intestinal stem cell

肠干细胞
  • 文章类型: Journal Article
    已知高脂肪饮食(HFD)喂养会导致肠道屏障破坏,从而引发严重的肠道炎症性疾病。吲哚-3-醛(IAld)已成为减轻炎症反应和维持肠道稳态的潜在候选者。然而,ILD在HFD相关肠道破坏中的作用尚不清楚.在这项研究中,将48只7周龄雄性C57BL/6J小鼠分为四组:正常饮食(NCD)组接受NCD;HFD组喂养HFD;HFDIAld200组在HFD中补充了200mg/kgIAld;HFDIAld600组在HFD中补充了600mg/kgIAld。结果表明,膳食补充ILD可以改善脂肪积累和代谢紊乱,与肠道通透性降低有关。这种减少潜在地导致HFD喂养的小鼠中全身性炎症减少和肠屏障功能增强。此外,我们发现ILD通过体内和离体激活芳香烃受体(AHRs)促进肠干细胞(ISC)的增殖。这些发现表明Ild通过促进AHR介导的ISC增殖来恢复HFD诱导的肠屏障破坏。
    High-fat diet (HFD) feeding is known to cause intestinal barrier disruption, thereby triggering severe intestinal inflammatory disease. Indole-3-aldehyde (IAld) has emerged as a potential candidate for mitigating inflammatory responses and maintaining intestinal homeostasis. However, the role of IAld in the HFD-related intestinal disruption remains unclear. In this study, 48 7 week-old male C57BL/6J mice were assigned to four groups: the normal chow diet (NCD) group received a NCD; the HFD group was fed an HFD; the HFD + IAld200 group was supplemented with 200 mg/kg IAld in the HFD; and the HFD + IAld600 group was supplemented with 600 mg/kg IAld in the HFD. The results showed that dietary IAld supplementation ameliorated fat accumulation and metabolic disorders, which are associated with reduced intestinal permeability. This reduction potentially led to decreased systemic inflammation and enhanced intestinal barrier function in HFD-fed mice. Furthermore, we found that IAld promoted intestinal stem cell (ISC) proliferation by activating aryl hydrocarbon receptors (AHRs) in vivo and ex vivo. These findings suggest that IAld restores the HFD-induced intestinal barrier disruption by promoting AHR-mediated ISC proliferation.
    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:凉血固原益肾汤的疗效,中药,在高剂量辐射暴露后,已在治疗辐射引起的肠损伤(RIII)和保持肠完整性和功能方面得到了科学证明。然而,需要进一步的研究来全面阐明LGYD治疗效果的确切机制,从而为辐射防护提供潜在的药物选择.
    目的:本研究旨在阐明LGYD通过调节肠道菌群(GM)对RIII发挥治疗作用的潜在机制。
    方法:采用16srRNA分析来评估不同剂量的全身照射(WBI)对GM的影响,以建立适合本研究的模型。使用16srRNA和SCFA定量评价LGYD对GM和SCFA的作用。利用UHPLC-QE-MS鉴定LGYD以及含LGYD药物的血清(LGYD-DS)中的活性组分。随后,免疫荧光和免疫组织化学染色用于验证LGYD和/或特征性微生物群对体内RIII恢复的影响.LGYD-DS的影响,特征植物区系,通过测量肠道类器官模型中的类器官表面积来评估肠干细胞(ISC)和SCFA。
    结果:剂量为8.5Gy的全身照射对GM的物种组成和丰度有显著影响,用作体内模型。LGYD显著提高存活率并促进从RIII的恢复。此外,LGYD表现出显着增加的Akkermansiamuciniphia(AKK)的丰度和SCFA的水平,特别是异丁酸。LGYD-DS由来自LGYD草药的七个主要成分组成。体内实验表明,LGYD和AKK均大大提高了辐射后的存活率,并促进了肠结构和功能的恢复过程。在类器官模型中,用LGYD-DS治疗,AKK上清液或异丁酸显著增加类器官表面积。
    结论:LGYD有可能通过促进肠干细胞的恢复来增强RIII,这与AKK丰度的上调和SCFA的产生密切相关,特别是异丁酸。
    BACKGROUND: The efficacy of Liangxue Guyuan Yishen Decoction (LGYD), a traditional Chinese medicine, has been scientifically proven in the treatment of radiation-induced intestinal injury (RIII) and preservation of intestinal integrity and function following high-dose radiation exposure. However, further investigation is required to comprehensively elucidate the precise mechanisms underlying the therapeutic effects of LGYD in order to provide potential pharmaceutical options for radiation protection.
