signaling in intestinal epithelial cells and dysregulated protective probiotic immune responses mediated by ASB3 collectively contribute to the exacerbation of inflammatory bowel disease. These findings provide insights into the pathogenesis of inflammatory bowel disease and suggest a novel mechanism by which ASB3 increases the risk of colitis. Our results suggest that future inhibition of ASB3 in intestinal epithelial cells may be a novel clinical strategy.
    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
    柠檬烯,一种常见于柑橘类水果和各种芳香植物中的膳食单环单萜,作为胃肠保护剂已经引起了越来越多的兴趣。这项研究旨在评估柠檬烯对肠道上皮屏障功能的影响,并探讨大麻素受体1型(CB1R)在体外的参与。此外,该研究的重点是研究柠檬烯在肠上皮细胞(Caco-2)中引起的代谢组学变化。跨上皮电阻(TEER)的初步分析表明,1-柠檬烯和d-柠檬烯,柠檬烯的异构体,导致正常细胞和由促炎细胞因子混合物(CytoMix)引起的TEER的剂量和时间依赖性增加。此外,两种柠檬烯都能降低CytoMix诱导的细胞旁通透性,路西法黄色通量的减少证明了这一点。此外,d-柠檬烯和l-柠檬烯处理增加了紧密连接分子(TJs)的表达,例如occludin,claudin-1和ZO-1,在转录和翻译水平。d-柠檬烯上调E-钙粘蛋白,参与粘附连接(AJs)的分子。机理研究表明,d-柠檬烯和l-柠檬烯处理显着抑制蛋白的CB1R,而mRNA水平保持不变。值得注意的是,d-柠檬烯对CB1R的抑制作用与药理学CB1R拮抗剂的抑制作用非常相似,如rimonabant和ORG27569。d-柠檬烯还改变Caco-2细胞代谢物。检测到β-葡萄糖和2-琥珀酰胺的大幅减少,提示柠檬烯可能影响肠上皮细胞葡萄糖摄取和谷氨酸代谢。这些发现表明,d-柠檬烯的CB1R拮抗特性可以有效地帮助恢复肠屏障损伤,标志着它是一个有前途的胃肠保护剂。
    Limonene, a dietary monocyclic monoterpene commonly found in citrus fruits and various aromatic plants, has garnered increasing interest as a gastrointestinal protectant. This study aimed to assess the effects of limonene on intestinal epithelial barrier function and investigate the involvement of cannabinoid receptor type-1 (CB1R) in vitro. Additionally, the study focused on examining the metabolomic changes induced by limonene in the intestinal epithelial cells (Caco-2). Initial analysis of transepithelial electrical resistance (TEER) revealed that both l-limonene and d-limonene, isomers of limonene, led to a dose- and time-dependent increase in TEER in normal cells and those inflamed by pro-inflammatory cytokines mixture (CytoMix). Furthermore, both types of limonene reduced CytoMix-induced paracellular permeability, as demonstrated by a decrease in Lucifer yellow flux. Moreover, d-limonene and l-limonene treatment increased the expression of tight junction molecules (TJs) such as occludin, claudin-1, and ZO-1, at both the transcriptional and translational levels. d-Limonene upregulates E-cadherin, a molecule involved in adherens junctions (AJs). Mechanistic investigations demonstrated that d-limonene and l-limonene treatment significantly inhibited CB1R at the protein, while the mRNA level remained unchanged. Notably, the inhibitory effect of d-limonene on CB1R was remarkably similar to that of pharmacological CB1R antagonists, such as rimonabant and ORG27569. d-limonene also alters Caco-2 cell metabolites. A substantial reduction in β-glucose and 2-succinamate was detected, suggesting limonene may impact intestinal epithelial cells\' glucose uptake and glutamate metabolism. These findings suggest that d-limonene\'s CB1R antagonistic property could effectively aid in the recovery of intestinal barrier damage, marking it a promising gastrointestinal protectant.
