gut barrier function

肠屏障功能
  • 文章类型: Journal Article
    植物蛋白食品消费的增加和肉类替代品的出现促使人们对此类产品的健康益处产生了兴趣,除蛋白质外还含有纤维。这篇综述研究了纤维对植物性蛋白质代谢的影响,并评估了其对肠道健康影响的贡献。植物蛋白,通常会添加纤维,可以有不同的健康结果。加工以及纤维和淀粉的存在等因素会影响植物蛋白的消化率,可能导致肠道蛋白水解发酵增加和有害代谢物的产生。然而,可发酵纤维可以通过作为肠道微生物的主要底物来抵消这种作用,降低蛋白水解活性。纤维量的增加,而不是蛋白质来源本身,在人类研究中观察到的植物性饮食的健康益处中起着重要作用。食品基质中的外在纤维和内在纤维之间的差异进一步影响蛋白质发酵和消化率。因此,在没有天然纤维的新型蛋白质产品中,对健康的影响可能不同于传统的植物蛋白来源。各种纤维对整个胃肠道植物性蛋白质代谢的影响尚未完全了解,需要进一步研究。
    The rising consumption of plant protein foods and the emergence of meat alternatives have prompted interest in the health benefits of such products, which contain fiber in addition to protein. This review investigates the effect of fiber on plant-based protein metabolism and evaluates its contribution to gut-derived health impacts. Plant proteins, which often come with added fiber, can have varying health outcomes. Factors such as processing and the presence of fiber and starch influence the digestibility of plant proteins, potentially leading to increased proteolytic fermentation in the gut and the production of harmful metabolites. However, fermentable fiber can counteract this effect by serving as a primary substrate for gut microbes, decreasing proteolytic activity. The increased amount of fiber, rather than the protein source itself, plays a significant role in the observed health benefits of plant-based diets in human studies. Differences between extrinsic and intrinsic fiber in the food matrix further impact protein fermentation and digestibility. Thus, in novel protein products without naturally occurring fiber, the health impact may differ from conventional plant protein sources. The influence of various fibers on plant-based protein metabolism throughout the gastrointestinal tract is not fully understood, necessitating further research.
    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:酒精相关性肝病(ALD)是肝脏相关发病率和死亡率的主要原因,但目前尚无预防ALD相关肝纤维化的治疗靶点和方法。过氧化物酶体增殖物激活受体(PPAR)α和δ在脂质代谢和肠屏障稳态中起关键作用,是ALD病理进展的主要原因。同时,Elafibranor(EFN),它是PPARα和PPARδ的双重激动剂,已达到治疗代谢功能障碍相关的脂肪变性肝病和原发性胆汁性胆管炎的III期临床试验。然而,EFN对ALD治疗的益处尚不清楚.
    目的:评价EFN对ALD小鼠肝纤维化和肠屏障功能障碍的抑制作用。
    方法:通过饲喂含有2.5%乙醇(EtOH)的Lieber-DeCarli液体饮食并腹膜内注射四氯化碳,在雌性C57BL/6J小鼠中诱导ALD相关的肝纤维化每周三次(1mL/kg),持续8周。在实验期间口服施用EFN(3和10mg/kg/天)。进行组织学和分子分析以评估EFN对脂肪性肝炎的影响,纤维化,和肠道屏障的完整性。通过基于细胞的测定来评估EFN对HepG2脂毒性和Caco-2屏障功能的影响。
    结果:肝脂肪变性,凋亡,通过EFN治疗,ALD小鼠模型中的纤维化显著减轻。EFN促进EtOH刺激的HepG2细胞的脂解和β-氧化,并增强自噬和抗氧化能力,主要通过PPARα激活。此外,EFN抑制Kupffer细胞介导的炎症反应,肝暴露于脂多糖(LPS)和Toll样受体4(TLR4)/核因子κB(NF-κB)信号传导。EFN通过恢复紧密连接蛋白和自噬以及通过抑制细胞凋亡和促炎反应来改善肠道通透性。EtOH刺激的Caco-2细胞对肠屏障功能的保护作用主要由PPARδ活化介导。
    结论:EFN通过抑制脂质积累和细胞凋亡减少ALD相关纤维化,增强肝细胞自噬和抗氧化能力,并通过恢复肠屏障功能抑制LPS/TLR4/NF-κB介导的炎症反应。
    BACKGROUND: Alcohol-associated liver disease (ALD) is a leading cause of liver-related morbidity and mortality, but there are no therapeutic targets and modalities to prevent ALD-related liver fibrosis. Peroxisome proliferator activated receptor (PPAR) α and δ play a key role in lipid metabolism and intestinal barrier homeostasis, which are major contributors to the pathological progression of ALD. Meanwhile, elafibranor (EFN), which is a dual PPARα and PPARδ agonist, has reached a phase III clinical trial for the treatment of metabolic dysfunction-associated steatotic liver disease and primary biliary cholangitis. However, the benefits of EFN for ALD treatment is unknown.