    OBJECTIVE: This study aims to elucidate the potential mechanism through which LGYD exerts its therapeutic effects on RIII by modulating the gut microbiota (GM).
    METHODS: 16 s rRNA analysis was employed to assess the impact of varying doses of whole body irradiation (WBI) on GM in order to establish an appropriate model for this study. The effects of LGYD on GM and SCFA were evaluated using 16 s rRNA and Quantification of SCFA. UHPLC-QE-MS was utilized to identify the active components in LGYD as well as LGYD drug containing serum (LGYD-DS). Subsequently, immunofluorescence and immunohistochemical staining were conducted to validate the influence of LGYD and/or characteristic microbiota on RIII recovery in vivo. The effects of LGYD-DS, characteristic flora, and SCFA on intestinal stem cell (ISC) were assessed by measuring organoid surface area in intestinal organoid model.
    RESULTS: The species composition and abundance of GM were significantly influenced by whole-body irradiation with a dose of 8.5 Gy, which was used as in vivo model. LGYD significantly improves the survival rate and promotes recovery from RIII. Additionally, LGYD exhibited a notable increase in the abundance of Akkermansia muciniphila (AKK) and levels of SCFA, particularly isobutyric acid. LGYD-DS consisted of seven main components derived from herbs of LGYD. In vivo experiments indicated that both LGYD and AKK substantially enhanced the survival rate after radiation and facilitated the recovery process for intestinal structure and function. In the organoid model, treatment with LGYD-DS, AKK supernatant or isobutyric acid significantly increased organoid surface area.
    CONCLUSIONS: LGYD has the potential to enhance RIII by promoting the restoration of intestinal stem cell, which is closely associated with the upregulation of AKK abundance and production of SCFA, particularly isobutyric acid.
    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
    过氧化物酶体动力学对肠干细胞(ISC)分化和肠再生至关重要。然而,在肠道再生过程中,控制ISC内过氧化物酶体动力学的确切机制尚不清楚.使用小鼠结肠炎和果蝇肠模型,我们已经确定了涉及转录因子过氧化物酶体增殖物激活受体(PPARs)和SOX21的负反馈控制机制.这种反馈机制有效地调节肠道再生过程中的过氧化物酶体丰度。肠道损伤后,释放的游离极长链脂肪酸(VLCFAs)通过刺激PPARs-PEX11s信号传导增加过氧化物酶体丰度。PPARs具有刺激过氧化物酶体裂变和抑制嗜血杆菌的作用。SOX21在ISC分化过程中作用于过氧化物酶体的下游,通过自噬诱导过氧化物酶体消除,同时抑制PPAR表达。因此,PPAR和SOX21构成了调节过氧化物酶体动力学的精细调节的负反馈回路。这些发现揭示了ISC中过氧化物酶体调节的复杂分子机制,有助于我们对肠道更新和修复的理解。
    Peroxisome dynamics are crucial for intestinal stem cell (ISC) differentiation and gut regeneration. However, the precise mechanisms that govern peroxisome dynamics within ISCs during gut regeneration remain unknown. Using mouse colitis and Drosophila intestine models, we have identified a negative-feedback control mechanism involving the transcription factors peroxisome proliferator-activated receptors (PPARs) and SOX21. This feedback mechanism effectively regulates peroxisome abundance during gut regeneration. Following gut injury, the released free very long-chain fatty acids (VLCFAs) increase peroxisome abundance by stimulating PPARs-PEX11s signaling. PPARs act to stimulate peroxisome fission and inhibit pexophagy. SOX21, which acts downstream of peroxisomes during ISC differentiation, induces peroxisome elimination through pexophagy while repressing PPAR expression. Hence, PPARs and SOX21 constitute a finely tuned negative-feedback loop that regulates peroxisome dynamics. These findings shed light on the complex molecular mechanisms underlying peroxisome regulation in ISCs, contributing to our understanding of gut renewal and 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
    肠上皮动态控制细胞周期,然而,目前尚无直接分析非永生化人肠上皮细胞(IECs)细胞周期时相的实验平台.这里,我们提供了两个记者和一个完整的平台,用于分析活的原代人IEC的细胞周期阶段。我们询问在软胶原蛋白上生长的IECs的转录身份,开发两个荧光细胞周期报告IEC细胞系,设计和3D打印胶原蛋白压机,以制作腔室载玻片,以实现最佳成像,同时支持原发性人类IEC生长,实时图像细胞周期动力学,然后在免费使用的程序上组装一个计算管道,用于细胞周期阶段的半自动分析。PIP-FUCCI构建体允许从活细胞的单个图像中分配细胞周期阶段,我们的PIP-H2A构建体允许使用我们公开的计算管道对细胞周期阶段长度进行半自动直接定量。用寡霉素治疗PIP-FUCCIIEC表明抑制线粒体呼吸延长G1期,和PIP-H2A细胞使我们能够测量寡霉素在异质IEC中差异延长S和G2/M期。这些平台为未来研究对肠上皮的药物作用提供了机会,细胞周期调节,还有更多.