    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
    促进肠道健康,现代畜牧业中普遍存在的问题,积极影响机体健康,生产力,和经济学。猪肠上皮细胞(IPEC-J2)不断增殖以维持肠道稳态,包括屏障,免疫,和吸收功能。肠道稳态是机体健康的基础。ADP-核糖基化因子1(Arf1),一个小的GTPase,在协调mTORC1对营养素的反应中起着至关重要的作用,尤其是肠道中的氨基酸。mTORC1是扩散的中心枢纽。因此,Arf1可能促进IPEC-J2细胞增殖。然而,Arf1在猪肠道中的确切作用尚不清楚。因此,我们通过Arf1在IPEC-J2细胞中的过表达和敲低,评估了Arf1在猪肠道中的功能作用和可能的机制。Arf1过表达和敲低显著增强和抑制,分别,IPEC-J2细胞活力,PCNA表达随Arf1表达而变化。此外,Arf1过表达组Ki67阳性细胞比例显著高于对照组.这些结果表明Arf1改善IPEC-J2细胞增殖。通过Western印迹探索了潜在的机制。Arf1过表达和敲低显著增强和抑制,分别,p-S6K1和p-RPS6的水平是mTORC1信号通路的关键下游靶标。总的来说,我们的发现揭示了Arf1-mTORC1轴在IPEC-J2细胞增殖中的作用及其在调节肠道稳态和健康方面的潜在功能.
    The promotion of gut health, a pervasive problem in modern animal husbandry, positively affects organismal health, productivity, and economics. Porcine intestinal epithelial cells (IPEC-J2) continuously proliferate to maintain intestinal homeostasis, including barrier, immune, and absorptive functions. Gut homeostasis is fundamental to organismal health. ADP-ribosylation factor 1 (Arf1), a small GTPase, plays a crucial role in coordinating mTORC1 in response to nutrients, especially amino acid availability in the gut. mTORC1 is the central hub of proliferation. Thus, it seems likely that Arf1 promotes IPEC-J2 cell proliferation. However, the exact role of Arf1 in the porcine gut remains unclear. Therefore, we evaluated the functional role and possible mechanisms of Arf1 in the porcine intestine through Arf1 overexpression and knockdown in IPEC-J2 cells. Arf1 overexpression and knockdown significantly enhanced and inhibited, respectively, IPEC-J2 cell viability, and PCNA expression varied with Arf1 expression. Moreover, the proportion of Ki67-positive cells was significantly greater in the Arf1-overexpressing group than in the control group. These results suggest that Arf1 improves IPEC-J2 cell proliferation. The underlying mechanism was explored by Western blotting. Arf1 overexpression and knockdown significantly enhanced and suppressed, respectively, the levels of p-S6K1 and p-RPS6, which are key downstream targets of the mTORC1 signaling pathway. Collectively, our findings reveal the role of the Arf1-mTORC1 axis in IPEC-J2 cell proliferation and its potential function in regulating intestinal homeostasis and health.
    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
    肺炎克雷伯菌是一种机会致病菌,可通过肠道引起人粘膜病变,导致菌血症和肝脾脓肿形成。以前的研究表明,肺炎克雷伯菌可以通过肠上皮进入或穿过细胞,但机制未知.在这项研究中,我们用KP1195处理肠上皮细胞系Caco-2,这是一种临床分离的菌株,对肠上皮细胞具有高粘附性和侵袭性。结果表明,肺炎克雷伯菌处理可增加整合素基因的表达,进一步破坏细胞骨架的变化。用细胞骨架抑制剂细胞松弛素D处理Caco-2可以显着提高肺炎克雷伯菌入侵Caco-2细胞的效率。这些数据表明,通过整联蛋白破坏细胞骨架可能是肺炎克雷伯菌增加细胞内侵袭的机制之一。本研究为进一步了解肺炎克雷伯菌进入肠上皮细胞的机制提供了理论依据。
    Klebsiella pneumoniae is an opportunistic pathogen and it can cause human mucosal lesions through the intestine, leading to bacteremia and abscess formation in liver and spleen. Previous studies have shown that K. pneumoniae can enter or cross cells through the intestinal epithelium, but the mechanism is unknown. In this study, we treated the intestinal epithelial cell line Caco-2 with KP1195, a clinically isolated strain with high adhesion and invasion of intestinal epithelial cells. The results showed that the treatment of K. pneumoniae could increase the expression of integrin gene and further disrupt the changes of cytoskeleton. Treating Caco-2 with cytoskeletal inhibitor cytorelaxin D can significantly increase the efficiency of K. pneumoniae invading Caco-2 cells. These data suggest that disruption of the cytoskeleton through integrins may be one of the mechanisms by which K. pneumoniae increases intracellular invasion. This study provides a theoretical basis for further understanding of the mechanism of K. pneumoniae entering intestinal epithelial cells.