    OBJECTIVE: To evaluate the inhibitory effects of EFN on liver fibrosis and gut-intestinal barrier dysfunction in an ALD mouse model.
    METHODS: ALD-related liver fibrosis was induced in female C57BL/6J mice by feeding a 2.5% ethanol (EtOH)-containing Lieber-DeCarli liquid diet and intraperitoneally injecting carbon tetrachloride thrice weekly (1 mL/kg) for 8 weeks. EFN (3 and 10 mg/kg/day) was orally administered during the experimental period. Histological and molecular analyses were performed to assess the effect of EFN on steatohepatitis, fibrosis, and intestinal barrier integrity. The EFN effects on HepG2 lipotoxicity and Caco-2 barrier function were evaluated by cell-based assays.
    RESULTS: The hepatic steatosis, apoptosis, and fibrosis in the ALD mice model were significantly attenuated by EFN treatment. EFN promoted lipolysis and β-oxidation and enhanced autophagic and antioxidant capacities in EtOH-stimulated HepG2 cells, primarily through PPARα activation. Moreover, EFN inhibited the Kupffer cell-mediated inflammatory response, with blunted hepatic exposure to lipopolysaccharide (LPS) and toll like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) signaling. EFN improved intestinal hyperpermeability by restoring tight junction proteins and autophagy and by inhibiting apoptosis and proinflammatory responses. The protective effect on intestinal barrier function in the EtOH-stimulated Caco-2 cells was predominantly mediated by PPARδ activation.
    CONCLUSIONS: EFN reduced ALD-related fibrosis by inhibiting lipid accumulation and apoptosis, enhancing hepatocyte autophagic and antioxidant capacities, and suppressing LPS/TLR4/NF-κB-mediated inflammatory responses by restoring intestinal barrier function.
    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
    不同饮食剂量的铜与果糖的相互作用通过肠-肝轴促进代谢功能障碍相关的脂肪变性肝病(MASLD)的发展。潜在的机制仍然难以捉摸。这项研究的目的是在大鼠模型中使用蛋白质组学方法鉴定导致回肠肠屏障功能障碍的特定途径。雄性断奶SpragueDawley大鼠饲喂充足铜(CuA)的饮食,边际铜(CuM),或在不存在或存在果糖补充剂的情况下补充铜(CuS)(CuAF,CuMF,和CuSF)持续4周。提取回肠蛋白并用LC-MS进行分析。鉴定了总共2847种差异表达的蛋白质(DEP),并进行了功能富集分析。因此,回肠蛋白质组和信号通路的差异改变被揭示.值得注意的是,CuAF的特征是氧化磷酸化和核糖体的富集,如KEGG分析的;CuMF的特征是富含花生四烯酸代谢途径;和粘着斑,肌动蛋白细胞骨架的调节,CuSF显著富集了紧密连接。总之,我们的蛋白质组学分析确定了回肠中与不同饮食剂量的铜-果糖相互作用相关的特定途径,这表明肠道中不同的机制参与了MASLD的发展。
    The interactions of different dietary doses of copper with fructose contribute to the development of metabolic dysfunction-associated steatotic liver disease (MASLD) via the gut-liver axis. The underlying mechanisms remain elusive. The aim of this study was to identify the specific pathways leading to gut barrier dysfunction in the ileum using a proteomics approach in a rat model. Male weanling Sprague Dawley rats were fed diets with adequate copper (CuA), marginal copper (CuM), or supplemented copper (CuS) in the absence or presence of fructose supplementation (CuAF, CuMF, and CuSF) for 4 weeks. Ileum protein was extracted and analyzed with an LC-MS. A total of 2847 differentially expressed proteins (DEPs) were identified and submitted to functional enrichment analysis. As a result, the ileum proteome and signaling pathways that were differentially altered were revealed. Of note, the CuAF is characterized by the enrichment of oxidative phosphorylation and ribosome as analyzed with the KEGG; the CuMF is characterized by an enriched arachidonic acid metabolism pathway; and focal adhesion, the regulation of the actin cytoskeleton, and tight junction were significantly enriched by the CuSF. In conclusion, our proteomics analysis identified the specific pathways in the ileum related to the different dietary doses of copper-fructose interactions, suggesting that distinct mechanisms in the gut are involved in the development of MASLD.