    The intestinal epithelium dynamically controls cell cycle, yet no experimental platform exists for directly analyzing cell cycle phases in non-immortalized human intestinal epithelial cells (IECs). Here, we present two reporters and a complete platform for analyzing cell cycle phases in live primary human IECs. We interrogate the transcriptional identity of IECs grown on soft collagen, develop two fluorescent cell cycle reporter IEC lines, design and 3D print a collagen press to make chamber slides for optimal imaging while supporting primary human IEC growth, live image cell cycle dynamics, then assemble a computational pipeline building upon free-to-use programs for semi-automated analysis of cell cycle phases. The PIP-FUCCI construct allows for assigning cell cycle phase from a single image of living cells, and our PIP-H2A construct allows for semi-automated direct quantification of cell cycle phase lengths using our publicly available computational pipeline. Treating PIP-FUCCI IECs with oligomycin demonstrates that inhibiting mitochondrial respiration lengthens G1 phase, and PIP-H2A cells allow us to measure that oligomycin differentially lengthens S and G2/M phases across heterogeneous IECs. These platforms provide opportunities for future studies on pharmaceutical effects on the intestinal epithelium, cell cycle regulation, and more.
    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
    遗传瘦和肥胖个体具有不同的肠道微生物群和功能。然而,微生物组异质性的潜在机制及其对上皮功能如肠干细胞(ISC)命运的调节仍不清楚。使用遗传上不同品种的猪(肥胖的梅山和瘦的约克郡),这项研究揭示了空肠微生物生态的转录组范围的变化,以富集活性乳酸菌为特征,特别是主要的淀粉乳杆菌(L.淀粉样蛋白),和肥胖品种的乳酸代谢网络。如高表达的GPR81,更多的增殖性ISC和激活的Wnt/β-catenin信号所反映的,淀粉样乳杆菌显性异质性与上皮功能差异平行。使用内部开发的猪空肠类器官的实验证明,活的淀粉样乳杆菌及其代谢产物乳酸可促进肠道类器官的生长。机械上,淀粉乳杆菌和乳酸盐以GPR81依赖性方式激活Wnt/β-连环蛋白信号以促进ISC介导的上皮增殖。然而,热抑制淀粉样乳杆菌不能引起这些变化。这些发现揭示了淀粉样乳杆菌在通过乳杆菌-乳酸-GPR81轴调节空肠干细胞中以前代表性不足的作用,连接品种驱动的肠道微生物组异质性与ISC命运的关键机制。因此,这项研究的结果为肠道微生物组和干细胞相互作用在维持肠道稳态中的作用提供了新的见解。
    Genetically lean and obese individuals have distinct intestinal microbiota and function. However, the underlying mechanisms of the microbiome heterogeneity and its regulation on epithelial function such as intestinal stem cell (ISC) fate remain unclear. Employing pigs of genetically distinct breeds (obese Meishan and lean Yorkshire), this study reveals transcriptome-wide variations in microbial ecology of the jejunum, characterized by enrichment of active Lactobacillus species, notably the predominant Lactobacillus amylovorus (L. amylovorus), and lactate metabolism network in obese breeds. The L. amylovorus-dominant heterogeneity is paralleled with epithelial functionality difference as reflected by highly expressed GPR81, more proliferative ISCs and activated Wnt/β-catenin signaling. Experiments using in-house developed porcine jejunal organoids prove that live L. amylovorus and its metabolite lactate promote intestinal organoid growth. Mechanistically, L. amylovorus and lactate activate Wnt/β-catenin signaling in a GPR81-dependent manner to promote ISC-mediated epithelial proliferation. However, heat-killed L. amylovorus fail to cause these changes. These findings uncover a previously underrepresented role of L. amylovorus in regulating jejunal stem cells via Lactobacillus-lactate-GPR81 axis, a key mechanism bridging breed-driven intestinal microbiome heterogeneity with ISC fate. Thus, results from this study provide new insights into the role of gut microbiome and stem cell interactions in maintaining 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
    肠上皮经历快速细胞更新,依靠当地的利基市场,以支持肠干细胞(ISC)的功能和自我更新。对ISC与疾病之间关联的研究继续以快速的速度扩展。然而,ISC与肠道微生物的详细相互作用尚待阐明.因此,这篇综述见证了ISC和肠道微生物之间的串扰方面的重大进展,提供关键见解(1)ISC生态位的构建和如何在Wnt的参与下共同控制上皮内稳态和预防肠道疾病的分子机制,骨形态发生蛋白,和Notch;(2)ISC的分化命运影响肠道微生物群。同时,肠道微生物的存在也调节ISC的功能;(3)通过模式识别受体的Wnt和Notch信号对ISC的微生物群调节;(4)特定的微生物群相关的postbiotics如何影响ISC以维持肠上皮再生和稳态,从而为肠道疾病的替代治疗方法提供见解。考虑到详细的互动还不清楚,有必要进一步探讨肠道菌群对ISC的调节作用,以利用微生物缓解肠道疾病。此外,这些重大进展共同推动我们在再生医学和临床微生物移植治疗癌症方面取得突破。
    Intestinal epithelium undergoes rapid cellular turnover, relying on the local niche, to support intestinal stem cells (ISCs) function and self-renewal. Research into the association between ISCs and disease continues to expand at a rapid rate. However, the detailed interaction of ISCs and gut microbes remains to be elucidated. Thus, this review witnessed major advances in the crosstalk between ISCs and gut microbes, delivering key insights into (1) construction of ISC niche and molecular mechanism of how to jointly govern epithelial homeostasis and protect against intestinal diseases with the participation of Wnt, bone morphogenetic protein, and Notch; (2) differentiation fate of ISCs affect the gut microbiota. Meanwhile, the presence of intestinal microbes also regulates ISC function; (3) microbiota regulation on ISCs by Wnt and Notch signals through pattern recognition receptors; (4) how do specific microbiota-related postbiotics influence ISCs to maintain intestinal epithelial regeneration and homeostasis that provide insights into a promising alternative therapeutic method for intestinal diseases. Considering the detailed interaction is still unclear, it is necessary to further explore the regulatory role of gut microbiota on ISCs to utilize microbes to alleviate gut disorders. Furthermore, these major advances collectively drive us ever closer to breakthroughs in regenerative medicine and cancer treatment by microbial transplantation in the clinic.