    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
    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
    据报道,肠道微生物来源的细胞外囊泡(mEV)通过将细菌产物递送到宿主细胞中来调节炎症反应。免疫球蛋白超家族巨噬细胞的补体受体(CRIgMφ)可以清除入侵的细菌及其衍生物。这里,我们研究了CRIgMφ的作用以及mEV调节肠道炎症的机制。我们发现,在IBD患者和结肠炎小鼠中,肠道微生物群受到干扰的mEVs泄漏会加剧这种情况。在肠粘膜中富集微生物DNA。IBD患者CRIg+Mφ显著降低,允许mEV扩散到粘膜中。mEV内的微生物DNA是炎症和屏障功能损伤的关键触发因素。cGAS/STING途径在mEV介导的炎症损伤中至关重要。阻断cGAS/STING信号可有效缓解由mEV渗漏和CRIg+Mφ缺乏引起的炎症。含微生物DNA的mEV,随着CRIg+Mφ缺乏,刺激IBD的炎症,与cGAS/STING途径起关键作用。
    Gut microbiota-derived extracellular vesicles (mEVs) are reported to regulate inflammatory response by delivering bacterial products into host cells. The complement receptor of the immunoglobulin superfamily macrophages (CRIg+ Mφ) could clear invading bacteria and their derivatives. Here, we investigate the role of CRIg+ Mφ and the mechanism by which mEVs regulate intestinal inflammation. We found that it is exacerbated in IBD patients and colitis mice by mEVs\' leakage from disturbed gut microbiota, enriching microbial DNA in the intestinal mucosa. CRIg+ Mφ significantly decrease in IBD patients, allowing the spread of mEVs into the mucosa. The microbial DNA within mEVs is the key trigger for inflammation and barrier function damage. The cGAS/STING pathway is crucial in mEVs-mediated inflammatory injury. Blocking cGAS/STING signaling effectively alleviates inflammation caused by mEVs leakage and CRIg+ Mφ deficiency. Microbial DNA-containing mEVs, along with CRIg+ Mφ deficiency, stimulate inflammation in IBD, with the cGAS/STING pathway playing a crucial role.
    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
    线粒体功能障碍与炎症性肠病(IBD)有关。为了了解微生物代谢回路如何导致肠道损伤,我们通过删除线粒体伴侣来破坏上皮中的线粒体功能,热休克蛋白60(Hsp60Δ/ΔIEC)。这种代谢扰动导致自我解决的组织损伤。再生在缺乏参与肠道稳态或炎症调节剂白细胞介素(IL)-10(Hsp60Δ/ΔIEC;Il10-/-)的芳香烃受体(Hsp60Δ/ΔIEC;AhR-/-)的情况下被破坏,导致IBD样病理。无菌(GF)Hsp60Δ/ΔIEC小鼠的远端结肠不存在损伤,强调细菌对代谢损伤的控制。用合成群落OMM12定殖GFHsp60Δ/ΔIEC小鼠揭示了代谢灵活的拟杆菌的扩张,盲肠杆菌单定植概括了损伤。代谢受损上皮的转录谱分析揭示了参与氧化应激的基因特征(Ido1,Nos2,Duox2)。这些特征在克罗恩病患者的样本中观察到,区分活性和非活性炎症。因此,上皮的线粒体扰动会导致微生物群依赖性损伤,并具有与IBD相关的区分性炎症基因谱。
    Mitochondrial dysfunction is associated with inflammatory bowel diseases (IBDs). To understand how microbial-metabolic circuits contribute to intestinal injury, we disrupt mitochondrial function in the epithelium by deleting the mitochondrial chaperone, heat shock protein 60 (Hsp60Δ/ΔIEC). This metabolic perturbation causes self-resolving tissue injury. Regeneration is disrupted in the absence of the aryl hydrocarbon receptor (Hsp60Δ/ΔIEC;AhR-/-) involved in intestinal homeostasis or inflammatory regulator interleukin (IL)-10 (Hsp60Δ/ΔIEC;Il10-/-), causing IBD-like pathology. Injury is absent in the distal colon of germ-free (GF) Hsp60Δ/ΔIEC mice, highlighting bacterial control of metabolic injury. Colonizing GF Hsp60Δ/ΔIEC mice with the synthetic community OMM12 reveals expansion of metabolically flexible Bacteroides, and B. caecimuris mono-colonization recapitulates the injury. Transcriptional profiling of the metabolically impaired epithelium reveals gene signatures involved in oxidative stress (Ido1, Nos2, Duox2). These signatures are observed in samples from Crohn\'s disease patients, distinguishing active from inactive inflammation. Thus, mitochondrial perturbation of the epithelium causes microbiota-dependent injury with discriminative inflammatory gene profiles relevant for 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