    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:脓毒症发病率高,发病率,和死亡率,是对人类安全的巨大威胁。肠道健康在脓毒症的发展中起着重要作用。齐黄方(QHF)含有黄芪,大黄,zhishi,还有白术.用于治疗气虚和正虚综合征。本研究旨在探讨QHF是否通过NLRP3炎性囊泡介导的细胞局灶性死亡改善小鼠肠屏障功能和微生物。
    方法:采用无特异性病原体(SPF)级C57BL/6小鼠盲肠结扎穿孔(CLP)法,中等,和高剂量的黄芪配方或益生菌4周。术后24小时,QHF缓解脓毒症肠功能障碍和恢复肠道微生态的作用机制,从而减轻肠道损伤,通过病理观察进行评估,免疫组织化学,西方印迹,ELISA,和16SrDNA高通量测序。
    结果:不同剂量的QHF和益生菌均有不同程度的改善小鼠肠道损伤和减少结肠细胞凋亡(P<0.05)。同时,不同剂量的QHF和益生菌能够降低血清IL-6,IL-1β,和TNF-α(P<0.05);下调NLRP3,caspase-1和caspase-11的蛋白表达(P<0.05);上调zonulaoccluden-1(ZO-1)和occludin的蛋白表达(P<0.05),改善小鼠肠道屏障功能。此外,QHF降低了有害细菌的相对丰度(Firmicutes,Muribaculaceae,弯曲杆菌,螺杆菌,和Alistipes),并增加了有益菌(拟杆菌和放线菌)的相对丰度(P<0.05)。
    结论:QHF通过NLRP3炎性体介导的细胞焦亡改善小鼠肠屏障功能和肠道微生物学。
    OBJECTIVE: Sepsis has a high incidence, morbidity, and mortality rate and is a great threat to human safety. Gut health plays an important role in sepsis development. Qi Huang Fang (QHF) contains astragalus, rhubarb, zhishi, and atractylodes. It is used to treat syndromes of obstructive qi and deficiency of righteousness. This study aimed to investigate whether QHF improves intestinal barrier function and microorganisms in mice through NLRP3 inflammatory vesicle-mediated cellular focal death.
    METHODS: A mouse model of sepsis was constructed by cecal ligation and puncture (CLP) of specific pathogen-free (SPF)-grade C57BL/6 mice after continuous gavage of low, medium, and high doses of astragalus formula or probiotics for 4 weeks. Twenty-four hours postoperatively, the mechanism of action of QHF in alleviating septic intestinal dysfunction and restoring intestinal microecology, thereby alleviating intestinal injury, was evaluated by pathological observation, immunohistochemistry, western blotting, ELISA, and 16S rDNA high-throughput sequencing.
    RESULTS: Different doses of QHF and probiotics ameliorated intestinal injury and reduced colonic apoptosis in mice to varying degrees (P < 0.05). Meanwhile, different doses of QHF and probiotics were able to reduce the serum levels of IL-6, IL-1β, and TNF-α (P < 0.05); down-regulate the protein expression of NLRP3, caspase-1, and caspase-11 (P < 0.05); and up-regulate the protein expression of zonula occluden-1 (ZO-1) and occludin (P < 0.05), which improved the intestinal barrier function in mice. In addition, QHF decreased the relative abundance of harmful bacteria (Firmicutes, Muribaculaceae, Campilobacterota, Helicobacter, and Alistipes) and increased the relative abundance of beneficial bacteria (Bacteroidetes and Actinobacteria) (P < 0.05).