    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
    宿主-微生物相互作用影响肠干细胞(ISC)活性以调节上皮更新和组成。这里,我们研究了病毒感染对肠道稳态的功能影响,以及病毒感染改变ISC活性的机制.我们报道果蝇A病毒(DAV)感染通过诱导持续的ISC增殖来破坏果蝇的肠道稳态,导致肠道发育不良,肠道屏障功能的丧失,减少寿命。我们发现实验室饲养的果蝇中常见的其他病毒也促进ISC增殖。DAV诱导的ISC增殖的机制涉及祖细胞自主表皮生长因子受体(EGFR)信号,肠上皮细胞c-Jun氨基末端激酶(JNK)活性,并且需要Sting依赖性核因子κB(NF-κB)(Relish)活性。我们进一步证明激活Sting-Relish信号足以诱导ISC增殖,促进肠道发育不良,在没有感染的情况下缩短寿命。我们的结果表明,病毒感染可以显著破坏肠道生理,突出了Sting-Relish信号的一个新角色,并支持病毒感染在衰老中的作用。
    Host-microbe interactions influence intestinal stem cell (ISC) activity to modulate epithelial turnover and composition. Here, we investigated the functional impacts of viral infection on intestinal homeostasis and the mechanisms by which viral infection alters ISC activity. We report that Drosophila A virus (DAV) infection disrupts intestinal homeostasis in Drosophila by inducing sustained ISC proliferation, resulting in intestinal dysplasia, loss of gut barrier function, and reduced lifespan. We found that additional viruses common in laboratory-reared Drosophila also promote ISC proliferation. The mechanism of DAV-induced ISC proliferation involves progenitor-autonomous epidermal growth factor receptor (EGFR) signaling, c-Jun N-terminal kinase (JNK) activity in enterocytes, and requires Sting-dependent nuclear factor κB (NF-κB) (Relish) activity. We further demonstrate that activating Sting-Relish signaling is sufficient to induce ISC proliferation, promote intestinal dysplasia, and reduce lifespan in the absence of infection. Our results reveal that viral infection can significantly disrupt intestinal physiology, highlight a novel role for Sting-Relish signaling, and support a role for viral infection in aging.
    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
    先于恶性肿瘤的自发性肿瘤发生的早期机制在很大程度上是未知的。我们表明,降低的aPKC水平与干细胞损失和复苏的诱导和化生程序相关的serrate-和常规启动的癌前病变,在结直肠癌(CRC)中长期存在。体内和小鼠类器官中PKCλ/1的急性失活足以刺激非转化肠上皮细胞(IECs)中的JNK,促进细胞死亡和肠干细胞(ISC)的快速损失,包括LGR5+。随后是隐窝底部的复兴干细胞(RSC)和顶部的胎儿化生细胞(FMC)的积累,创建两个时空不同的细胞群体,依赖于JNK诱导的AP-1和YAP。这些细胞谱系变化在癌症发生和发展过程中得以维持,并决定了人类CRC的侵袭性表型。不管它们的锯齿或传统来源。
    The early mechanisms of spontaneous tumor initiation that precede malignancy are largely unknown. We show that reduced aPKC levels correlate with stem cell loss and the induction of revival and metaplastic programs in serrated- and conventional-initiated premalignant lesions, which is perpetuated in colorectal cancers (CRCs). Acute inactivation of PKCλ/ι in vivo and in mouse organoids is sufficient to stimulate JNK in non-transformed