    最近的进展使体外模拟人类小肠成为可能,但要完全概括它们的结构和功能特征仍然是一个挑战。我们怀疑肠道内的间质流,在胚胎器官发生过程中由循环血浆提供动力,成为一个重要因素。我们旨在通过将间质流纳入系统来构建体内多层小肠组织,反过来,通过在能够复制间质流的微流体装置上同时区分定形内胚层和中胚层细胞与人多能干细胞,开发了小肠系统。这种方法增强了细胞成熟,并导致了具有绒毛样上皮和对齐的间充质层的三维小肠样组织的发育。我们的小肠系统不仅克服了传统肠道模型的局限性,而且还提供了一个独特的机会来深入了解肠道组织发育的详细机制。
    Recent advances have made modeling human small intestines in vitro possible, but it remains a challenge to recapitulate fully their structural and functional characteristics. We suspected interstitial flow within the intestine, powered by circulating blood plasma during embryonic organogenesis, to be a vital factor. We aimed to construct an in vivo-like multilayered small intestinal tissue by incorporating interstitial flow into the system and, in turn, developed the micro-small intestine system by differentiating definitive endoderm and mesoderm cells from human pluripotent stem cells simultaneously on a microfluidic device capable of replicating interstitial flow. This approach enhanced cell maturation and led to the development of a three-dimensional small intestine-like tissue with villi-like epithelium and an aligned mesenchymal layer. Our micro-small intestine system not only overcomes the limitations of conventional intestine models but also offers a unique opportunity to gain insights into the detailed mechanisms underlying intestinal tissue development.
    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
    细胞培养是生物和医学研究中用于理解生理和病理过程的模型。根据细胞类型和所需物种,细胞系并不总是可用的。此外,永生化过程经常影响细胞生物学。原代细胞通常在短期培养中与组织中的细胞保持更大程度的相似性。这项研究的目的是验证鸡原代上皮盲肠细胞(PECCs)在宿主病原体相互作用的体外研究中的适用性。通过检测紧密和粘附连接以及鹅卵石样细胞形态来确认PECs的上皮性质。唾液酸的分布与盲肠膀胱切开术相似。为了了解PECC应对微生物挑战的能力,确定了Toll样受体(TLRs)库。PECC暴露于聚肌苷酸-聚胞嘧啶酸(聚(I:C))或脂多糖(LPS)导致I型和III型干扰素(IFN)以及白介素(IL-)1β的上调,IL-6和IL-8mRNA表达。总的来说,PECC显示出极化上皮细胞的特性。TLRs的存在,它们的差异表达,以及模式识别受体依赖性免疫反应使PECCs能够作为宿主病原体相互作用研究的合适体外模型,在体内条件下很难进行。
    Cell cultures are models in biological and medical research to understand physiological and pathological processes. Cell lines are not always available depending on cell type and required species. In addition, the immortalization process often affects cell biology. Primary cells generally maintain a greater degree of similarity in short-term culture to the cells in tissue. Goal of this study was to verify the suitability of chicken primary epithelial caecal cells (PECCs) for in vitro investigations of host‒pathogen interactions. Epithelial nature of PECCs was confirmed by detection of tight and adherens junctions and cobblestone-like cell morphology. Sialic acids distribution was similar to that in caecal cyrosections. To understand the capacity of PECCs to respond to microbial challenges, the Toll-like receptors (TLRs) repertoire was determined. Exposure of PECCs to polyinosinic-polycytidylic acid (poly(I:C)) or lipopolysaccharide (LPS) led to upregulation of type I and III interferon (IFN) as well as interleukin (IL-) 1β, IL-6 and IL-8 mRNA expression. Overall, the PECCs showed properties of polarized epithelial cells. The presence of TLRs, their differential expression, as well as pattern recognition receptor dependent immune responses enable PECCs to act as suitable in vitro model for host‒pathogen interaction studies, which are difficult to conduct under in vivo conditions.