    CONCLUSIONS: QHF improves intestinal barrier function and gut microbiology in mice via NLRP3 inflammasome-mediated cellular pyroptosis.
    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
    在这项研究中,本研究建立了一种体外共培养模型,该模型使用细胞-底物阻抗传感系统(ECIS)来测试肠道微生物群实时发酵非消化性碳水化合物(NDC)对肠道屏障功能的影响.我们将植物乳杆菌WCFS1用作模型肠道细菌,将藻酸盐-果胶用作固定化聚合物以及NDC的来源,以确定果胶发酵对T84肠上皮细胞屏障功能的影响。在第一个设计中,将植物乳杆菌WCFS1封装在藻酸盐胶囊中,然后包埋在琼脂层中,以模拟可能存在于结肠中的坚硬粘液层。在这个实验设计中,琼脂层的存在干扰了T84细胞的跨上皮电阻(TEER)测量。随后,我们去除了琼脂层,并在藻酸盐凝胶中使用包封的细菌,发现TEER测量是足够的。植物乳杆菌WCFS1的包封避免了与细胞的直接接触。此外,封装系统允许植物乳杆菌WCFS1在有限的空间中具有较高量的堆积密度,这可以限制胶囊内的氧浓度并因此产生厌氧条件。要测试此设计,将T84细胞与植物乳杆菌藻酸盐胶囊共孵育,该胶囊补充有分级负载的可发酵果胶(每个胶囊0、4和8mg/ml),以研究果胶发酵对肠屏障功能的影响。我们观察到,随着植物乳杆菌胶囊中果胶含量的增加,果胶对肠上皮的TEER表现出逐渐更强的保护作用。这可以部分解释为SCFA产量增加,因为含有8mg/ml果胶的植物乳杆菌藻酸盐胶囊中的乳酸和乙酸都得到了增强。总的来说,这种新设计的体外共培养模型可以研究细菌来源的发酵产物的影响,也可以以相对高通量的方式研究NDC对肠道屏障功能的直接影响。
    In this study, an in vitro co-culture model using an electric cell-substrate impedance sensing system (ECIS) for testing the impact of real-time fermentation of non-digestible carbohydrates (NDCs) by the intestinal microbiota on gut barrier function was established. We applied Lactobacillus plantarum WCFS1 as a model intestinal bacterium and alginate-pectin as immobilization polymers as well as a source of NDCs to determine the impact of pectin fermentation on the barrier function of T84 gut epithelial cells. In the first design, L. plantarum WCFS1 was encapsulated in an alginate capsule followed by embedding in an agar layer to mimic a firm mucus layer that might be present in the colon. In this experimental design, the presence of the agar layer interfered with the transepithelial electrical resistance (TEER) measurement of T84 cells. Subsequently, we removed the agar layer and used encapsulated bacteria in an alginate gel and found that the TEER measurement was adequate. The encapsulation of the L. plantarum WCFS1 does avoid direct contact with cells. Also, the encapsulation system allows higher amounts of packing densities of L. plantarum WCFS1 in a limited space which can limit the oxygen concentration within the capsule and therefore create anaerobic conditions. To test this design, T84 cells were co-incubated with L. plantarum alginate-capsules supplemented with graded loads of fermentable pectin (0, 4, and 8 mg/ml per capsule) to investigate the effect of pectin fermentation on gut barrier function. We observed that as the pectin content in the L. plantarum capsules increased, pectin showed a gradually stronger protective effect on the TEER of the gut epithelium. This could partly be explained by enhanced SCFA production as both lactate and acetate were enhanced in L. plantarum containing alginate capsules with 8 mg/ml pectin. Overall, this newly designed in vitro co-culture model allows for studying the impact of bacteria-derived fermentation products but also for studying the direct effects of NDCs on gut barrier function in a relatively high-throughput way.