intestinal epithelial cells (IECs), which promotes cell death and the rapid loss of the intestinal stem cells (ISCs), including those that are LGR5+. This is followed by the accumulation of revival stem cells (RSCs) at the bottom of the crypt and fetal-metaplastic cells (FMCs) at the top, creating two spatiotemporally distinct cell populations that depend on JNK-induced AP-1 and YAP. These cell lineage changes are maintained during cancer initiation and progression and determine the aggressive phenotype of human CRC, irrespective of their serrated or conventional origin.
    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
    肠干细胞(ISC)对于维持和再生肠上皮至关重要。小檗碱(BBR)表现出多样化的生物活性,但目前尚不清楚BBR是否可以调节ISC的功能。因此,我们研究了BBR对健康和辐射损伤小鼠ISC的影响,并探讨了潜在的潜在机制。结果表明,BBR显著增加小肠的长度,绒毛的高度,以及地穴的深度和密度,促进隐窝上皮细胞的增殖,增加OLFM4+ISSC和杯状细胞的数量。来自BBR处理的小鼠的隐窝比未处理的小鼠的隐窝更能够生长为肠状肠。BBR减轻WAI诱导的肠损伤。BBR抑制隐窝上皮细胞凋亡,增加杯状细胞的数量,并在8GyWAI诱导的损伤后增加了OLFM4ISC和tdTomatoISC后代的数量。机械上,在生理条件下和WAI诱导的损伤后,BBR处理导致p-S6,p-STAT3和p-ERK1/2阳性的密码上皮细胞数量显着增加。总之,BBR能够在生理上或在辐射诱导的损伤后增强ISC的功能,表明BBR在治疗放射性肠损伤方面具有潜在价值。
    Intestinal stem cells (ISCs) are pivotal for the maintenance and regeneration of the intestinal epithelium. Berberine (BBR) exhibits diverse biological activities, but it remains unclear whether BBR can modulate ISCs\' function. Therefore, we investigated the effects of BBR on ISCs in healthy and radiation-injured mice and explored the potential underlying mechanisms involved. The results showed that BBR significantly increased the length of the small intestines, the height of the villi, and the depth and density of the crypts, promoted the proliferation of cryptal epithelial cells and increased the number of OLFM4+ ISCs and goblet cells. Crypts from the BBR-treated mice were more capable of growing into enteroids than those from untreated mice. BBR alleviated WAI-induced intestinal injury. BBR suppressed the apoptosis of crypt epithelial cells, increased the quantity of goblet cells, and increased the quantity of OLFM4+ ISCs and tdTomato+ progenies of ISCs after 8 Gy WAI-induced injury. Mechanistically, BBR treatment caused a significant increase in the quantity of p-S6, p-STAT3 and p-ERK1/2 positive cryptal epithelial cells under physiological conditions and after WAI-induced injury. In conclusion, BBR is capable of enhancing the function of ISCs either physiologically or after radiation-induced injury, indicating that BBR has potential value in the treatment of radiation-induced intestinal injury.