    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
    溃疡性结肠炎(UC)是一种以肠上皮损害为特征的主要炎症性肠病(IBD)。最近,脂质运载蛋白-2(LCN2)已被确定为UC患者的潜在粪便生物标志物。然而,需要进一步研究以探讨其在UC中的促炎作用及其潜在机制.生物学分析表明,Lcn2在UC患者的结肠粘膜中充当推定的特征基因,并与UC的capsase/pyrotocol信号通路有关。在患有DSS诱导的结肠炎的野生型小鼠中,通过体内施用Lcn2过表达质粒在结肠粘膜中的LCN2过表达导致结肠炎症状加重和上皮损伤,以及增加的表达水平的焦亡标志物(裂解的caspase1,GSDMD,IL-1β,HMGB1和IL-18)。此外,我们观察到LCN2体内沉默后,焦凋亡标志物表达水平的下调。然而,LCN2过表达对GSDMD-KO小鼠的促炎作用有明显的抑制作用。此外,单细胞RNA测序分析显示,Lcn2主要在UC患者结肠粘膜的肠上皮细胞(IECs)中表达.我们发现,LCN2通过调节LPS和ATP刺激的NCM460细胞中NF-κB/NLRP3/GSDMD信号轴,有效地调节了细胞焦亡事件。这些发现证明了LCN2在结肠上皮中的促炎作用,并为抑制UC中的焦亡提供了潜在的靶标。
    Ulcerative colitis (UC) is a major inflammatory bowel disease (IBD) characterized by intestinal epithelium damage. Recently, Lipocalin-2 (LCN2) has been identified as a potential fecal biomarker for patients with UC. However, further investigation is required to explore its pro-inflammatory role in UC and the underlying mechanism. The biological analysis revealed that Lcn2 serves as a putative signature gene in the colon mucosa of patients with UC and its association with the capsase/pyroptosis signaling pathway in UC. In wild-type mice with DSS-induced colitis, LCN2 overexpression in colon mucosa via in vivo administration of Lcn2 overexpression plasmid resulted in exacerbation of colitis symptoms and epithelium damage, as well as increased expression levels of pyroptosis markers (cleaved caspase1, GSDMD, IL-1β, HMGB1 and IL-18). Additionally, we observed downregulation in the expression levels of pyroptosis markers following in vivo silencing of LCN2. However, the pro-inflammatory effect of LCN2 overexpression was effectively restrained in GSDMD-KO mice. Moreover, single-cell RNA-sequencing analysis revealed that Lcn2 was predominantly expressed in the intestinal epithelial cells (IECs) within the colon mucosa of patients with UC. We found that LCN2 effectively regulated pyroptosis events by modulating the NF-κB/NLRP3/GSDMD signaling axis in NCM460 cells stimulated by LPS and ATP. These findings demonstrate the pro-inflammatory role of LCN2 in colon epithelium and provide a potential target for inhibiting pyroptosis in UC.
    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