    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
    微生物管理是水产养殖效率的核心。乳酸片球菌MA18/5M已显示出良好的促进生长的结果,调节免疫反应,和许多鱼类的抗病性。然而,这种菌株赋予鱼类健康益处的机制知之甚少,特别是在太平洋鲑鱼模型中。简而言之,这项研究的目的是i)通过检查肠道屏障功能和紧密连接(TJ)和免疫基因在体外和体内的表达来评估嗜酸乳杆菌MA18/5M的保护作用,和ii)确定该菌株对常见盐水病原体的保护作用,anguillarum弧菌J382。利用细胞系RTgutGC采用沙门氏菌肠的体外模型。通过RTgutGC中的TEER测量评估的屏障形成和完整性,显示仅暴露于GuirilarumJ38224小时的细胞的抗性显着降低,但用乳酸假单胞菌MA18/5M预处理48小时可减轻这些影响。而嗜酸假单胞菌MA18/5M并没有显著上调紧密连接和免疫分子,用这种菌株进行预处理可以防止病原体诱导的对肠道屏障的损害。特别是,Ocldn的表达被V.anguillarumJ382显着诱导,表明该分子可能在宿主对该病原体的反应中起作用。为了证实活鱼的这些观察结果,在实际水产养殖条件下饲养的奇努克鲑鱼中评估了磷酸P.MA18/5M的影响。10周后,补充嗜酸乳杆菌MA18/5M对奇努克鲑鱼的生长参数没有影响。有趣的是,组织病理学结果未显示与补充嗜酸假单胞菌MA18/5M相关的改变,表明该菌株在工业中使用是安全的。最后,在所有处理中编码TJ和免疫基因的转录本的表达模式表明,表达的变化更可能是由于发育过程而不是嗜酸乳杆菌MA18/5M补充。总的来说,我们的结果表明,嗜酸假单胞菌MA18/5M是用于鱼类生产的安全菌株,然而,为了评估以前在其他鲑鱼物种中观察到的对生长和免疫反应的影响,需要对成年鱼进行评估。
    Microbial management is central to aquaculture\'s efficiency. Pediococcus acidilactici MA18/5M has shown promising results promoting growth, modulation of the immune response, and disease resistance in many fishes. However, the mechanisms through which this strain confers health benefits in fish are poorly understood, particularly in Pacific salmonid models. Briefly, the aims of this study were to i) assess the protective effects of P. acidilactici MA18/5M by examining gut barrier function and the expression of tight junction (TJ) and immune genes in vitro and in vivo, and ii) to determine the protective effects of this strain against a common saltwater pathogen, Vibrio anguillarum J382. An in vitro model of the salmonid gut was employed utilizing the cell line RTgutGC. Barrier formation and integrity assessed by TEER measurements in RTgutGC, showed a significant decrease in resistance in cells exposed only to V. anguillarum J382 for 24 h, but pre-treatment with P. acidilactici MA18/5M for 48 h mitigated these effects. While P. acidilactici MA18/5M did not significantly upregulate tight junction and immune molecules, pre-treatment with this strain protected against pathogen-induced insults to the gut barrier. In particular, the expression of ocldn was significantly induced by V. anguillarum J382, suggesting that this molecule might play a role in the host response against this pathogen. To corroborate these observations in live fish, the effects of P. acidilactici MA18/5M was evaluated in Chinook salmon reared in real aquaculture conditions. Supplementation with P. acidilactici MA18/5M had no effect on Chinook salmon growth parameters after 10 weeks. Interestingly, histopathological results did not show alterations associated with P. acidilactici MA18/5M supplementation, indicating that this strain is safe to be used in the industry. Finally, the expression pattern of transcripts encoding TJ and immune genes in all the treatments suggest that variation in expression is more likely to be due to developmental processes rather than P. acidilactici MA18/5M supplementation. Overall, our results showed that P. acidilactici MA18/5M is a safe strain for use in fish production, however, to assess the effects on growth and immune response previously observed in other salmonid species, an assessment in adult fish is needed.