    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:五粮液浓香白酒(以下,五粮液白酒)是中国传统的谷物白酒,含有短链脂肪酸,己酸乙酯,乳酸乙酯,其他微量成分,和很大比例的乙醇。五粮液白酒对肠干细胞和肠上皮发育的影响尚未阐明。这里,通过在小鼠模型中施用Lieber-DeCarli慢性乙醇液体饮食,以模拟健康成年人的长期(8周)轻/中度饮酒(1.6gkg-1day-1),研究了五粮液白酒在肠上皮再生和肠道微生物群调节中的作用。
    结果:五粮液白酒促进小鼠结肠隐窝增殖。根据免疫荧光和逆转录定量聚合酶链反应分析,与仅使用乙醇的治疗相比,五粮液白酒增加了小鼠结肠中肠干细胞和杯状细胞的数量以及肠内分泌细胞分化标志物的表达。此外,肠道菌群分析显示,五粮液白酒给药后,与肠道稳态相关的菌群相对丰度增加。值得注意的是,细菌的丰度增加,Faecalibaculum,落叶松科,Blautia可能在促进干细胞介导的肠上皮发育和维持肠稳态中起重要作用。
    结论:总之,这些发现表明,五粮液白酒可用于调节小鼠肠道干细胞的增殖和分化,并改变肠道菌群分布,从而促进肠道稳态。本研究阐明了五粮液白酒促进肠道健康的机制。©2024化学工业学会。
    BACKGROUND: Wuliangye strong aroma baijiu (hereafter, Wuliangye baijiu) is a traditional Chinese grain liquor containing short-chain fatty acids, ethyl caproate, ethyl lactate, other trace components, and a large proportion of ethanol. The effects of Wuliangye baijiu on intestinal stem cells and intestinal epithelial development have not been elucidated. Here, the role of Wuliangye baijiu in intestinal epithelial regeneration and gut microbiota modulation was investigated by administering a Lieber-DeCarli chronic ethanol liquid diet in a mouse model to mimic long-term (8 weeks\') light/moderate alcohol consumption (1.6 g kg-1 day-1) in healthy human adults.
    RESULTS: Wuliangye baijiu promoted colonic crypt proliferation in mice. According to immunofluorescence and reverse transcription-quantitative polymerase chain reaction analyses, compared with the ethanol-only treatment, Wuliangye baijiu increased the number of intestinal stem cells and goblet cells and the expression of enteroendocrine cell differentiation markers in the mouse colon. Furthermore, gut microbiota analysis showed an increase in the relative abundance of microbiota related to intestinal homeostasis following Wuliangye baijiu administration. Notably, increased abundance of Bacteroidota, Faecalibaculum, Lachnospiraceae, and Blautia may play an essential role in promoting stem-cell-mediated intestinal epithelial development and maintaining intestinal homeostasis.
    CONCLUSIONS: In summary, these findings suggest that Wuliangye baijiu can be used to regulate intestinal stem cell proliferation and differentiation in mice and to alter gut microbiota distributions, thereby promoting intestinal homeostasis. This research elucidates the mechanism by which Wuliangye baijiu promotes intestinal health. © 2024 Society of Chemical Industry.
    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