    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
    膳食纤维剥夺与益生菌灭绝有关,粘液屏障菌群失调,和粘蛋白降解细菌的过度生长。然而,粘蛋白是否以及如何挽救纤维剥夺引起的肠屏障缺陷仍未被研究。这里,我们试图研究外源性粘蛋白维持肠屏障功能的潜在作用和机制.结果表明,膳食粘蛋白减轻了纤维剥夺引起的结肠屏障完整性破坏,并减少了体内精胺的产生。重要的是,我们强调了微生物衍生的精胺生产,但不是宿主产生的精胺,粘蛋白补充后显著增加,与结肠乳酸杆菌丰度升高呈正相关。采用体外模型后,微生物来源的精胺始终由粘蛋白和乳杆菌属占主导地位。此外,粘膜Limosilactobacillus被鉴定为必需的产生精胺的乳杆菌属。,这个分离的菌株负责精胺的积累,特别是在体外粘附粘蛋白后。具体来说,通过增加精胺的产生并依赖于增强的精氨酸代谢,证实了添加粘蛋白的粘膜Limosilactobacillus的细菌上清液可促进肠屏障功能。总的来说,这些发现共同提供了证据,表明粘蛋白调节的微生物精氨酸代谢桥接了微生物和肠道屏障功能之间的相互作用,说明对宿主肠道健康的可能影响。
    目的:膳食纤维发酵产生的短链脂肪酸等微生物代谢产物已被证明对肠道健康有益。然而,必须承认某些进入结肠的氨基酸可以被微生物代谢产生多胺。多胺可以促进肠上皮细胞的更新并维持宿主-微生物的稳态。我们的研究强调了粘蛋白对促进粘膜Limosilactobacillus中精氨酸代谢产生精胺的特异性富集,表明微生物衍生的多胺支持杯状细胞增殖和屏障功能的显着增强。
    Dietary fiber deprivation is linked to probiotic extinction, mucus barrier dysbiosis, and the overgrowth of mucin-degrading bacteria. However, whether and how mucin could rescue fiber deprivation-induced intestinal barrier defects remains largely unexplored. Here, we sought to investigate the potential role and mechanism by which exogenous mucin maintains the gut barrier function. The results showed that dietary mucin alleviated fiber deprivation-induced disruption of colonic barrier integrity and reduced spermine production in vivo. Importantly, we highlighted that microbial-derived spermine production, but not host-produced spermine, increased significantly after mucin supplementation, with a positive association with upgraded colonic Lactobacillus abundance. After employing an in vitro model, the microbial-derived spermine was consistently dominated by both mucin and Lactobacillus spp. Furthermore, Limosilactobacillus mucosae was identified as an essential spermine-producing Lactobacillus spp., and this isolated strain was responsible for spermine accumulation, especially after adhering to mucin in vitro. Specifically, the mucin-supplemented bacterial supernatant of Limosilactobacillus mucosae was verified to promote intestinal barrier functions through the increased spermine production with a dependence on enhanced arginine metabolism. Overall, these findings collectively provide evidence that mucin-modulated microbial arginine metabolism bridged the interplay between microbes and gut barrier function, illustrating possible implications for host gut health.
    OBJECTIVE: Microbial metabolites like short-chain fatty acids produced by dietary fiber fermentation have been demonstrated to have beneficial effects on intestinal health. However, it is essential to acknowledge that certain amino acids entering the colon can be metabolized by microorganisms to produce polyamines. The polyamines can promote the renewal of intestinal epithelial cell and maintain host-microbe homeostasis. Our study highlighted the specific enrichment by mucin on promoting the arginine metabolism in Limosilactobacillus mucosae to produce spermine, suggesting that microbial-derived polyamines support a significant enhancement on the goblet cell proliferation and barrier function.
    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
    富含儿茶素的绿茶提取物(GTE)在肥胖啮齿动物中的抗炎活性通过降低肠道通透性和肠源性内毒素的吸收来预防代谢性内毒素血症。然而,人类健康的翻译尚未建立。我们假设与健康人相比,GTE可以通过降低代谢综合征(MetS)患者的肠道通透性以及肠道和全身性炎症来减少内毒素血症。一个随机的,双盲,安慰剂对照,在健康成年人(n=19,34±2岁)和患有MetS的成年人(n=21,40±3岁)中的交叉试验检查了对血清内毒素的脱咖啡因GTE甜食(890mg/d总儿茶素)的4周给药,肠通透性,肠道和全身炎症,和心脏代谢参数。与安慰剂相比,GTE甜食降低了健康人和MetS患者的血清内毒素(P=0.023),同时增加循环儿茶素(P<0.0001)和γ-戊内酯(P=0.0001)的浓度。无论健康状况如何,GTE均降低了粪便钙卫蛋白(P=.029)和髓过氧化物酶(P=.048)的浓度。摄入肠道通透性探针后,尿乳糖/甘露醇(P=.043),但三氯半乳蔗糖/赤藓糖醇(P>.05)不考虑健康状况。血浆转氨酶无治疗间差异(P>0.05),血压,血浆脂质,或体重,血浆肿瘤坏死因子-α,白细胞介素6或脂多糖结合蛋白/可溶性分化簇14的比例受到影响。然而,与治疗内组基线浓度相比,GTE甜食降低了两个研究组的空腹血糖(P=0.029).这些发现表明,富含儿茶素的GTE可有效减少健康成人和MetS患者的循环内毒素并改善血糖控制。可能通过减少肠道炎症和小肠通透性,但不影响全身炎症。
    Anti-inflammatory activities of catechin-rich green tea extract (GTE) in obese rodents protect against metabolic endotoxemia by decreasing intestinal permeability and absorption of gut-derived endotoxin. However, translation to human health has not been established. We hypothesized that GTE would reduce endotoxemia by decreasing gut permeability and intestinal and systemic inflammation in persons with metabolic syndrome (MetS) compared with healthy persons. A randomized, double-blind, placebo-controlled, crossover trial in healthy adults (n = 19, 34 ± 2 years) and adults with MetS (n = 21, 40 ± 3 years) examined 4-week administration of a decaffeinated GTE confection (890 mg/d total catechins) on serum endotoxin, intestinal permeability, gut and systemic inflammation, and cardiometabolic parameters. Compared with the placebo, the GTE confection decreased serum endotoxin (P = .023) in both healthy persons and those with MetS, while increasing concentrations of circulating catechins (P < .0001) and γ-valerolactones (P = .0001). Fecal calprotectin (P = .029) and myeloperoxidase (P = .048) concentrations were decreased by GTE regardless of health status. Following the ingestion of gut permeability probes, urinary lactose/mannitol (P = .043) but not sucralose/erythritol (P > .05) was decreased by GTE regardless of health status. No between-treatment differences (P > .05) were observed for plasma aminotransferases, blood pressure, plasma lipids, or body mass nor were plasma tumor necrosis factor-α, interleukin-6, or the ratio of lipopolysaccharide-binding protein/soluble cluster of differentiation-14 affected. However, fasting glucose in both study groups was decreased (P = .029) by the GTE confection compared with within-treatment arm baseline concentrations. These findings demonstrate that catechin-rich GTE is effective to decrease circulating endotoxin and improve glycemic control in healthy adults and those with MetS, likely by reducing gut inflammation and small intestinal permeability but without affecting systemic inflammation.
    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:艰难梭菌感染的治疗包括使用抗生素,免疫抑制剂,和粪便微生物移植。然而,这些治疗方法有几个缺点,包括失去对定殖的抵抗力,促进自身免疫性疾病,以及供体样本中未知病原体的可能性。迄今为止,微生物代谢物在CDI诱导的结肠炎中的潜在益处尚未得到充分研究.这里,我们首次报道,微生物代谢产物尿石素A有可能阻断艰难梭菌产生毒素,并增强肠道屏障功能,从而减轻CDI诱导的结肠炎.
    Clostridioides difficile is a Gram-positive, anaerobic, spore-forming bacterium responsible for antibiotic-associated pseudomembranous colitis. Clostridioides difficile infection (CDI) symptoms can range from diarrhea to life-threatening colon damage. Toxins produced by C. difficile (TcdA and TcdB) cause intestinal epithelial injury and lead to severe gut barrier dysfunction, stem cell damage, and impaired regeneration of the gut epithelium. Current treatment options for intestinal repair are limited. In this study, we demonstrate that treatment with the microbial metabolite urolithin A (UroA) attenuates CDI-induced adverse effects on the colon epithelium in a preclinical model of CDI-induced colitis. Moreover, our analysis suggests that UroA treatment protects against C. difficile-induced inflammation, disruption of gut barrier integrity, and intestinal tight junction proteins in the colon of CDI mice. Importantly, UroA treatment significantly reduced the expression and release of toxins from C. difficile without inducing bacterial cell death. These results indicate the direct regulatory effects of UroA on bacterial gene regulation. Overall, our findings reveal a novel aspect of UroA activity, as it appears to act at both the bacterial and host levels to protect against CDI-induced colitis pathogenesis. This research sheds light on a promising avenue for the development of novel treatments for C. difficile infection.IMPORTANCETherapy for Clostridioides difficile infections includes the use of antibiotics, immunosuppressors, and fecal microbiota transplantation. However, these treatments have several drawbacks, including the loss of colonization resistance, the promotion of autoimmune disorders, and the potential for unknown pathogens in donor samples. To date, the potential benefits of microbial metabolites in CDI-induced colitis have not been fully investigated. Here, we report for the first time that the microbial metabolite urolithin A has the potential to block toxin production from C. difficile and enhance gut barrier function to mitigate CDI-induced 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
    近视是导致远距视力模糊的最常见的眼病之一。炎症性疾病可引发或加剧近视改变。尽管肠道菌群与各种炎症性疾病有关,人们对它在近视中的作用知之甚少。
    将小鼠随机分为对照组和模型组,将模型组的30D镜片贴在眼睛上,持续3周。然后,收集小鼠盲肠内容物和血浆,分析其肠道菌群和血浆代谢组。
    我们发现近视和非近视小鼠之间的微生物组成差异很大,近视眼中Firmicutes门的相对丰度明显下降,而放线菌门的相对丰度增加。此外,放线菌和双歧杆菌与眼球的轴向长度(AL)呈正相关,而与屈光度呈负相关。非靶向代谢组学分析鉴定了141种差异表达的代谢物,和京都百科全书的基因和基因组途径富集分析揭示了相当大的富集主要在氨基酸代谢途径。值得注意的是,涉及谷氨酸代谢的途径,包括“谷氨酰胺和D-谷氨酸代谢”和“丙氨酸,天冬氨酸和谷氨酸代谢发生了巨大变化,表现为近视患者L-谷氨酸和L-谷氨酰胺浓度明显下降。有趣的是,近视中的微生物群失调和代谢产物的变化具有破坏肠道屏障的特征。我们进一步证明了近视小鼠的肠屏障功能受损,表现为Occludin的表达降低,ZO-1和增加的FITC-葡聚糖的渗透。
    与非近视小鼠相比,近视小鼠的肠道微生物组和代谢产物谱明显改变。近视的生态失调和血浆代谢组学转变具有中断的肠道屏障特征。我们的研究为肠道微生物群在近视中的可能作用提供了新的见解,并加强了基于微生物群的治疗在近视中的潜在可行性。
    UNASSIGNED: Myopia is one of the most common eye diseases leading to blurred distance vision. Inflammatory diseases could trigger or exacerbate myopic changes. Although gut microbiota bacteria are associated with various inflammatory diseases, little is known about its role in myopia.
    UNASSIGNED: The mice were randomly divided into control and model groups, with the model group being attached-30D lens onto the eyes for 3 weeks. Then, mouse cecal contents and plasma were collected to analyze their intestinal microbiota and plasma metabolome.
    UNASSIGNED: We identified that the microbial composition differed considerably between the myopic and non-myopic mice, with the relative abundance of Firmicutes phylum decreased obviously while that of Actinobacteria phylum was increased in myopia. Furthermore, Actinobacteria and Bifidobacterium were positively correlated with axial lengths (ALs) of eyeballs while negatively correlated with refractive diopters. Untargeted metabolomic analysis identified 141 differentially expressed metabolites, and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis revealed considerable enrichment mainly in amino acid metabolism pathways. Notably, pathways involved glutamate metabolism including \"Glutamine and D-glutamate metabolism\" and \"Alanine, aspartate and glutamate metabolism\" was changed dramatically, which presented as the concentrations of L-Glutamate and L-Glutamine decreased obviously in myopia. Interestingly, microbiome dysbiosis and metabolites alternations in myopia have a disrupting gut barrier feature. We further demonstrated that the gut barrier function was impaired in myopic mice manifesting in decreased expression of Occludin, ZO-1 and increased permeation of FITC-dextran.
    UNASSIGNED: Myopic mice had obviously altered gut microbiome and metabolites profiles compared to non-myopic mice. The dysbiosis and plasma metabolomics shift in myopia had an interrupting gut barrier feature. Our study provides new insights into the possible role of the gut microbiota in myopia and reinforces the potential feasibility of microbiome-based therapies in myopia.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号